1
|
Kirk RW, Sun L, Xiao R, Clark EA, Nelson S. Multiplexed CRISPRi Reveals a Transcriptional Switch Between KLF Activators and Repressors in the Maturing Neocortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.07.636951. [PMID: 39975013 PMCID: PMC11839100 DOI: 10.1101/2025.02.07.636951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
A critical phase of mammalian brain development takes place after birth. Neurons of the mouse neocortex undergo dramatic changes in their morphology, physiology, and synaptic connections during the first postnatal month, while properties of immature neurons, such as the capacity for robust axon outgrowth, are lost. The genetic and epigenetic programs controlling prenatal development are well studied, but our understanding of the transcriptional mechanisms that regulate postnatal neuronal maturation is comparatively lacking. By integrating chromatin accessibility and gene expression data from two subtypes of neocortical pyramidal neurons in the neonatal and maturing brain, we predicted a role for the Krüppel-Like Factor (KLF) family of Transcription Factors in the developmental regulation of neonatally expressed genes. Using a multiplexed CRISPR Interference (CRISPRi) knockdown strategy, we found that a shift in expression from KLF activators (Klf6, Klf7) to repressors (Klf9, Klf13) during early postnatal development functions as a transcriptional 'switch' to first activate, then repress a set of shared targets with cytoskeletal functions including Tubb2b and Dpysl3. We demonstrate that this switch is buffered by redundancy between KLF paralogs, which our multiplexed CRISPRi strategy is equipped to overcome and study. Our results indicate that competition between activators and repressors within the KLF family regulates a conserved component of the postnatal maturation program that may underlie the loss of intrinsic axon growth in maturing neurons. This could facilitate the transition from axon growth to synaptic refinement required to stabilize mature circuits.
Collapse
Affiliation(s)
- Ryan W Kirk
- Department of Biology, Brandeis University, Waltham, MA 02453, USA
| | - Liwei Sun
- Department of Biology, Brandeis University, Waltham, MA 02453, USA
| | - Ruixuan Xiao
- Department of Biology, Brandeis University, Waltham, MA 02453, USA
| | - Erin A Clark
- Department of Biology, Brandeis University, Waltham, MA 02453, USA
| | - Sacha Nelson
- Department of Biology, Brandeis University, Waltham, MA 02453, USA
| |
Collapse
|
2
|
Siebert JR, Kennedy K, Osterhout DJ. Neurons Are Not All the Same: Diversity in Neuronal Populations and Their Intrinsic Responses to Spinal Cord Injury. ASN Neuro 2025; 17:2440299. [PMID: 39819292 DOI: 10.1080/17590914.2024.2440299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2025] Open
Abstract
Functional recovery following spinal cord injury will require the regeneration and repair of damaged neuronal pathways. It is well known that the tissue response to injury involves inflammation and the formation of a glial scar at the lesion site, which significantly impairs the capacity for neuronal regeneration and functional recovery. There are initial attempts by both supraspinal and intraspinal neurons to regenerate damaged axons, often influenced by the neighboring tissue pathology. Many experimental therapeutic strategies are targeted to further stimulate the initial axonal regrowth, with little consideration for the diversity of the affected neuronal populations. Notably, recent studies reveal that the neuronal response to injury is variable, based on multiple factors, including the location of the injury with respect to the neuronal cell bodies and the affected neuronal populations. New insights into regenerative mechanisms have shown that neurons are not homogenous but instead exhibit a wide array of diversity in their gene expression, physiology, and intrinsic responses to injury. Understanding this diverse intrinsic response is crucial, as complete functional recovery requires the successful coordinated regeneration and reorganization of various neuron pathways.
Collapse
Affiliation(s)
- Justin R Siebert
- Physician Assistant Studies Program, Department of Health Care and Administration, Slippery Rock University of Pennsylvania, Slippery Rock, PA, USA
| | - Kiersten Kennedy
- Norton College of Medicine, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Donna J Osterhout
- Department of Cell & Developmental Biology, SUNY Upstate Medical University, Syracuse, NY, USA
| |
Collapse
|
3
|
Talsma AD, Niemi JP, Zigmond RE. Neither injury induced macrophages within the nerve, nor the environment created by Wallerian degeneration is necessary for enhanced in vivo axon regeneration after peripheral nerve injury. J Neuroinflammation 2024; 21:134. [PMID: 38802868 PMCID: PMC11131297 DOI: 10.1186/s12974-024-03132-5] [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: 04/23/2024] [Accepted: 05/13/2024] [Indexed: 05/29/2024] Open
Abstract
BACKGROUND Since the 1990s, evidence has accumulated that macrophages promote peripheral nerve regeneration and are required for enhancing regeneration in the conditioning lesion (CL) response. After a sciatic nerve injury, macrophages accumulate in the injury site, the nerve distal to that site, and the axotomized dorsal root ganglia (DRGs). In the peripheral nervous system, as in other tissues, the macrophage response is derived from both resident macrophages and recruited monocyte-derived macrophages (MDMs). Unresolved questions are: at which sites do macrophages enhance nerve regeneration, and is a particular population needed. METHODS Ccr2 knock-out (KO) and Ccr2gfp/gfp knock-in/KO mice were used to prevent MDM recruitment. Using these strains in a sciatic CL paradigm, we examined the necessity of MDMs and residents for CL-enhanced regeneration in vivo and characterized injury-induced nerve inflammation. CL paradigm variants, including the addition of pharmacological macrophage depletion methods, tested the role of various macrophage populations in initiating or sustaining the CL response. In vivo regeneration, measured from bilateral proximal test lesions (TLs) after 2 d, and macrophages were quantified by immunofluorescent staining. RESULTS Peripheral CL-enhanced regeneration was equivalent between crush and transection CLs and was sustained for 28 days in both Ccr2 KO and WT mice despite MDM depletion. Similarly, the central CL response measured in dorsal roots was unchanged in Ccr2 KO mice. Macrophages at both the TL and CL, but not between them, stained for the pro-regenerative marker, arginase 1. TL macrophages were primarily CCR2-dependent MDMs and nearly absent in Ccr2 KO and Ccr2gfp/gfp KO mice. However, there were only slightly fewer Arg1+ macrophages in CCR2 null CLs than controls due to resident macrophage compensation. Zymosan injection into an intact WT sciatic nerve recruited Arg1+ macrophages but did not enhance regeneration. Finally, clodronate injection into Ccr2gfp KO CLs dramatically reduced CL macrophages. Combined with the Ccr2gfp KO background, depleting MDMs and TL macrophages, and a transection CL, physically removing the distal nerve environment, nearly all macrophages in the nerve were removed, yet CL-enhanced regeneration was not impaired. CONCLUSIONS Macrophages in the sciatic nerve are neither necessary nor sufficient to produce a CL response.
Collapse
Affiliation(s)
- Aaron D Talsma
- Department of Neurosciences, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH, 44106-4975, USA
| | - Jon P Niemi
- Department of Neurosciences, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH, 44106-4975, USA
| | - Richard E Zigmond
- Department of Neurosciences, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH, 44106-4975, USA.
| |
Collapse
|
4
|
Lisek M, Tomczak J, Boczek T, Zylinska L. Calcium-Associated Proteins in Neuroregeneration. Biomolecules 2024; 14:183. [PMID: 38397420 PMCID: PMC10887043 DOI: 10.3390/biom14020183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 01/27/2024] [Accepted: 02/01/2024] [Indexed: 02/25/2024] Open
Abstract
The dysregulation of intracellular calcium levels is a critical factor in neurodegeneration, leading to the aberrant activation of calcium-dependent processes and, ultimately, cell death. Ca2+ signals vary in magnitude, duration, and the type of neuron affected. A moderate Ca2+ concentration can initiate certain cellular repair pathways and promote neuroregeneration. While the peripheral nervous system exhibits an intrinsic regenerative capability, the central nervous system has limited self-repair potential. There is evidence that significant variations exist in evoked calcium responses and axonal regeneration among neurons, and individual differences in regenerative capacity are apparent even within the same type of neurons. Furthermore, some studies have shown that neuronal activity could serve as a potent regulator of this process. The spatio-temporal patterns of calcium dynamics are intricately controlled by a variety of proteins, including channels, ion pumps, enzymes, and various calcium-binding proteins, each of which can exert either positive or negative effects on neural repair, depending on the cellular context. In this concise review, we focus on several calcium-associated proteins such as CaM kinase II, GAP-43, oncomodulin, caldendrin, calneuron, and NCS-1 in order to elaborate on their roles in the intrinsic mechanisms governing neuronal regeneration following traumatic damage processes.
Collapse
Affiliation(s)
| | | | | | - Ludmila Zylinska
- Department of Molecular Neurochemistry, Medical University of Lodz, 92-215 Lodz, Poland; (M.L.); (J.T.); (T.B.)
| |
Collapse
|
5
|
Kheirollahi A, Sadeghi S, Orandi S, Moayedi K, Khajeh K, Khoobi M, Golestani A. Chondroitinase as a therapeutic enzyme: Prospects and challenges. Enzyme Microb Technol 2024; 172:110348. [PMID: 37898093 DOI: 10.1016/j.enzmictec.2023.110348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 09/28/2023] [Accepted: 10/19/2023] [Indexed: 10/30/2023]
Abstract
The chondroitinases (Chase) are bacterial lyases that specifically digest chondroitin sulfate and/or dermatan sulfate glycosaminoglycans via a β-elimination reaction and generate unsaturated disaccharides. In recent decades, these enzymes have attracted the attention of many researchers due to their potential applications in various aspects of medicine from the treatment of spinal cord injury to use as an analytical tool. In spite of this diverse spectrum, the application of Chase is faced with several limitations and challenges such as thermal instability and lack of a suitable delivery system. In the current review, we address potential therapeutic applications of Chase with emphasis on the challenges ahead. Then, we summarize the latest achievements to overcome the problems by considering the studies carried out in the field of enzyme engineering, drug delivery, and combination-based therapy.
Collapse
Affiliation(s)
- Asma Kheirollahi
- Department of Comparative Biosciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Solmaz Sadeghi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Shirin Orandi
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Kiana Moayedi
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Khosro Khajeh
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran 14115-154, Iran
| | - Mehdi Khoobi
- Department of Radiopharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Abolfazl Golestani
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
6
|
Benowitz LI, Xie L, Yin Y. Inflammatory Mediators of Axon Regeneration in the Central and Peripheral Nervous Systems. Int J Mol Sci 2023; 24:15359. [PMID: 37895039 PMCID: PMC10607492 DOI: 10.3390/ijms242015359] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/13/2023] [Accepted: 10/13/2023] [Indexed: 10/29/2023] Open
Abstract
Although most pathways in the mature central nervous system cannot regenerate when injured, research beginning in the late 20th century has led to discoveries that may help reverse this situation. Here, we highlight research in recent years from our laboratory identifying oncomodulin (Ocm), stromal cell-derived factor (SDF)-1, and chemokine CCL5 as growth factors expressed by cells of the innate immune system that promote axon regeneration in the injured optic nerve and elsewhere in the central and peripheral nervous systems. We also review the role of ArmC10, a newly discovered Ocm receptor, in mediating many of these effects, and the synergy between inflammation-derived growth factors and complementary strategies to promote regeneration, including deleting genes encoding cell-intrinsic suppressors of axon growth, manipulating transcription factors that suppress or promote the expression of growth-related genes, and manipulating cell-extrinsic suppressors of axon growth. In some cases, combinatorial strategies have led to unprecedented levels of nerve regeneration. The identification of some similar mechanisms in human neurons offers hope that key discoveries made in animal models may eventually lead to treatments to improve outcomes after neurological damage in patients.
Collapse
Affiliation(s)
- Larry I. Benowitz
- Department of Neurosurgery, Boston Children’s Hospital, Boston, MA 02115, USA; (L.X.); (Y.Y.)
- F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Neurosurgery, Harvard Medical School, Boston, MA 02115, USA
- Department of Ophthalmology, Harvard Medical School, Boston, MA 02115, USA
- Department of Ophthalmology, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | - Lili Xie
- Department of Neurosurgery, Boston Children’s Hospital, Boston, MA 02115, USA; (L.X.); (Y.Y.)
- Department of Neurosurgery, Harvard Medical School, Boston, MA 02115, USA
- Department of Ophthalmology, Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Yuqin Yin
- Department of Neurosurgery, Boston Children’s Hospital, Boston, MA 02115, USA; (L.X.); (Y.Y.)
- Department of Neurosurgery, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
7
|
Rocha DN, Carvalho ED, Relvas JB, Oliveira MJ, Pêgo AP. Mechanotransduction: Exploring New Therapeutic Avenues in Central Nervous System Pathology. Front Neurosci 2022; 16:861613. [PMID: 35573316 PMCID: PMC9096357 DOI: 10.3389/fnins.2022.861613] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 03/22/2022] [Indexed: 11/13/2022] Open
Abstract
Cells are continuously exposed to physical forces and the central nervous system (CNS) is no exception. Cells dynamically adapt their behavior and remodel the surrounding environment in response to forces. The importance of mechanotransduction in the CNS is illustrated by exploring its role in CNS pathology development and progression. The crosstalk between the biochemical and biophysical components of the extracellular matrix (ECM) are here described, considering the recent explosion of literature demonstrating the powerful influence of biophysical stimuli like density, rigidity and geometry of the ECM on cell behavior. This review aims at integrating mechanical properties into our understanding of the molecular basis of CNS disease. The mechanisms that mediate mechanotransduction events, like integrin, Rho/ROCK and matrix metalloproteinases signaling pathways are revised. Analysis of CNS pathologies in this context has revealed that a wide range of neurological diseases share as hallmarks alterations of the tissue mechanical properties. Therefore, it is our belief that the understanding of CNS mechanotransduction pathways may lead to the development of improved medical devices and diagnostic methods as well as new therapeutic targets and strategies for CNS repair.
Collapse
Affiliation(s)
- Daniela Nogueira Rocha
- Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
| | - Eva Daniela Carvalho
- Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- Faculdade de Engenharia (FEUP), Universidade do Porto, Porto, Portugal
| | - João Bettencourt Relvas
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal
- Departamento de Biomedicina, Faculdade de Medicina, Universidade do Porto, Porto, Portugal
| | - Maria José Oliveira
- Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Ana Paula Pêgo
- Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| |
Collapse
|
8
|
Woods I, O'Connor C, Frugoli L, Kerr S, Gutierrez Gonzalez J, Stasiewicz M, McGuire T, Cavanagh B, Hibbitts A, Dervan A, O'Brien FJ. Biomimetic Scaffolds for Spinal Cord Applications Exhibit Stiffness-Dependent Immunomodulatory and Neurotrophic Characteristics. Adv Healthc Mater 2022; 11:e2101663. [PMID: 34784649 DOI: 10.1002/adhm.202101663] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/04/2021] [Indexed: 01/14/2023]
Abstract
After spinal cord injury (SCI), tissue engineering scaffolds offer a potential bridge for regeneration across the lesion and support repair through proregenerative signaling. Ideal biomaterial scaffolds that mimic the physicochemical properties of native tissue have the potential to provide innate trophic signaling while also minimizing damaging inflammation. To address this challenge, taking cues from the spinal cord's structure, the proregenerative signaling capabilities of native cord components are compared in vitro. A synergistic mix of collagen-IV and fibronectin (Coll-IV/Fn) is found to optimally enhance axonal extension from neuronal cell lines (SHSY-5Y and NSC-34) and induce morphological features typical of quiescent astrocytes. This optimal composition is incorporated into hyaluronic acid scaffolds with aligned pore architectures but varying stiffnesses (0.8-3 kPa). Scaffolds with biomimetic mechanical properties (<1 kPa), functionalized with Coll-IV/Fn, not only modulate primary astrocyte behavior but also stimulate the production of anti-inflammatory cytokine IL-10 in a stiffness-dependent manner. Seeded SHSY-5Y neurons generate distributed neuronal networks, while softer biomimetic scaffolds promote axonal outgrowth in an ex vivo model of axonal regrowth. These results indicate that the interaction of stiffness and biomaterial composition plays an essential role in vitro in generating repair-critical cellular responses and demonstrates the potential of biomimetic scaffold design.
Collapse
Affiliation(s)
- Ian Woods
- Tissue Engineering Research Group Department of Anatomy & Regenerative Medicine Royal College of Surgeons in Ireland (RCSI) 123 St. Stephen's Green, Dublin 2, D02YN77 Ireland
| | - Cian O'Connor
- Tissue Engineering Research Group Department of Anatomy & Regenerative Medicine Royal College of Surgeons in Ireland (RCSI) 123 St. Stephen's Green, Dublin 2, D02YN77 Ireland
| | - Lisa Frugoli
- Tissue Engineering Research Group Department of Anatomy & Regenerative Medicine Royal College of Surgeons in Ireland (RCSI) 123 St. Stephen's Green, Dublin 2, D02YN77 Ireland
| | - Seán Kerr
- Tissue Engineering Research Group Department of Anatomy & Regenerative Medicine Royal College of Surgeons in Ireland (RCSI) 123 St. Stephen's Green, Dublin 2, D02YN77 Ireland
| | - Javier Gutierrez Gonzalez
- Tissue Engineering Research Group Department of Anatomy & Regenerative Medicine Royal College of Surgeons in Ireland (RCSI) 123 St. Stephen's Green, Dublin 2, D02YN77 Ireland
- Advanced Materials and Bioengineering Research (AMBER) Centre RCSI 123 St Stephen's Green, Dublin 2, D02YN77 Ireland
| | - Martyna Stasiewicz
- Tissue Engineering Research Group Department of Anatomy & Regenerative Medicine Royal College of Surgeons in Ireland (RCSI) 123 St. Stephen's Green, Dublin 2, D02YN77 Ireland
| | - Tara McGuire
- Tissue Engineering Research Group Department of Anatomy & Regenerative Medicine Royal College of Surgeons in Ireland (RCSI) 123 St. Stephen's Green, Dublin 2, D02YN77 Ireland
| | - Brenton Cavanagh
- Tissue Engineering Research Group Department of Anatomy & Regenerative Medicine Royal College of Surgeons in Ireland (RCSI) 123 St. Stephen's Green, Dublin 2, D02YN77 Ireland
- Cellular and Molecular Imaging Core Royal College of Surgeons in Ireland 123 St. Stephen's Green, Dublin 2, D02YN77 Ireland
| | - Alan Hibbitts
- Tissue Engineering Research Group Department of Anatomy & Regenerative Medicine Royal College of Surgeons in Ireland (RCSI) 123 St. Stephen's Green, Dublin 2, D02YN77 Ireland
| | - Adrian Dervan
- Tissue Engineering Research Group Department of Anatomy & Regenerative Medicine Royal College of Surgeons in Ireland (RCSI) 123 St. Stephen's Green, Dublin 2, D02YN77 Ireland
| | - Fergal J. O'Brien
- Tissue Engineering Research Group Department of Anatomy & Regenerative Medicine Royal College of Surgeons in Ireland (RCSI) 123 St. Stephen's Green, Dublin 2, D02YN77 Ireland
- Advanced Materials and Bioengineering Research (AMBER) Centre RCSI 123 St Stephen's Green, Dublin 2, D02YN77 Ireland
- Trinity Centre for Biomedical Engineering Trinity College Dublin Dublin 2, D02R590 Ireland
| |
Collapse
|
9
|
Middleton SJ, Perez-Sanchez J, Dawes JM. The structure of sensory afferent compartments in health and disease. J Anat 2021; 241:1186-1210. [PMID: 34528255 PMCID: PMC9558153 DOI: 10.1111/joa.13544] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 08/12/2021] [Accepted: 08/30/2021] [Indexed: 12/20/2022] Open
Abstract
Primary sensory neurons are a heterogeneous population of cells able to respond to both innocuous and noxious stimuli. Like most neurons they are highly compartmentalised, allowing them to detect, convey and transfer sensory information. These compartments include specialised sensory endings in the skin, the nodes of Ranvier in myelinated axons, the cell soma and their central terminals in the spinal cord. In this review, we will highlight the importance of these compartments to primary afferent function, describe how these structures are compromised following nerve damage and how this relates to neuropathic pain.
Collapse
Affiliation(s)
- Steven J Middleton
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | | | - John M Dawes
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| |
Collapse
|
10
|
Jang EH, Bae YH, Yang EM, Gim Y, Suh HJ, Kim S, Park SM, Park JB, Hur EM. Comparing axon regeneration in male and female mice after peripheral nerve injury. J Neurosci Res 2021; 99:2874-2887. [PMID: 34510521 DOI: 10.1002/jnr.24955] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 07/07/2021] [Accepted: 08/17/2021] [Indexed: 02/06/2023]
Abstract
Axons in the adult mammalian central nervous system fail to regenerate after injury. By contrast, spontaneous axon regeneration occurs in the peripheral nervous system (PNS) due to a supportive PNS environment and an increase in the intrinsic growth potential induced by injury via cooperative activation of multifaceted biological pathways. This study compared axon regeneration and injury responses in C57BL/6 male and female mice after sciatic nerve crush (SNC) injury. The extent of axon regeneration in vivo was indistinguishable in male and female mice when observed at 3 days after SNC injury, and primary dorsal root ganglion (DRG) neurons from injured, male and female mice extended axons to a similar length. Moreover, the induction of selected regeneration-associated genes (RAGs), such as Atf3, Sprr1a, Gap43, Sox11, Jun, Gadd45a, and Smad1 were comparable in male and female DRGs when assessed by quantitative real-time reverse transcription polymerase chain reaction. Furthermore, the RNA-seq analysis of male and female DRGs revealed that differentially expressed genes (DEGs) in SNC groups compared to sham-operated groups included many common genes associated with neurite outgrowth. However, we also found that a large number of genes in the DEGs were sex dependent, implicating the involvement of distinct gene regulatory network in the two sexes following peripheral nerve injury. In conclusion, we found that male and female mice mounted a comparable axon regeneration response and many RAGs were commonly induced in response to SNC. However, given that many DEGs were sex-dependently expressed, future studies are needed to investigate whether they contribute to peripheral axon regeneration, and if so, to what extent.
Collapse
Affiliation(s)
- Eun-Hae Jang
- Laboratory of Neuroscience, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Yun-Hee Bae
- Laboratory of Neuroscience, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Eun Mo Yang
- Laboratory of Neuroscience, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea.,BK21 Four Future Veterinary Medicine Leading Education & Research Center, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Yunho Gim
- Laboratory of Neuroscience, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Hyun-Jun Suh
- Laboratory of Neuroscience, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Subin Kim
- Laboratory of Neuroscience, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Seong-Min Park
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Republic of Korea
| | - Jong Bae Park
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Republic of Korea.,Rare Cancer Branch, Research Institute and Hospital, National Cancer Center, Goyang, Republic of Korea
| | - Eun-Mi Hur
- Laboratory of Neuroscience, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea.,BK21 Four Future Veterinary Medicine Leading Education & Research Center, College of Veterinary Medicine, Seoul National University, Seoul, South Korea.,Interdisciplinary Program in Neuroscience, College of Natural Sciences, Seoul National University, Seoul, South Korea
| |
Collapse
|
11
|
Deerhake ME, Shinohara ML. Emerging roles of Dectin-1 in noninfectious settings and in the CNS. Trends Immunol 2021; 42:891-903. [PMID: 34489167 DOI: 10.1016/j.it.2021.08.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/13/2021] [Accepted: 08/14/2021] [Indexed: 12/15/2022]
Abstract
Dectin-1 is a C-type lectin receptor (CLR) expressed on the surface of various mammalian myeloid cells. Dectin-1 recognizes β-glucans and elicits antifungal proinflammatory immune responses. Recent studies have begun to examine the biology of Dectin-1 in previously less explored settings, such as homeostasis, sterile inflammation, and in the central nervous system. Indeed, in certain contexts, Dectin-1 is now known to promote tolerance, and anti-inflammatory and neuroprotective responses. In this review, we provide an overview of the current understanding of the roles of Dectin-1 in immunology beyond the context of fungal infections, mainly focusing on in vivo neuroimmunology studies, which could reveal new therapeutic approaches to modify innate immune responses in neurologic disorders.
Collapse
Affiliation(s)
- M Elizabeth Deerhake
- Department of Immunology, Duke University School of Medicine, Durham, NC 27705, USA
| | - Mari L Shinohara
- Department of Immunology, Duke University School of Medicine, Durham, NC 27705, USA; Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27705, USA.
| |
Collapse
|
12
|
Aldskogius H, Kozlova EN. Dorsal Root Injury-A Model for Exploring Pathophysiology and Therapeutic Strategies in Spinal Cord Injury. Cells 2021; 10:2185. [PMID: 34571835 PMCID: PMC8470715 DOI: 10.3390/cells10092185] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 08/19/2021] [Accepted: 08/20/2021] [Indexed: 12/12/2022] Open
Abstract
Unraveling the cellular and molecular mechanisms of spinal cord injury is fundamental for our possibility to develop successful therapeutic approaches. These approaches need to address the issues of the emergence of a non-permissive environment for axonal growth in the spinal cord, in combination with a failure of injured neurons to mount an effective regeneration program. Experimental in vivo models are of critical importance for exploring the potential clinical relevance of mechanistic findings and therapeutic innovations. However, the highly complex organization of the spinal cord, comprising multiple types of neurons, which form local neural networks, as well as short and long-ranging ascending or descending pathways, complicates detailed dissection of mechanistic processes, as well as identification/verification of therapeutic targets. Inducing different types of dorsal root injury at specific proximo-distal locations provide opportunities to distinguish key components underlying spinal cord regeneration failure. Crushing or cutting the dorsal root allows detailed analysis of the regeneration program of the sensory neurons, as well as of the glial response at the dorsal root-spinal cord interface without direct trauma to the spinal cord. At the same time, a lesion at this interface creates a localized injury of the spinal cord itself, but with an initial neuronal injury affecting only the axons of dorsal root ganglion neurons, and still a glial cell response closely resembling the one seen after direct spinal cord injury. In this review, we provide examples of previous research on dorsal root injury models and how these models can help future exploration of mechanisms and potential therapies for spinal cord injury repair.
Collapse
Affiliation(s)
- Håkan Aldskogius
- Laboratory of Regenertive Neurobiology, Biomedical Center, Department of Neuroscience, Uppsala University, 75124 Uppsala, Sweden;
| | | |
Collapse
|
13
|
EFA6 in Axon Regeneration, as a Microtubule Regulator and as a Guanine Nucleotide Exchange Factor. Cells 2021; 10:cells10061325. [PMID: 34073530 PMCID: PMC8226579 DOI: 10.3390/cells10061325] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/23/2021] [Accepted: 05/24/2021] [Indexed: 11/17/2022] Open
Abstract
Axon regeneration after injury is a conserved biological process that involves a large number of molecular pathways, including rapid calcium influx at injury sites, retrograde injury signaling, epigenetic transition, transcriptional reprogramming, polarized transport, and cytoskeleton reorganization. Despite the numerous efforts devoted to understanding the underlying cellular and molecular mechanisms of axon regeneration, the search continues for effective target molecules for improving axon regeneration. Although there have been significant historical efforts towards characterizing pro-regenerative factors involved in axon regeneration, the pursuit of intrinsic inhibitors is relatively recent. EFA6 (exchange factor for ARF6) has been demonstrated to inhibit axon regeneration in different organisms. EFA6 inhibition could be a promising therapeutic strategy to promote axon regeneration and functional recovery after axon injury. This review summarizes the inhibitory role on axon regeneration through regulating microtubule dynamics and through affecting ARF6 (ADP-ribosylation factor 6) GTPase-mediated integrin transport.
Collapse
|
14
|
Zhai J, Kim H, Han SB, Manire M, Yoo R, Pang S, Smith GM, Son YJ. Co-targeting myelin inhibitors and CSPGs markedly enhances regeneration of GDNF-stimulated, but not conditioning-lesioned, sensory axons into the spinal cord. eLife 2021; 10:63050. [PMID: 33942723 PMCID: PMC8139830 DOI: 10.7554/elife.63050] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 05/03/2021] [Indexed: 12/20/2022] Open
Abstract
A major barrier to intraspinal regeneration after dorsal root (DR) injury is the DR entry zone (DREZ), the CNS/PNS interface. DR axons stop regenerating at the DREZ, even if regenerative capacity is increased by a nerve conditioning lesion. This potent blockade has long been attributed to myelin-associated inhibitors and (CSPGs), but incomplete lesions and conflicting reports have prevented conclusive agreement. Here, we evaluated DR regeneration in mice using novel strategies to facilitate complete lesions and analyses, selective tracing of proprioceptive and mechanoreceptive axons, and the first simultaneous targeting of Nogo/Reticulon-4, MAG, OMgp, CSPGs, and GDNF. Co-eliminating myelin inhibitors and CSPGs elicited regeneration of only a few conditioning-lesioned DR axons across the DREZ. Their absence, however, markedly and synergistically enhanced regeneration of GDNF-stimulated axons, highlighting the importance of sufficiently elevating intrinsic growth capacity. We also conclude that myelin inhibitors and CSPGs are not the primary mechanism stopping axons at the DREZ.
Collapse
Affiliation(s)
- Jinbin Zhai
- Shriners Hospitals Pediatric Research Center and Center for Neural Repair and Rehabilitation, Lewis Katz School of Medicine, Temple University, Philadelphia, United States.,Center for Neural Repair and Rehabilitation, Lewis Katz School of Medicine, Temple University, Philadelphia, United States
| | - Hyukmin Kim
- Shriners Hospitals Pediatric Research Center and Center for Neural Repair and Rehabilitation, Lewis Katz School of Medicine, Temple University, Philadelphia, United States.,Center for Neural Repair and Rehabilitation, Lewis Katz School of Medicine, Temple University, Philadelphia, United States
| | - Seung Baek Han
- Shriners Hospitals Pediatric Research Center and Center for Neural Repair and Rehabilitation, Lewis Katz School of Medicine, Temple University, Philadelphia, United States.,Center for Neural Repair and Rehabilitation, Lewis Katz School of Medicine, Temple University, Philadelphia, United States
| | - Meredith Manire
- Shriners Hospitals Pediatric Research Center and Center for Neural Repair and Rehabilitation, Lewis Katz School of Medicine, Temple University, Philadelphia, United States.,Center for Neural Repair and Rehabilitation, Lewis Katz School of Medicine, Temple University, Philadelphia, United States
| | - Rachel Yoo
- Shriners Hospitals Pediatric Research Center and Center for Neural Repair and Rehabilitation, Lewis Katz School of Medicine, Temple University, Philadelphia, United States.,Center for Neural Repair and Rehabilitation, Lewis Katz School of Medicine, Temple University, Philadelphia, United States
| | - Shuhuan Pang
- Shriners Hospitals Pediatric Research Center and Center for Neural Repair and Rehabilitation, Lewis Katz School of Medicine, Temple University, Philadelphia, United States.,Center for Neural Repair and Rehabilitation, Lewis Katz School of Medicine, Temple University, Philadelphia, United States
| | - George M Smith
- Shriners Hospitals Pediatric Research Center and Center for Neural Repair and Rehabilitation, Lewis Katz School of Medicine, Temple University, Philadelphia, United States.,Center for Neural Repair and Rehabilitation, Lewis Katz School of Medicine, Temple University, Philadelphia, United States
| | - Young-Jin Son
- Shriners Hospitals Pediatric Research Center and Center for Neural Repair and Rehabilitation, Lewis Katz School of Medicine, Temple University, Philadelphia, United States.,Center for Neural Repair and Rehabilitation, Lewis Katz School of Medicine, Temple University, Philadelphia, United States
| |
Collapse
|
15
|
Prager J, Ito D, Carwardine DR, Jiju P, Chari DM, Granger N, Wong LF. Delivery of chondroitinase by canine mucosal olfactory ensheathing cells alongside rehabilitation enhances recovery after spinal cord injury. Exp Neurol 2021; 340:113660. [PMID: 33647272 DOI: 10.1016/j.expneurol.2021.113660] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 02/21/2021] [Accepted: 02/24/2021] [Indexed: 01/11/2023]
Abstract
Spinal cord injury (SCI) can cause chronic paralysis and incontinence and remains a major worldwide healthcare burden, with no regenerative treatment clinically available. Intraspinal transplantation of olfactory ensheathing cells (OECs) and injection of chondroitinase ABC (chABC) are both promising therapies but limited and unpredictable responses are seen, particularly in canine clinical trials. Sustained delivery of chABC presents a challenge due to its thermal instability; we hypothesised that transplantation of canine olfactory mucosal OECs genetically modified ex vivo by lentiviral transduction to express chABC (cOEC-chABC) would provide novel delivery of chABC and synergistic therapy. Rats were randomly divided into cOEC-chABC, cOEC, or vehicle transplanted groups and received transplant immediately after dorsal column crush corticospinal tract (CST) injury. Rehabilitation for forepaw reaching and blinded behavioural testing was conducted for 8 weeks. We show that cOEC-chABC transplanted animals recover greater forepaw reaching accuracy on Whishaw testing and more normal gait than cOEC transplanted or vehicle control rats. Increased CST axon sprouting cranial to the injury and serotonergic fibres caudal to the injury suggest a mechanism for recovery. We therefore demonstrate that cOECs can deliver sufficient chABC to drive modest functional improvement, and that this genetically engineered cellular and molecular approach is a feasible combination therapy for SCI.
Collapse
Affiliation(s)
- Jon Prager
- Bristol Veterinary School, University of Bristol, Bristol, UK; The Royal Veterinary College, University of London, Hatfield, UK
| | - Daisuke Ito
- Bristol Medical School, University of Bristol, Bristol, UK; School of Veterinary Medicine, Nihon University, Japan
| | | | - Prince Jiju
- Bristol Medical School, University of Bristol, Bristol, UK
| | - Divya M Chari
- Neural Tissue Engineering, Keele School of Medicine, Keele University, Keele, UK
| | - Nicolas Granger
- The Royal Veterinary College, University of London, Hatfield, UK
| | | |
Collapse
|
16
|
Wu D, Jin Y, Shapiro TM, Hinduja A, Baas PW, Tom VJ. Chronic neuronal activation increases dynamic microtubules to enhance functional axon regeneration after dorsal root crush injury. Nat Commun 2020; 11:6131. [PMID: 33257677 PMCID: PMC7705672 DOI: 10.1038/s41467-020-19914-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 11/05/2020] [Indexed: 12/26/2022] Open
Abstract
After a dorsal root crush injury, centrally-projecting sensory axons fail to regenerate across the dorsal root entry zone (DREZ) to extend into the spinal cord. We find that chemogenetic activation of adult dorsal root ganglion (DRG) neurons improves axon growth on an in vitro model of the inhibitory environment after injury. Moreover, repeated bouts of daily chemogenetic activation of adult DRG neurons for 12 weeks post-crush in vivo enhances axon regeneration across a chondroitinase-digested DREZ into spinal gray matter, where the regenerating axons form functional synapses and mediate behavioral recovery in a sensorimotor task. Neuronal activation-mediated axon extension is dependent upon changes in the status of tubulin post-translational modifications indicative of highly dynamic microtubules (as opposed to stable microtubules) within the distal axon, illuminating a novel mechanism underlying stimulation-mediated axon growth. We have identified an effective combinatory strategy to promote functionally-relevant axon regeneration of adult neurons into the CNS after injury.
Collapse
Affiliation(s)
- Di Wu
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Ying Jin
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Tatiana M Shapiro
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Abhishek Hinduja
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Peter W Baas
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Veronica J Tom
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
17
|
Stoyanov S, Sun W, Düsedau HP, Cangalaya C, Choi I, Mirzapourdelavar H, Baidoe-Ansah D, Kaushik R, Neumann J, Dunay IR, Dityatev A. Attenuation of the extracellular matrix restores microglial activity during the early stage of amyloidosis. Glia 2020; 69:182-200. [PMID: 32865286 DOI: 10.1002/glia.23894] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 07/10/2020] [Accepted: 07/15/2020] [Indexed: 12/16/2022]
Abstract
In the advanced stages of Alzheimer's disease (AD), microglia are transformed to an activated phenotype with thickened and retracted processes, migrate to the site of amyloid-beta (Aβ) plaques, and proliferate. In the early stages of AD, it is still poorly understood whether the microglial function is altered and which factors may regulate these changes. Here, we focused on studying microglia in the retrosplenial cortex (RSC) in 3- to 4-month-old 5xFAD mice as a transgenic mouse model of AD. At this age, there are neither Aβ plaques, nor activation of microglia, nor dysregulation in the expression of genes encoding major extracellular matrix (ECM) molecules or extracellular proteases in the RSC. Still, histochemical evaluation of the fine structure of neural ECM revealed increased levels of Wisteria floribunda agglutinin labeling in holes of perineuronal nets and changes in the perimeter of ECM barriers around the holes in 5xFAD mice. Two-photon vital microscopy demonstrated normal morphology and resting motility of microglia but strongly diminished number of microglial cells that migrated to the photolesion site in 5xFAD mice. Enzymatic digestion of ECM by chondroitinase ABC (ChABC) ameliorated this defect. Accordingly, the characterization of cell surface markers by flow cytometry demonstrated altered expression of microglial CD45. Moreover, ChABC treatment reduced the invasion of myeloid-derived mononuclear cells into the RSC of 5xFAD mice. Hence, the migration of both microglia and myeloid cells is altered during the early stages of amyloidosis and can be restored at least partially by the attenuation of the ECM.
Collapse
Affiliation(s)
- Stoyan Stoyanov
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Weilun Sun
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Henning Peter Düsedau
- Institute of Inflammation and Neurodegeneration, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Carla Cangalaya
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Institute of Biochemistry, Otto-von-Guericke University, Magdeburg, Germany
| | - Ilseob Choi
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Hadi Mirzapourdelavar
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - David Baidoe-Ansah
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Rahul Kaushik
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Jens Neumann
- Department of Neurology, Otto-von-Guericke University, Magdeburg, Germany
| | - Ildiko Rita Dunay
- Institute of Inflammation and Neurodegeneration, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
| | - Alexander Dityatev
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
- Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| |
Collapse
|
18
|
Jara JS, Agger S, Hollis ER. Functional Electrical Stimulation and the Modulation of the Axon Regeneration Program. Front Cell Dev Biol 2020; 8:736. [PMID: 33015031 PMCID: PMC7462022 DOI: 10.3389/fcell.2020.00736] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 07/15/2020] [Indexed: 01/07/2023] Open
Abstract
Neural injury in mammals often leads to persistent functional deficits as spontaneous repair in the peripheral nervous system (PNS) is often incomplete, while endogenous repair mechanisms in the central nervous system (CNS) are negligible. Peripheral axotomy elicits growth-associated gene programs in sensory and motor neurons that can support reinnervation of peripheral targets given sufficient levels of debris clearance and proximity to nerve targets. In contrast, while damaged CNS circuitry can undergo a limited amount of sprouting and reorganization, this innate plasticity does not re-establish the original connectivity. The utility of novel CNS circuitry will depend on effective connectivity and appropriate training to strengthen these circuits. One method of enhancing novel circuit connectivity is through the use of electrical stimulation, which supports axon growth in both central and peripheral neurons. This review will focus on the effects of CNS and PNS electrical stimulation in activating axon growth-associated gene programs and supporting the recovery of motor and sensory circuits. Electrical stimulation-mediated neuroplasticity represents a therapeutically viable approach to support neural repair and recovery. Development of appropriate clinical strategies employing electrical stimulation will depend upon determining the underlying mechanisms of activity-dependent axon regeneration and the heterogeneity of neuronal subtype responses to stimulation.
Collapse
Affiliation(s)
| | - Sydney Agger
- Burke Neurological Institute, White Plains, NY, United States
| | - Edmund R Hollis
- Burke Neurological Institute, White Plains, NY, United States.,Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
19
|
Yin Y, De Lima S, Gilbert HY, Hanovice NJ, Peterson SL, Sand RM, Sergeeva EG, Wong KA, Xie L, Benowitz LI. Optic nerve regeneration: A long view. Restor Neurol Neurosci 2020; 37:525-544. [PMID: 31609715 DOI: 10.3233/rnn-190960] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The optic nerve conveys information about the outside world from the retina to multiple subcortical relay centers. Until recently, the optic nerve was widely believed to be incapable of re-growing if injured, with dire consequences for victims of traumatic, ischemic, or neurodegenerative diseases of this pathway. Over the past 10-20 years, research from our lab and others has made considerable progress in defining factors that normally suppress axon regeneration and the ability of retinal ganglion cells, the projection neurons of the retina, to survive after nerve injury. Here we describe research from our lab on the role of inflammation-derived growth factors, suppression of inter-cellular signals among diverse retinal cell types, and combinatorial therapies, along with related studies from other labs, that enable animals with optic nerve injury to regenerate damaged retinal axons back to the brain. These studies raise the possibility that vision might one day be restored to people with optic nerve damage.
Collapse
Affiliation(s)
- Yuqin Yin
- Laboratories for Neuroscience Research in Neurosurgery, Boston Children's Hospital, Boston, MA, USA.,Department of Neurosurgery, Boston Children's Hospital, Boston, MA, USA.,F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA.,Department of Neurosurgery, Harvard Medical School, Boston, MA, USA
| | - Silmara De Lima
- Laboratories for Neuroscience Research in Neurosurgery, Boston Children's Hospital, Boston, MA, USA.,Department of Neurosurgery, Boston Children's Hospital, Boston, MA, USA.,F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA.,Department of Neurosurgery, Harvard Medical School, Boston, MA, USA
| | - Hui-Ya Gilbert
- Laboratories for Neuroscience Research in Neurosurgery, Boston Children's Hospital, Boston, MA, USA.,Department of Neurosurgery, Boston Children's Hospital, Boston, MA, USA
| | - Nicholas J Hanovice
- Laboratories for Neuroscience Research in Neurosurgery, Boston Children's Hospital, Boston, MA, USA.,Department of Neurosurgery, Boston Children's Hospital, Boston, MA, USA.,F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA.,Department of Neurosurgery, Harvard Medical School, Boston, MA, USA
| | - Sheri L Peterson
- Laboratories for Neuroscience Research in Neurosurgery, Boston Children's Hospital, Boston, MA, USA.,Department of Neurosurgery, Boston Children's Hospital, Boston, MA, USA.,F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA.,Department of Neurosurgery, Harvard Medical School, Boston, MA, USA
| | - Rheanna M Sand
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA.,Department of Neurology, Boston Children's Hospital, Boston, MA, USA.,Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Elena G Sergeeva
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA.,Department of Neurology, Boston Children's Hospital, Boston, MA, USA.,Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Kimberly A Wong
- Laboratories for Neuroscience Research in Neurosurgery, Boston Children's Hospital, Boston, MA, USA.,Department of Neurosurgery, Boston Children's Hospital, Boston, MA, USA.,F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA.,Department of Neurosurgery, Harvard Medical School, Boston, MA, USA
| | - Lili Xie
- Laboratories for Neuroscience Research in Neurosurgery, Boston Children's Hospital, Boston, MA, USA.,Department of Neurosurgery, Boston Children's Hospital, Boston, MA, USA.,F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA.,Department of Neurosurgery, Harvard Medical School, Boston, MA, USA
| | - Larry I Benowitz
- Laboratories for Neuroscience Research in Neurosurgery, Boston Children's Hospital, Boston, MA, USA.,Department of Neurosurgery, Boston Children's Hospital, Boston, MA, USA.,F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA.,Department of Neurosurgery, Harvard Medical School, Boston, MA, USA.,Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
20
|
Recent advances in the therapeutic uses of chondroitinase ABC. Exp Neurol 2019; 321:113032. [PMID: 31398353 DOI: 10.1016/j.expneurol.2019.113032] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 07/19/2019] [Accepted: 08/03/2019] [Indexed: 12/18/2022]
Abstract
Many studies, using pre-clinical models of SCI, have demonstrated the efficacy of chondroitinase ABC as a treatment for spinal cord injury and this has been confirmed in laboratories worldwide and in several animal models. The aim of this review is report the current state of research in the field and to compare the relative efficacies of these new interventions to improve outcomes in both acute and chronic models of SCI. We also report new methods of chondroitinase delivery and the outcomes of two clinical trials using the enzyme to treat spinal cord injury in dogs and disc herniation in human patients. Recent studies have assessed the outcomes of combining chondroitinase with other strategies known to promote recovery following spinal cord injury and new approaches. Evidence is emerging that one of the most powerful combinations is that of chondroitinase with cell transplants. The particular benefits of each of the different cell types used for these transplant experiments are discussed. Combining chondroitinase with rehabilitation also improves outcomes. Gene therapy is an efficient method of enzyme delivery to the injured spinal cord and circumvents the issue of the enzyme's thermo-instability. Other methods of delivery, such as via nanoparticles or synthetic scaffolds, have shown promise; however, the outcomes from these experiments suggest that these methods of delivery require further optimization to achieve similar levels of efficacy to that obtained by a gene therapy approach. Pre-clinical models have also shown chondroitinase is efficacious in the treatment of other conditions, such as peripheral nerve injury, stroke, coronary reperfusion, Parkinson's disease and certain types of cancer. The wide range of conditions where the benefits of chondroitinase treatment have been demonstrated reflects the complex roles that chondroitin sulphate proteoglycans (its substrate) play in health and disease and warrants the enzyme's further development as a therapy.
Collapse
|
21
|
Long-Distance Axon Regeneration Promotes Recovery of Diaphragmatic Respiratory Function after Spinal Cord Injury. eNeuro 2019; 6:ENEURO.0096-19.2019. [PMID: 31427403 PMCID: PMC6794082 DOI: 10.1523/eneuro.0096-19.2019] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 06/11/2019] [Accepted: 06/14/2019] [Indexed: 12/14/2022] Open
Abstract
Compromise in inspiratory breathing following cervical spinal cord injury (SCI) is caused by damage to descending bulbospinal axons originating in the rostral ventral respiratory group (rVRG) and consequent denervation and silencing of phrenic motor neurons (PhMNs) that directly control diaphragm activation. In a rat model of high-cervical hemisection SCI, we performed systemic administration of an antagonist peptide directed against phosphatase and tensin homolog (PTEN), a central inhibitor of neuron-intrinsic axon growth potential. PTEN antagonist peptide (PAP4) robustly restored diaphragm function, as determined with electromyography (EMG) recordings in living SCI animals. PAP4 promoted substantial, long-distance regeneration of injured rVRG axons through the lesion and back toward PhMNs located throughout the C3–C5 spinal cord. These regrowing rVRG axons also formed putative excitatory synaptic connections with PhMNs, demonstrating reconnection of rVRG-PhMN-diaphragm circuitry. Lastly, re-lesion through the hemisection site completely ablated functional recovery induced by PAP4. Collectively, our findings demonstrate that axon regeneration in response to systemic PAP4 administration promoted recovery of diaphragmatic respiratory function after cervical SCI.
Collapse
|
22
|
Ong W, Pinese C, Chew SY. Scaffold-mediated sequential drug/gene delivery to promote nerve regeneration and remyelination following traumatic nerve injuries. Adv Drug Deliv Rev 2019; 149-150:19-48. [PMID: 30910595 DOI: 10.1016/j.addr.2019.03.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 02/27/2019] [Accepted: 03/19/2019] [Indexed: 02/06/2023]
Abstract
Neural tissue regeneration following traumatic injuries is often subpar. As a result, the field of neural tissue engineering has evolved to find therapeutic interventions and has seen promising outcomes. However, robust nerve and myelin regeneration remain elusive. One possible reason may be the fact that tissue regeneration often follows a complex sequence of events in a temporally-controlled manner. Although several other fields of tissue engineering have begun to recognise the importance of delivering two or more biomolecules sequentially for more complete tissue regeneration, such serial delivery of biomolecules in neural tissue engineering remains limited. This review aims to highlight the need for sequential delivery to enhance nerve regeneration and remyelination after traumatic injuries in the central nervous system, using spinal cord injuries as an example. In addition, possible methods to attain temporally-controlled drug/gene delivery are also discussed for effective neural tissue regeneration.
Collapse
|
23
|
Rosenzweig ES, Salegio EA, Liang JJ, Weber JL, Weinholtz CA, Brock JH, Moseanko R, Hawbecker S, Pender R, Cruzen CL, Iaci JF, Caggiano AO, Blight AR, Haenzi B, Huie JR, Havton LA, Nout-Lomas YS, Fawcett JW, Ferguson AR, Beattie MS, Bresnahan JC, Tuszynski MH. Chondroitinase improves anatomical and functional outcomes after primate spinal cord injury. Nat Neurosci 2019; 22:1269-1275. [PMID: 31235933 PMCID: PMC6693679 DOI: 10.1038/s41593-019-0424-1] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 05/10/2019] [Indexed: 01/07/2023]
Abstract
Inhibitory extracellular matrices form around mature neurons as perineuronal nets containing chondroitin sulfate proteoglycans that limit axonal sprouting after CNS injury. The enzyme chondroitinase (Chase) degrades inhibitory chondroitin sulfate proteoglycans and improves axonal sprouting and functional recovery after spinal cord injury in rodents. We evaluated the effects of Chase in rhesus monkeys that had undergone C7 spinal cord hemisection. Four weeks after hemisection, we administered multiple intraparenchymal Chase injections below the lesion, targeting spinal cord circuits that control hand function. Hand function improved significantly in Chase-treated monkeys relative to vehicle-injected controls. Moreover, Chase significantly increased corticospinal axon growth and the number of synapses formed by corticospinal terminals in gray matter caudal to the lesion. No detrimental effects were detected. This approach appears to merit clinical translation in spinal cord injury.
Collapse
Affiliation(s)
- Ephron S Rosenzweig
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Ernesto A Salegio
- California National Primate Research Center, University of California, Davis, Davis, CA, USA
| | - Justine J Liang
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Janet L Weber
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Chase A Weinholtz
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - John H Brock
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
- Veterans Administration Medical Center, La Jolla, CA, USA
| | - Rod Moseanko
- California National Primate Research Center, University of California, Davis, Davis, CA, USA
| | - Stephanie Hawbecker
- California National Primate Research Center, University of California, Davis, Davis, CA, USA
| | - Roger Pender
- California National Primate Research Center, University of California, Davis, Davis, CA, USA
| | - Christina L Cruzen
- California National Primate Research Center, University of California, Davis, Davis, CA, USA
| | | | | | | | | | - J Russell Huie
- Department of Neurosurgery, University of California, San Francisco, San Francisco, CA, USA
| | - Leif A Havton
- Department of Neurology, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Neurobiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yvette S Nout-Lomas
- College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | | | - Adam R Ferguson
- Department of Neurosurgery, University of California, San Francisco, San Francisco, CA, USA
| | - Michael S Beattie
- Department of Neurosurgery, University of California, San Francisco, San Francisco, CA, USA
| | - Jacqueline C Bresnahan
- Department of Neurosurgery, University of California, San Francisco, San Francisco, CA, USA
| | - Mark H Tuszynski
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA.
- Veterans Administration Medical Center, La Jolla, CA, USA.
| |
Collapse
|
24
|
Swieck K, Conta-Steencken A, Middleton FA, Siebert JR, Osterhout DJ, Stelzner DJ. Effect of lesion proximity on the regenerative response of long descending propriospinal neurons after spinal transection injury. BMC Neurosci 2019; 20:10. [PMID: 30885135 PMCID: PMC6421714 DOI: 10.1186/s12868-019-0491-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 03/05/2019] [Indexed: 02/07/2023] Open
Abstract
Background The spinal cord is limited in its capacity to repair after damage caused by injury or disease. However, propriospinal (PS) neurons in the spinal cord have demonstrated a propensity for axonal regeneration after spinal cord injury. They can regrow and extend axonal projections to re-establish connections across a spinal lesion. We have previously reported differential reactions of two distinct PS neuronal populations—short thoracic propriospinal (TPS) and long descending propriospinal tract (LDPT) neurons—following a low thoracic (T10) spinal cord injury in a rat model. Immediately after injury, TPS neurons undergo a strong initial regenerative response, defined by the upregulation of transcripts to several growth factor receptors, and growth associated proteins. Many also initiate a strong apoptotic response, leading to cell death. LDPT neurons, on the other hand, show neither a regenerative nor an apoptotic response. They show either a lowered expression or no change in genes for a variety of growth associated proteins, and these neurons survive for at least 2 months post-axotomy. There are several potential explanations for this lack of cellular response for LDPT neurons, one of which is the distance of the LDPT cell body from the T10 lesion. In this study, we examined the molecular response of LDPT neurons to axotomy caused by a proximal spinal cord lesion. Results Utilizing laser capture microdissection and RNA quantification with branched DNA technology, we analyzed the change in gene expression in LDPT neurons following axotomy near their cell body. Expression patterns of 34 genes selected for their robust responses in TPS neurons were analyzed 3 days following a T2 spinal lesion. Our results show that after axonal injury nearer their cell bodies, there was a differential response of the same set of genes evaluated previously in TPS neurons after proximal axotomy, and LDPT neurons after distal axotomy (T10 spinal transection). The genetic response was much less robust than for TPS neurons after proximal axotomy, included both increased and decreased expression of certain genes, and did not suggest either a major regenerative or apoptotic response within the population of genes examined. Conclusions The data collectively demonstrate that the location of axotomy in relation to the soma of a neuron has a major effect on its ability to mount a regenerative response. However, the data also suggest that there are endogenous differences in the LDPT and TPS neuronal populations that affect their response to axotomy. These phenotypic differences may indicate that different or multiple therapies may be needed following spinal cord injury to stimulate maximal regeneration of all PS axons.
Collapse
Affiliation(s)
- Kristen Swieck
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, NY, 13210, USA
| | - Amanda Conta-Steencken
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, NY, 13210, USA
| | - Frank A Middleton
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, NY, 13210, USA
| | - Justin R Siebert
- Department of Biology, Slippery Rock University, 1 Morrow Way, Slippery Rock, PA, 16057, USA
| | - Donna J Osterhout
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, NY, 13210, USA.
| | - Dennis J Stelzner
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, NY, 13210, USA
| |
Collapse
|
25
|
Anti-Chondroitin Sulfate Proteoglycan Strategies in Spinal Cord Injury: Temporal and Spatial Considerations Explain the Balance between Neuroplasticity and Neuroprotection. J Neurotrauma 2018. [DOI: 10.1089/neu.2018.5928] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
|
26
|
Kigerl KA, Lai W, Wallace LM, Yang H, Popovich PG. High mobility group box-1 (HMGB1) is increased in injured mouse spinal cord and can elicit neurotoxic inflammation. Brain Behav Immun 2018; 72:22-33. [PMID: 29175543 PMCID: PMC6681463 DOI: 10.1016/j.bbi.2017.11.018] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 11/09/2017] [Accepted: 11/22/2017] [Indexed: 01/26/2023] Open
Abstract
Inflammation is a ubiquitous but poorly understood consequence of spinal cord injury (SCI). The mechanisms controlling this response are unclear but culminate in the sequential activation of resident and recruited immune cells. Collectively, these cells can exert divergent effects on cell survival and tissue repair. HMGB1 is a ubiquitously expressed DNA binding protein and also a potent inflammatory stimulus. Necrotic cells release HGMB1, but HMGB1 also is actively secreted by inflammatory macrophages. A goal of this study was to quantify spatio-temporal patterns of cellular HMGB1 expression in a controlled mouse model of experimental SCI then determine the effects of HMGB1 on post-SCI neuroinflammation and recovery of function. We documented SCI-induced changes in nuclear and cytoplasmic distribution of HMGB1 in various cell types after SCI. The data reveal a time-dependent increase in HMGB1 mRNA and protein with protein reaching maximal levels 24-72 h post-injury then declining toward baseline 14-28 days post-SCI. Although most cells expressed nuclear HMGB1, reduced nuclear labeling with increased cytoplasmic expression was found in a subset of CNS macrophages suggesting that those cells begin to secrete HMGB1 at the injury site. In vitro data indicate that extracelluar HMGB1 helps promote the development of macrophages with a neurotoxic phenotype. The ability of HMGB1 to elicit neurotoxic macrophage functions was confirmed in vivo; 72 h after injecting 500 ng of recombinant HMGB1 into intact spinal cord ventral horn, inflammatory CNS macrophages co-localized with focal areas of neuronal killing. However, attempts to confer neuroprotection after SCI by blocking HMGB1 with a neutralizing antibody were unsuccessful. Collectively, these data implicate HMGB1 as a novel regulator of post-SCI inflammation and suggest that inhibition of HMGB1 could be a novel therapeutic target after SCI. Future studies will need to identify better methods to deliver optimal concentrations of HMGB1 antagonists to the injured spinal cord.
Collapse
Affiliation(s)
- Kristina A. Kigerl
- Center for Brain and Spinal Cord Repair, Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Wenmin Lai
- Center for Brain and Spinal Cord Repair, Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Lindsay M. Wallace
- Center for Gene Therapy, The Research Institute at Nationwide Children’s Hospital, Columbus, OH, USA
| | - Huan Yang
- Feinstein Institute for Medical Research, Manhasset, NY, USA
| | - Phillip G. Popovich
- Center for Brain and Spinal Cord Repair, Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, USA,corresponding author: Phillip G. Popovich, Center for Brain and Spinal Cord Repair, 786 Biomedical Research Tower, 460 W. 12th Ave, Columbus, OH 43210, (614) 688-8576,
| |
Collapse
|
27
|
Patil N, Truong V, Holmberg MH, Lavoie NS, McCoy MR, Dutton JR, Holmberg EG, Parr AM. Safety and Efficacy of Rose Bengal Derivatives for Glial Scar Ablation in Chronic Spinal Cord Injury. J Neurotrauma 2018; 35:1745-1754. [PMID: 29373946 PMCID: PMC6033306 DOI: 10.1089/neu.2017.5398] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
There are no effective therapies available currently to ameliorate loss of function for patients with spinal cord injuries (SCIs). In addition, proposed treatments that demonstrated functional recovery in animal models of acute SCI have failed almost invariably when applied to chronic injury models. Glial scar formation in chronic injury is a likely contributor to limitation on regeneration. We have removed existing scar tissue in chronically contused rat spinal cord using a rose Bengal-based photo ablation approach. In this study, we compared two chemically modified rose bengal derivatives to unmodified rose bengal, both confirming and expanding on our previously published report. Rats were treated with unmodified rose bengal (RB1) or rose bengal modified with hydrocarbon (RB2) or polyethylene glycol (RB3), to determine the effects on scar components and spared tissue post-treatment. Our results showed that RB1 was more efficacious than RB2, while still maintaining minimal collateral effects on spared tissue. RB3 was not taken up by the cells, likely because of its size, and therefore had no effect. Treatment with RB1 also resulted in an increase in serotonin eight days post-treatment in chronically injured spinal cords. Thus, we suggest that unmodified rose Bengal is a potent candidate agent for the development of a therapeutic strategy for scar ablation in chronic SCI.
Collapse
Affiliation(s)
- Nandadevi Patil
- Department of Neurosurgery, Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota
| | - Vincent Truong
- Department of Neurosurgery, Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota
| | - Mackenzie H. Holmberg
- Department of Chemistry, University of Alaska Anchorage, Anchorage, Alaska
- University of Washington School of Medicine, Seattle, Washington
| | - Nicolas S. Lavoie
- Department of Neurosurgery, Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota
| | - Mark R. McCoy
- Department of Chemistry, University of Alaska Anchorage, Anchorage, Alaska
| | - James R. Dutton
- Department of Genetics, Cell Biology and Development, Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota
| | - Eric G. Holmberg
- Department of Chemistry, University of Alaska Anchorage, Anchorage, Alaska
| | - Ann M. Parr
- Department of Neurosurgery, Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
28
|
Nieuwenhuis B, Haenzi B, Andrews MR, Verhaagen J, Fawcett JW. Integrins promote axonal regeneration after injury of the nervous system. Biol Rev Camb Philos Soc 2018; 93:1339-1362. [PMID: 29446228 PMCID: PMC6055631 DOI: 10.1111/brv.12398] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 12/23/2017] [Accepted: 01/11/2018] [Indexed: 12/13/2022]
Abstract
Integrins are cell surface receptors that form the link between extracellular matrix molecules of the cell environment and internal cell signalling and the cytoskeleton. They are involved in several processes, e.g. adhesion and migration during development and repair. This review focuses on the role of integrins in axonal regeneration. Integrins participate in spontaneous axonal regeneration in the peripheral nervous system through binding to various ligands that either inhibit or enhance their activation and signalling. Integrin biology is more complex in the central nervous system. Integrins receptors are transported into growing axons during development, but selective polarised transport of integrins limits the regenerative response in adult neurons. Manipulation of integrins and related molecules to control their activation state and localisation within axons is a promising route towards stimulating effective regeneration in the central nervous system.
Collapse
Affiliation(s)
- Bart Nieuwenhuis
- John van Geest Centre for Brain Repair, Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0PYU.K.
- Laboratory for Regeneration of Sensorimotor SystemsNetherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences (KNAW)1105 BAAmsterdamThe Netherlands
| | - Barbara Haenzi
- John van Geest Centre for Brain Repair, Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0PYU.K.
| | | | - Joost Verhaagen
- Laboratory for Regeneration of Sensorimotor SystemsNetherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences (KNAW)1105 BAAmsterdamThe Netherlands
- Centre for Neurogenomics and Cognitive Research, Amsterdam NeuroscienceVrije Universiteit Amsterdam1081 HVAmsterdamThe Netherlands
| | - James W. Fawcett
- John van Geest Centre for Brain Repair, Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0PYU.K.
- Centre of Reconstructive NeuroscienceInstitute of Experimental Medicine142 20Prague 4Czech Republic
| |
Collapse
|
29
|
Soluble TNFα Signaling within the Spinal Cord Contributes to the Development of Autonomic Dysreflexia and Ensuing Vascular and Immune Dysfunction after Spinal Cord Injury. J Neurosci 2018; 38:4146-4162. [PMID: 29610439 DOI: 10.1523/jneurosci.2376-17.2018] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 03/08/2018] [Accepted: 03/14/2018] [Indexed: 12/15/2022] Open
Abstract
Cardiovascular disease and susceptibility to infection are leading causes of morbidity and mortality for individuals with spinal cord injury (SCI). A major contributor to these is autonomic dysreflexia (AD), an amplified reaction of the autonomic nervous system (hallmarked by severe hypertension) in response to sensory stimuli below the injury. Maladaptive plasticity of the spinal sympathetic reflex circuit below the SCI results in AD intensification over time. Mechanisms underlying this maladaptive plasticity are poorly understood, restricting the identification of treatments. Thus, no preventative treatments are currently available. Neuroinflammation has been implicated in other pathologies associated with hyperexcitable neural circuits. Specifically, the soluble form of TNFα (sTNFα) is known to play a role in neuroplasticity. We hypothesize that persistent expression of sTNFα in spinal cord underlies AD exacerbation. To test this, we intrathecally administered XPro1595, a biologic that renders sTNFα nonfunctional, after complete, high-level SCI in female rats. This dramatically attenuated the intensification of colorectal distension-induced and naturally occurring AD events. This improvement is mediated via decreased sprouting of nociceptive primary afferents and activation of the spinal sympathetic reflex circuit. We also examined peripheral vascular function using ex vivo pressurized arterial preparations and immune function via flow cytometric analysis of splenocytes. Diminishing AD via pharmacological inhibition of sTNFα mitigated ensuing vascular hypersensitivity and immune dysfunction. This is the first demonstration that neuroinflammation-induced sTNFα is critical for altering the spinal sympathetic reflex circuit, elucidating a novel mechanism for AD. Importantly, we identify the first potential pharmacological, prophylactic treatment for this life-threatening syndrome.SIGNIFICANCE STATEMENT Autonomic dysreflexia (AD), a disorder that develops after spinal cord injury (SCI) and is hallmarked by sudden, extreme hypertension, contributes to cardiovascular disease and susceptibility to infection, respectively, two leading causes of mortality and morbidity in SCI patients. We demonstrate that neuroinflammation-induced expression of soluble TNFα plays a critical role in AD, elucidating a novel underlying mechanism. We found that intrathecal administration after SCI of a biologic that inhibits soluble TNFα signaling dramatically attenuates AD and significantly reduces AD-associated peripheral vascular and immune dysfunction. We identified mechanisms behind diminished plasticity of neuronal populations within the spinal sympathetic reflex circuit. This study is the first to pinpoint a potential pharmacological, prophylactic strategy to attenuate AD and ensuing cardiovascular and immune dysfunction.
Collapse
|
30
|
Blanquie O, Bradke F. Cytoskeleton dynamics in axon regeneration. Curr Opin Neurobiol 2018; 51:60-69. [PMID: 29544200 DOI: 10.1016/j.conb.2018.02.024] [Citation(s) in RCA: 129] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 02/22/2018] [Accepted: 02/26/2018] [Indexed: 11/19/2022]
Abstract
Recent years have seen cytoskeleton dynamics emerging as a key player in axon regeneration. The cytoskeleton, in particular microtubules and actin, ensures the growth of neuronal processes and maintains the singular, highly polarized shape of neurons. Following injury, adult central axons are tipped by a dystrophic structure, the retraction bulb, which prevents their regeneration. Abnormal cytoskeleton dynamics are responsible for the formation of this growth-incompetent structure but pharmacologically modulating cytoskeleton dynamics of injured axons can transform this structure into a growth-competent growth cone. The cytoskeleton also drives the migration of scar-forming cells after an injury. Targeting its dynamics modifies the composition of the inhibitory environment formed by scar tissue and renders it more permissive for regenerating axons. Hence, cytoskeleton dynamics represent an appealing target to promote axon regeneration. As some of cytoskeleton-targeting drugs are used in the clinics for other purposes, they hold the promise to be used as a basis for a regenerative therapy after a spinal cord injury.
Collapse
Affiliation(s)
- Oriane Blanquie
- Laboratory for Axon Growth and Regeneration, German Center for Neurodegenerative Diseases (DZNE), Sigmund-Freud-Strasse 27, 53127 Bonn, Germany.
| | - Frank Bradke
- Laboratory for Axon Growth and Regeneration, German Center for Neurodegenerative Diseases (DZNE), Sigmund-Freud-Strasse 27, 53127 Bonn, Germany.
| |
Collapse
|
31
|
Urban MW, Ghosh B, Strojny LR, Block CG, Blazejewski SM, Wright MC, Smith GM, Lepore AC. Cell-type specific expression of constitutively-active Rheb promotes regeneration of bulbospinal respiratory axons following cervical SCI. Exp Neurol 2018; 303:108-119. [PMID: 29453976 DOI: 10.1016/j.expneurol.2018.02.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 02/09/2018] [Accepted: 02/12/2018] [Indexed: 12/27/2022]
Abstract
Damage to respiratory neural circuitry and consequent loss of diaphragm function is a major cause of morbidity and mortality in individuals suffering from traumatic cervical spinal cord injury (SCI). Repair of CNS axons after SCI remains a therapeutic challenge, despite current efforts. SCI disrupts inspiratory signals originating in the rostral ventral respiratory group (rVRG) of the medulla from their phrenic motor neuron (PhMN) targets, resulting in loss of diaphragm function. Using a rat model of cervical hemisection SCI, we aimed to restore rVRG-PhMN-diaphragm circuitry by stimulating regeneration of injured rVRG axons via targeted induction of Rheb (ras homolog enriched in brain), a signaling molecule that regulates neuronal-intrinsic axon growth potential. Following C2 hemisection, we performed intra-rVRG injection of an adeno-associated virus serotype-2 (AAV2) vector that drives expression of a constitutively-active form of Rheb (cRheb). rVRG neuron-specific cRheb expression robustly increased mTOR pathway activity within the transduced rVRG neuron population ipsilateral to the hemisection, as assessed by levels of phosphorylated ribosomal S6 kinase. By co-injecting our novel AAV2-mCherry/WGA anterograde/trans-synaptic axonal tracer into rVRG, we found that cRheb expression promoted regeneration of injured rVRG axons into the lesion site, while we observed no rVRG axon regrowth with AAV2-GFP control. AAV2-cRheb also significantly reduced rVRG axonal dieback within the intact spinal cord rostral to the lesion. However, cRheb expression did not promote any recovery of ipsilateral hemi-diaphragm function, as assessed by inspiratory electromyography (EMG) burst amplitudes. This lack of functional recovery was likely because regrowing rVRG fibers did not extend back into the caudal spinal cord to synaptically reinnervate PhMNs that we retrogradely-labeled with cholera toxin B from the ipsilateral hemi-diaphragm. Our findings demonstrate that enhancing neuronal-intrinsic axon growth capacity can promote regeneration of injured bulbospinal respiratory axons after SCI, but this strategy may need to be combined with other manipulations to achieve reconnection of damaged neural circuitry and ultimately recovery of diaphragm function.
Collapse
Affiliation(s)
- Mark W Urban
- Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, 233 South 10th Street, BLSB 245, Philadelphia, PA 19107, United States.
| | - Biswarup Ghosh
- Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, 233 South 10th Street, BLSB 245, Philadelphia, PA 19107, United States.
| | - Laura R Strojny
- Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, 233 South 10th Street, BLSB 245, Philadelphia, PA 19107, United States
| | - Cole G Block
- Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, 233 South 10th Street, BLSB 245, Philadelphia, PA 19107, United States
| | - Sara M Blazejewski
- Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, 233 South 10th Street, BLSB 245, Philadelphia, PA 19107, United States.
| | - Megan C Wright
- Department of Biology, Arcadia University, 450 S. Easton Rd., 220 Boyer Hall, Glenside, PA 19038, United States.
| | - George M Smith
- Department of Neuroscience, Shriners Hospitals for Pediatric Research Center, Temple University School of Medicine, 3500 North Broad Street, Philadelphia, PA 19140-5104, United States.
| | - Angelo C Lepore
- Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, 233 South 10th Street, BLSB 245, Philadelphia, PA 19107, United States.
| |
Collapse
|
32
|
Simmons AB, Bloomsburg SJ, Sukeena JM, Miller CJ, Ortega-Burgos Y, Borghuis BG, Fuerst PG. DSCAM-mediated control of dendritic and axonal arbor outgrowth enforces tiling and inhibits synaptic plasticity. Proc Natl Acad Sci U S A 2017; 114:E10224-E10233. [PMID: 29114051 PMCID: PMC5703318 DOI: 10.1073/pnas.1713548114] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Mature mammalian neurons have a limited ability to extend neurites and make new synaptic connections, but the mechanisms that inhibit such plasticity remain poorly understood. Here, we report that OFF-type retinal bipolar cells in mice are an exception to this rule, as they form new anatomical connections within their tiled dendritic fields well after retinal maturity. The Down syndrome cell-adhesion molecule (Dscam) confines these anatomical rearrangements within the normal tiled fields, as conditional deletion of the gene permits extension of dendrite and axon arbors beyond these borders. Dscam deletion in the mature retina results in expanded dendritic fields and increased cone photoreceptor contacts, demonstrating that DSCAM actively inhibits circuit-level plasticity. Electrophysiological recordings from Dscam-/- OFF bipolar cells showed enlarged visual receptive fields, demonstrating that expanded dendritic territories comprise functional synapses. Our results identify cell-adhesion molecule-mediated inhibition as a regulator of circuit-level neuronal plasticity in the adult retina.
Collapse
Affiliation(s)
- Aaron B Simmons
- Department of Biological Sciences, University of Idaho, Moscow, ID 83844
| | | | - Joshua M Sukeena
- Department of Biological Sciences, University of Idaho, Moscow, ID 83844
| | - Calvin J Miller
- Department of Biological Sciences, University of Idaho, Moscow, ID 83844
| | - Yohaniz Ortega-Burgos
- Department of Chemistry, University of Puerto Rico-Humacao, Humacao Puerto Rico, 00792
| | - Bart G Borghuis
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY 40202;
| | - Peter G Fuerst
- Department of Biological Sciences, University of Idaho, Moscow, ID 83844;
- Washington-Wyoming-Alaska-Montana-Idaho Medical Education Program, University of Washington School of Medicine, Moscow, ID 83844
| |
Collapse
|
33
|
Filous AR, Schwab JM. Determinants of Axon Growth, Plasticity, and Regeneration in the Context of Spinal Cord Injury. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 188:53-62. [PMID: 29030051 DOI: 10.1016/j.ajpath.2017.09.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 09/15/2017] [Accepted: 09/21/2017] [Indexed: 12/30/2022]
Abstract
The mechanisms that underlie recovery after injury of the central nervous system have rarely been definitively established. Axon regrowth remains the major prerequisite for plasticity, regeneration, circuit formation, and eventually functional recovery. The attributed functional relevance of axon regrowth, however, will depend on several subsequent conditional neurobiological modifications, including myelination and synapse formation, but also pruning of aberrant connectivity. Despite the ability to revamp axon outgrowth by altering an increasing number of extracellular and intracellular targets, disentangling which axons are responsible for the recovery of function from those that are functionally silent, or even contributing to aberrant functions, represents a pertinent void in our understanding, challenging the intuitive translational link between anatomical and functional regeneration. Anatomic hallmarks of regeneration are not static and are largely activity dependent. Herein, we survey mechanisms leading to the formation of dystrophic growth cone at the injured axonal tip, the subsequent axonal dieback, and the molecular determinants of axon growth, plasticity, and regeneration in the context of spinal cord injury.
Collapse
Affiliation(s)
- Angela R Filous
- Spinal Cord Injury Division, Department of Neurology, The Ohio State University, Wexner Medical Center, Columbus, Ohio.
| | - Jan M Schwab
- Spinal Cord Injury Division, Department of Neurology, The Ohio State University, Wexner Medical Center, Columbus, Ohio; Department of Neuroscience, The Ohio State University, Wexner Medical Center, Columbus, Ohio; Department of Physical Medicine and Rehabilitation, The Ohio State University, Wexner Medical Center, Columbus, Ohio; Center for Brain and Spinal Cord Repair, Spinal Cord Injury Medicine, The Ohio State University, Wexner Medical Center, Columbus, Ohio.
| |
Collapse
|
34
|
Postinjury Induction of Activated ErbB2 Selectively Hyperactivates Denervated Schwann Cells and Promotes Robust Dorsal Root Axon Regeneration. J Neurosci 2017; 37:10955-10970. [PMID: 28982707 DOI: 10.1523/jneurosci.0903-17.2017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 09/20/2017] [Accepted: 09/27/2017] [Indexed: 11/21/2022] Open
Abstract
Following nerve injury, denervated Schwann cells (SCs) convert to repair SCs, which enable regeneration of peripheral axons. However, the repair capacity of SCs and the regenerative capacity of peripheral axons are limited. In the present studies we examined a potential therapeutic strategy to enhance the repair capacity of SCs, and tested its efficacy in enhancing regeneration of dorsal root (DR) axons, whose regenerative capacity is particularly weak. We used male and female mice of a doxycycline-inducible transgenic line to induce expression of constitutively active ErbB2 (caErbB2) selectively in SCs after DR crush or transection. Two weeks after injury, injured DRs of induced animals contained far more SCs and SC processes. These SCs had not redifferentiated and continued to proliferate. Injured DRs of induced animals also contained far more axons that regrew along SC processes past the transection or crush site. Remarkably, SCs and axons in uninjured DRs remained quiescent, indicating that caErbB2 enhanced regeneration of injured DRs, without aberrantly activating SCs and axons in intact nerves. We also found that intraspinally expressed glial cell line-derived neurotrophic factor (GDNF), but not the removal of chondroitin sulfate proteoglycans, greatly enhanced the intraspinal migration of caErbB2-expressing SCs, enabling robust penetration of DR axons into the spinal cord. These findings indicate that SC-selective, post-injury activation of ErbB2 provides a novel strategy to powerfully enhance the repair capacity of SCs and axon regeneration, without substantial off-target damage. They also highlight that promoting directed migration of caErbB2-expressing SCs by GDNF might be useful to enable axon regrowth in a non-permissive environment.SIGNIFICANCE STATEMENT Repair of injured peripheral nerves remains a critical clinical problem. We currently lack a therapy that potently enhances axon regeneration in patients with traumatic nerve injury. It is extremely challenging to substantially increase the regenerative capacity of damaged nerves without deleterious off-target effects. It was therefore of great interest to discover that caErbB2 markedly enhances regeneration of damaged dorsal roots, while evoking little change in intact roots. To our knowledge, these findings are the first demonstration that repair capacity of denervated SCs can be efficaciously enhanced without altering innervated SCs. Our study also demonstrates that oncogenic ErbB2 signaling can be activated in SCs but not impede transdifferentiation of denervated SCs to regeneration-promoting repair SCs.
Collapse
|
35
|
Santhosh KT, Alizadeh A, Karimi-Abdolrezaee S. Design and optimization of PLGA microparticles for controlled and local delivery of Neuregulin-1 in traumatic spinal cord injury. J Control Release 2017; 261:147-162. [PMID: 28668379 DOI: 10.1016/j.jconrel.2017.06.030] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 06/21/2017] [Accepted: 06/27/2017] [Indexed: 12/17/2022]
Abstract
Spinal cord injury (SCI) results in significant tissue damage that underlies functional impairments. Pharmacological interventions to confer neuroprotection and promote cell replacement are essential for SCI repair. We previously reported that Neuregulin-1 (Nrg-1) is acutely and permanently downregulated after SCI. Nrg-1 is a critical growth factor for differentiation of neural precursor cells (NPCs) into myelinating oligodendrocytes. We showed that intrathecal delivery of Nrg-1 enhances oligodendrocyte replacement following SCI. While an effective delivery system, intrathecal and systemic administration of growth factors with diverse biological targets may pose adverse off-target effects. Here, we have developed and optimized an injectable biodegradable poly(lactic-co-glycolic acid) (PLGA) microparticles system for sustained and prolonged intraspinal delivery of Nrg-1 in SCI. Recombinant human Nrg-1β1 peptide was encapsulated into PLGA microparticles. Optimal Nrg-1 release rate and duration were achieved by manipulating the porosity and size of PLGA particles. Our in vitro analysis showed a direct correlation between particle size and porosity with Nrg-1 release rate, while Nrg-1 loading efficiency in PLGA microparticles was inversely correlated with particle porosity. In SCI, local intraspinal injection of PLGA-Nrg-1 microparticles maintained significantly higher tissue levels of Nrg-1 for a long-term duration compared to Nrg-1 delivered intrathecally by osmotic pumps. Bioactivity of Nrg-1 in PLGA microparticles was verified by promoting oligodendrocyte differentiation of NPCs in vitro, and preservation of oligodendrocytes and axons in SCI. PLGA-Nrg-1 also attenuated neuroinflammation and glial scarring following SCI. We show, for the first time, the feasibility, efficacy and safety of PLGA microparticle system for local and controlled administration of Nrg-1 in SCI.
Collapse
Affiliation(s)
- Kallivalappil T Santhosh
- Regenerative Medicine Program, Department of Physiology and Pathophysiology, Spinal Cord Research Centre, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Arsalan Alizadeh
- Regenerative Medicine Program, Department of Physiology and Pathophysiology, Spinal Cord Research Centre, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Soheila Karimi-Abdolrezaee
- Regenerative Medicine Program, Department of Physiology and Pathophysiology, Spinal Cord Research Centre, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada.
| |
Collapse
|
36
|
Hill CE. A view from the ending: Axonal dieback and regeneration following SCI. Neurosci Lett 2017; 652:11-24. [DOI: 10.1016/j.neulet.2016.11.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 10/20/2016] [Accepted: 11/01/2016] [Indexed: 12/22/2022]
|
37
|
Hoeber J, König N, Trolle C, Lekholm E, Zhou C, Pankratova S, Åkesson E, Fredriksson R, Aldskogius H, Kozlova EN. A Combinatorial Approach to Induce Sensory Axon Regeneration into the Dorsal Root Avulsed Spinal Cord. Stem Cells Dev 2017; 26:1065-1077. [PMID: 28562227 DOI: 10.1089/scd.2017.0019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Spinal root injuries result in newly formed glial scar formation, which prevents regeneration of sensory axons causing permanent sensory loss. Previous studies showed that delivery of trophic factors or implantation of human neural progenitor cells supports sensory axon regeneration and partly restores sensory functions. In this study, we elucidate mechanisms underlying stem cell-mediated ingrowth of sensory axons after dorsal root avulsion (DRA). We show that human spinal cord neural stem/progenitor cells (hscNSPC), and also, mesoporous silica particles loaded with growth factor mimetics (MesoMIM), supported sensory axon regeneration. However, when hscNSPC and MesoMIM were combined, sensory axon regeneration failed. Morphological and tracing analysis showed that sensory axons grow through the newly established glial scar along "bridges" formed by migrating stem cells. Coimplantation of MesoMIM prevented stem cell migration, "bridges" were not formed, and sensory axons failed to enter the spinal cord. MesoMIM applied alone supported sensory axons ingrowth, but without affecting glial scar formation. In vitro, the presence of MesoMIM significantly impaired migration of hscNSPC without affecting their level of differentiation. Our data show that (1) the ability of stem cells to migrate into the spinal cord and organize cellular "bridges" in the newly formed interface is crucial for successful sensory axon regeneration, (2) trophic factor mimetics delivered by mesoporous silica may be a convenient alternative way to induce sensory axon regeneration, and (3) a combinatorial approach of individually beneficial components is not necessarily additive, but can be counterproductive for axonal growth.
Collapse
Affiliation(s)
- Jan Hoeber
- 1 Department of Neuroscience, Uppsala University , Uppsala, Sweden
| | - Niclas König
- 1 Department of Neuroscience, Uppsala University , Uppsala, Sweden
| | - Carl Trolle
- 1 Department of Neuroscience, Uppsala University , Uppsala, Sweden
| | - Emilia Lekholm
- 1 Department of Neuroscience, Uppsala University , Uppsala, Sweden .,2 Department of Pharmaceutical Biosciences, Uppsala University , Uppsala, Sweden
| | | | - Stanislava Pankratova
- 4 Institute of Neuroscience and Pharmacology, University of Copenhagen , Copenhagen, Denmark
| | - Elisabet Åkesson
- 5 Department of Neurobiology, Care Sciences and Society, Karolinska Institutet , Stockholm, Sweden
| | - Robert Fredriksson
- 2 Department of Pharmaceutical Biosciences, Uppsala University , Uppsala, Sweden
| | - Håkan Aldskogius
- 1 Department of Neuroscience, Uppsala University , Uppsala, Sweden
| | - Elena N Kozlova
- 1 Department of Neuroscience, Uppsala University , Uppsala, Sweden
| |
Collapse
|
38
|
Biscola NP, Cartarozzi LP, Ulian-Benitez S, Barbizan R, Castro MV, Spejo AB, Ferreira RS, Barraviera B, Oliveira ALR. Multiple uses of fibrin sealant for nervous system treatment following injury and disease. J Venom Anim Toxins Incl Trop Dis 2017; 23:13. [PMID: 28293254 PMCID: PMC5348778 DOI: 10.1186/s40409-017-0103-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 02/23/2017] [Indexed: 12/14/2022] Open
Abstract
Lesions to the nervous system often produce hemorrhage and tissue loss that are difficult, if not impossible, to repair. Therefore, scar formation, inflammation and cavitation take place, expanding the lesion epicenter. This significantly worsens the patient conditions and impairment, increasing neuronal loss and glial reaction, which in turn further decreases the chances of a positive outcome. The possibility of using hemostatic substances that also function as a scaffold, such as the fibrin sealant, reduces surgical time and improve postoperative recovery. To date, several studies have demonstrated that human blood derived fibrin sealant produces positive effects in different interventions, becoming an efficient alternative to suturing. To provide an alternative to homologous fibrin sealants, the Center for the Study of Venoms and Venomous Animals (CEVAP, Brazil) has proposed a new bioproduct composed of certified animal components, including a thrombin-like enzyme obtained from snake venom and bubaline fibrinogen. Thus, the present review brings up to date literature assessment on the use of fibrin sealant for nervous system repair and positions the new heterologous bioproduct from CEVAP as an alternative to the commercial counterparts. In this way, clinical and pre-clinical data are discussed in different topics, ranging from central nervous system to peripheral nervous system applications, specifying positive results as well as future enhancements that are necessary for improving the use of fibrin sealant therapy.
Collapse
Affiliation(s)
- Natalia Perussi Biscola
- Graduate Program in Tropical Diseases, Botucatu Medical School, Univ Estadual Paulista (UNESP), Botucatu, SP Brazil
- Center for the Study of Venoms and Venomous Animals (CEVAP), Univ Estadual Paulista (UNESP), Botucatu, SP Brazil
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Laboratory of Nerve Regeneration, CEP 13083-970 Campinas, SP Brazil
| | - Luciana Politti Cartarozzi
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Laboratory of Nerve Regeneration, CEP 13083-970 Campinas, SP Brazil
| | - Suzana Ulian-Benitez
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Laboratory of Nerve Regeneration, CEP 13083-970 Campinas, SP Brazil
- Neuro Development Lab, School of Biosciences, University of Birmingham, Birmingham, England UK
| | - Roberta Barbizan
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Laboratory of Nerve Regeneration, CEP 13083-970 Campinas, SP Brazil
- The School of Medicine at Mucuri (FAMMUC), Federal University of Jequitinhonha and Mucuri Valleys (UFVJM), 39803-371 Teófilo Otoni, MG Brazil
| | - Mateus Vidigal Castro
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Laboratory of Nerve Regeneration, CEP 13083-970 Campinas, SP Brazil
| | - Aline Barroso Spejo
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Laboratory of Nerve Regeneration, CEP 13083-970 Campinas, SP Brazil
| | - Rui Seabra Ferreira
- Graduate Program in Tropical Diseases, Botucatu Medical School, Univ Estadual Paulista (UNESP), Botucatu, SP Brazil
- Center for the Study of Venoms and Venomous Animals (CEVAP), Univ Estadual Paulista (UNESP), Botucatu, SP Brazil
| | - Benedito Barraviera
- Graduate Program in Tropical Diseases, Botucatu Medical School, Univ Estadual Paulista (UNESP), Botucatu, SP Brazil
- Center for the Study of Venoms and Venomous Animals (CEVAP), Univ Estadual Paulista (UNESP), Botucatu, SP Brazil
| | - Alexandre Leite Rodrigues Oliveira
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Laboratory of Nerve Regeneration, CEP 13083-970 Campinas, SP Brazil
| |
Collapse
|
39
|
Xia T, Huang B, Ni S, Gao L, Wang J, Wang J, Chen A, Zhu S, Wang B, Li G, Zhu S, Li X. The combination of db-cAMP and ChABC with poly(propylene carbonate) microfibers promote axonal regenerative sprouting and functional recovery after spinal cord hemisection injury. Biomed Pharmacother 2016; 86:354-362. [PMID: 28011383 DOI: 10.1016/j.biopha.2016.12.045] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Revised: 12/08/2016] [Accepted: 12/08/2016] [Indexed: 12/27/2022] Open
Abstract
This study describes the use of poly(propylene carbonate) (PPC) electrospun microfibres impregnated with a combination of dibutyryl cyclic adenosine monophosphate (db-cAMP) and chondroitinase ABC (ChABC) in the treatment of right-side hemisected spinal cord injury (SCI). Release of db-cAMP and/or ChABC from the microfibres was assessed in vitro using high-performance liquid chromatography (HPLC). Drug-impregnated microfibres were implanted into the hemisected thoracic spinal cord of rats, and treatment was evaluated using functional recovery examinations and immunohistochemistry. Our results demonstrated that the microfibres containing db-cAMP and/or ChABC displayed a stable and prolonged release of each agent. Sustained delivery of db-cAMP and/or ChABC was found to promote axonal regenerative sprouting, functional recovery, and reduced glial scar formation when compared to untreated control animals. The combination of both db-cAMP and ChABC was determined to be more effective than using either drug alone in the treatment of SCI. These findings demonstrate the feasibility of using PPC electrospun microfibres for multi-drug combination therapy in SCI.
Collapse
Affiliation(s)
- Tongliang Xia
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, 107# Wenhua Xi Road, Jinan, 250012, PR China
| | - Bin Huang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, 107# Wenhua Xi Road, Jinan, 250012, PR China
| | - Shilei Ni
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, 107# Wenhua Xi Road, Jinan, 250012, PR China
| | - Lei Gao
- Jiyang County People's Hospital of Shandong Provence, 9# Xinyuan Street, Jinyang County, Jinan, 251400, PR China
| | - Jiangang Wang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, 107# Wenhua Xi Road, Jinan, 250012, PR China
| | - Jian Wang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, 107# Wenhua Xi Road, Jinan, 250012, PR China; Department of Biomedicine, University of Bergen, Jonas Lies Vei 91, 5009 Bergen, Norway
| | - Anjing Chen
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, 107# Wenhua Xi Road, Jinan, 250012, PR China
| | - Shaowei Zhu
- Department of Neurology, Qilu Hospital of Shandong University, 107# Wenhua Xi Road, Jinan, 250012, PR China
| | - Benlin Wang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, 107# Wenhua Xi Road, Jinan, 250012, PR China
| | - Gang Li
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, 107# Wenhua Xi Road, Jinan, 250012, PR China
| | - Shugan Zhu
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, 107# Wenhua Xi Road, Jinan, 250012, PR China
| | - Xingang Li
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, 107# Wenhua Xi Road, Jinan, 250012, PR China.
| |
Collapse
|
40
|
Wang Z, Winsor K, Nienhaus C, Hess E, Blackmore MG. Combined chondroitinase and KLF7 expression reduce net retraction of sensory and CST axons from sites of spinal injury. Neurobiol Dis 2016; 99:24-35. [PMID: 27988344 DOI: 10.1016/j.nbd.2016.12.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 12/02/2016] [Accepted: 12/13/2016] [Indexed: 11/26/2022] Open
Abstract
Axon regeneration in the central nervous system is limited both by inhibitory extracellular cues and by an intrinsically low capacity for axon growth in some CNS populations. Chondroitin sulfate proteoglycans (CSPGs) are well-studied inhibitors of axon growth in the CNS, and degradation of CSPGs by chondroitinase has been shown to improve the extension of injured axons. Alternatively, axon growth can be improved by targeting the neuron-intrinsic growth capacity through forced expression of regeneration-associated transcription factors. For example, a transcriptionally active chimera of Krüppel-like Factor 7 (KLF7) and a VP16 domain improves axon growth when expressed in corticospinal tract neurons. Here we tested the hypothesis that combined expression of chondroitinase and VP16-KLF7 would lead to further improvements in axon growth after spinal injury. Chondroitinase was expressed by viral transduction of cells in the spinal cord, while VP16-KLF7 was virally expressed in sensory neurons of the dorsal root ganglia or corticospinal tract (CST) neurons. After transection of the dorsal columns, both chondroitinase and VP16-KLF7 increased the proximity of severed sensory axons to the injury site. Similarly, after complete crush injuries, VP16-KLF7 expression increased the approach of CST axons to the injury site. In neither paradigm however, did single or combined treatment with chondroitinase or VP16-KLF7 enable regenerative growth distal to the injury. These results substantiate a role for CSPG inhibition and low KLF7 activity in determining the net retraction of axons from sites of spinal injury, while suggesting that additional factors act to limit a full regenerative response.
Collapse
Affiliation(s)
- Zimei Wang
- Department of Biomedical Sciences, Marquette University, 53201, USA
| | - Kristen Winsor
- Department of Biomedical Sciences, Marquette University, 53201, USA
| | | | - Evan Hess
- Department of Biomedical Sciences, Marquette University, 53201, USA
| | | |
Collapse
|
41
|
Wu D, Klaw MC, Kholodilov N, Burke RE, Detloff MR, Côté MP, Tom VJ. Expressing Constitutively Active Rheb in Adult Dorsal Root Ganglion Neurons Enhances the Integration of Sensory Axons that Regenerate Across a Chondroitinase-Treated Dorsal Root Entry Zone Following Dorsal Root Crush. Front Mol Neurosci 2016; 9:49. [PMID: 27458339 PMCID: PMC4932115 DOI: 10.3389/fnmol.2016.00049] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 06/07/2016] [Indexed: 11/13/2022] Open
Abstract
While the peripheral branch of dorsal root ganglion neurons (DRG) can successfully regenerate after injury, lesioned central branch axons fail to regrow across the dorsal root entry zone (DREZ), the interface between the dorsal root and the spinal cord. This lack of regeneration is due to the limited regenerative capacity of adult sensory axons and the growth-inhibitory environment at the DREZ, which is similar to that found in the glial scar after a central nervous system (CNS) injury. We hypothesized that transduction of adult DRG neurons using adeno-associated virus (AAV) to express a constitutively-active form of the GTPase Rheb (caRheb) will increase their intrinsic growth potential after a dorsal root crush. Additionally, we posited that if we combined that approach with digestion of upregulated chondroitin sulfate proteoglycans (CSPG) at the DREZ with chondroitinase ABC (ChABC), we would promote regeneration of sensory axons across the DREZ into the spinal cord. We first assessed if this strategy promotes neuritic growth in an in vitro model of the glial scar containing CSPG. ChABC allowed for some regeneration across the once potently inhibitory substrate. Combining ChABC treatment with expression of caRheb in DRG significantly improved this growth. We then determined if this combination strategy also enhanced regeneration through the DREZ after dorsal root crush in adult rats in vivo. After unilaterally crushing C4-T1 dorsal roots, we injected AAV5-caRheb or AAV5-GFP into the ipsilateral C5-C8 DRGs. ChABC or PBS was injected into the ipsilateral dorsal horn at C5-C8 to digest CSPG, for a total of four animal groups (caRheb + ChABC, caRheb + PBS, GFP + ChABC, GFP + PBS). Regeneration was rarely observed in PBS-treated animals, whereas short-distance regrowth across the DREZ was observed in ChABC-treated animals. No difference in axon number or length between the ChABC groups was observed, which may be related to intraganglionic inflammation induced by the injection. ChABC-mediated regeneration is functional, as stimulation of ipsilateral median and ulnar nerves induced neuronal c-Fos expression in deafferented dorsal horn in both ChABC groups. Interestingly, caRheb + ChABC animals had significantly more c-Fos+ nuclei indicating that caRheb expression in DRGs promoted functional synaptogenesis of their axons that regenerated beyond a ChABC-treated DREZ.
Collapse
Affiliation(s)
- Di Wu
- Department of Neurobiology and Anatomy, Drexel University College of Medicine Philadelphia, PA, USA
| | - Michelle C Klaw
- Department of Neurobiology and Anatomy, Drexel University College of Medicine Philadelphia, PA, USA
| | - Nikolai Kholodilov
- Department of Neurology, Columbia University in the City of New York New York, NY, USA
| | - Robert E Burke
- Department of Neurology, Columbia University in the City of New YorkNew York, NY, USA; Department of Pathology and Cell Biology, Columbia University in the City of New YorkNew York, NY, USA
| | - Megan R Detloff
- Department of Neurobiology and Anatomy, Drexel University College of Medicine Philadelphia, PA, USA
| | - Marie-Pascale Côté
- Department of Neurobiology and Anatomy, Drexel University College of Medicine Philadelphia, PA, USA
| | - Veronica J Tom
- Department of Neurobiology and Anatomy, Drexel University College of Medicine Philadelphia, PA, USA
| |
Collapse
|
42
|
Liu Y, Kelamangalath L, Kim H, Han SB, Tang X, Zhai J, Hong JW, Lin S, Son YJ, Smith GM. NT-3 promotes proprioceptive axon regeneration when combined with activation of the mTor intrinsic growth pathway but not with reduction of myelin extrinsic inhibitors. Exp Neurol 2016; 283:73-84. [PMID: 27264357 DOI: 10.1016/j.expneurol.2016.05.021] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 05/13/2016] [Accepted: 05/17/2016] [Indexed: 11/25/2022]
Abstract
Although previous studies have identified several strategies to stimulate regeneration of CNS axons, extensive regeneration and functional recovery have remained a major challenge, particularly for large diameter myelinated axons. Within the CNS, myelin is thought to inhibit axon regeneration, while modulating activity of the mTOR pathway promotes regeneration of injured axons. In this study, we examined NT-3 mediated regeneration of sensory axons through the dorsal root entry zone in a triple knockout of myelin inhibitory proteins or after activation of mTOR using a constitutively active (ca) Rheb in DRG neurons to determine the influence of environmental inhibitory or activation of intrinsic growth pathways could enhance NT-3-mediate regeneration. Loss of myelin inhibitory proteins showed modest enhancement of sensory axon regeneration. In mTOR studies, we found a dramatic age related decrease in the mTOR activation as determined by phosphorylation of the downstream marker S6 ribosomal subunit. Expression of caRheb within adult DRG neurons in vitro increased S6 phosphorylation and doubled the overall length of neurite outgrowth, which was reversed in the presence of rapamycin. In adult female rats, combined expression of caRheb in DRG neurons and NT-3 within the spinal cord increased regeneration of sensory axons almost 3 fold when compared to NT-3 alone. Proprioceptive assessment using a grid runway indicates functionally significant regeneration of large-diameter myelinated sensory afferents. Our results indicate that caRheb-induced increase in mTOR activation enhances neurotrophin-3 induced regeneration of large-diameter myelinated axons.
Collapse
Affiliation(s)
- Yingpeng Liu
- Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Lakshmi Kelamangalath
- Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Hyukmin Kim
- Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Seung Baek Han
- Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Xiaoqing Tang
- Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Jinbin Zhai
- Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Jee W Hong
- Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Shen Lin
- Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Young-Jin Son
- Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - George M Smith
- Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Philadelphia, PA 19140, USA.
| |
Collapse
|
43
|
He Z, Jin Y. Intrinsic Control of Axon Regeneration. Neuron 2016; 90:437-51. [DOI: 10.1016/j.neuron.2016.04.022] [Citation(s) in RCA: 337] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 03/10/2016] [Accepted: 04/13/2016] [Indexed: 01/12/2023]
|
44
|
Affiliation(s)
- Jan Hoeber
- Uppsala University, Department of Neuroscience, Uppsala, Sweden
| |
Collapse
|
45
|
Kwon MJ, Yoon HJ, Kim BG. Regeneration-associated macrophages: a novel approach to boost intrinsic regenerative capacity for axon regeneration. Neural Regen Res 2016; 11:1368-1371. [PMID: 27857723 PMCID: PMC5090822 DOI: 10.4103/1673-5374.191194] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Axons in central nervous system (CNS) do not regenerate spontaneously after injuries such as stroke and traumatic spinal cord injury. Both intrinsic and extrinsic factors are responsible for the regeneration failure. Although intensive research efforts have been invested on extrinsic regeneration inhibitors, the extent to which glial inhibitors contribute to the regeneration failure in vivo still remains elusive. Recent experimental evidence has rekindled interests in intrinsic factors for the regulation of regeneration capacity in adult mammals. In this review, we propose that activating macrophages with pro-regenerative molecular signatures could be a novel approach for boosting intrinsic regenerative capacity of CNS neurons. Using a conditioning injury model in which regeneration of central branches of dorsal root ganglia sensory neurons is enhanced by a preceding injury to the peripheral branches, we have demonstrated that perineuronal macrophages surrounding dorsal root ganglia neurons are critically involved in the maintenance of enhanced regeneration capacity. Neuron-derived chemokine (C-C motif) ligand 2 (CCL2) seems to mediate neuron-macrophage interactions conveying injury signals to perineuronal macrophages taking on a soley pro-regenerative phenotype, which we designate as regeneration-associated macrophages (RAMs). Manipulation of the CCL2 signaling could boost regeneration potential mimicking the conditioning injury, suggesting that the chemokine-mediated RAM activation could be utilized as a regenerative therapeutic strategy for CNS injuries.
Collapse
Affiliation(s)
- Min Jung Kwon
- Department of Brain Science, Ajou University School of Medicine, Suwon, Republic of Korea; Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Republic of Korea
| | - Hyuk Jun Yoon
- Department of Brain Science, Ajou University School of Medicine, Suwon, Republic of Korea; Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Republic of Korea
| | - Byung Gon Kim
- Department of Brain Science, Ajou University School of Medicine, Suwon, Republic of Korea; Department of Neurology, Ajou University School of Medicine, Suwon, Republic of Korea; Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Republic of Korea
| |
Collapse
|
46
|
Toll-Like Receptors and Dectin-1, a C-Type Lectin Receptor, Trigger Divergent Functions in CNS Macrophages. J Neurosci 2015; 35:9966-76. [PMID: 26156997 DOI: 10.1523/jneurosci.0337-15.2015] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
UNLABELLED Spinal cord injury (SCI) activates macrophages, endowing them with both reparative and pathological functions. The mechanisms responsible for these divergent functions are unknown but are likely controlled through stochastic activation of different macrophage receptor subtypes. Various danger-associated molecular patterns released from dying cells in the injured spinal cord likely activate distinct subtypes of macrophage pattern recognition receptors, including bacterial toll-like receptors (TLRs) and fungal C-type lectin receptors (e.g., dectin-1). To determine the in vivo consequences of activating these receptors, ligands specific for TLR2 or dectin-1 were microinjected, alone or in combination, into intact spinal cord. Both ligands elicit a florid macrophage reaction; however, only dectin-1 activation causes macrophage-mediated demyelination and axonal injury. Coactivating TLR2 reduced the injurious effects of dectin-1 activation. When injected into traumatically injured spinal cord, TLR2 agonists enhance the endogenous macrophage reaction while conferring neuroprotection. Indeed, dieback of axons was reduced, leading to smaller lesion volumes at the peak of the macrophage response. Moreover, the density of NG2+ cells expressing vimentin increased in and near lesions that were enriched with TLR2-activated macrophages. In dectin-1-null mutant (knock-out) mice, dieback of corticospinal tract axons also is reduced after SCI. Collectively, these data support the hypothesis that the ability of macrophages to create an axon growth-permissive microenvironment or cause neurotoxicity is receptor dependent and it may be possible to exploit this functional dichotomy to enhance CNS repair. SIGNIFICANCE STATEMENT There is a growing appreciation that macrophages exert diverse functions in the injured and diseased CNS. Indeed, both macrophage-mediated repair and macrophage-mediated injury occur, and often these effector functions are elicited simultaneously. Understanding the mechanisms governing the reparative and pathological properties of activated macrophages is at the forefront of neuroscience research. In this report, using in vitro and in vivo models of relevance to traumatic spinal cord injury (SCI), new data indicate that stochastic activation of toll-like and c-type lectin receptors on macrophages causes neuroprotection or neurotoxicity, respectively. Although this manuscript focuses on SCI, these two innate immune receptor subtypes are also involved in developmental processes and become activated in macrophages that respond to various neurological diseases.
Collapse
|
47
|
DeFrancesco-Lisowitz A, Lindborg JA, Niemi JP, Zigmond RE. The neuroimmunology of degeneration and regeneration in the peripheral nervous system. Neuroscience 2015; 302:174-203. [PMID: 25242643 PMCID: PMC4366367 DOI: 10.1016/j.neuroscience.2014.09.027] [Citation(s) in RCA: 125] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Revised: 09/08/2014] [Accepted: 09/10/2014] [Indexed: 12/25/2022]
Abstract
Peripheral nerves regenerate following injury due to the effective activation of the intrinsic growth capacity of the neurons and the formation of a permissive pathway for outgrowth due to Wallerian degeneration (WD). WD and subsequent regeneration are significantly influenced by various immune cells and the cytokines they secrete. Although macrophages have long been known to play a vital role in the degenerative process, recent work has pointed to their importance in influencing the regenerative capacity of peripheral neurons. In this review, we focus on the various immune cells, cytokines, and chemokines that make regeneration possible in the peripheral nervous system, with specific attention placed on the role macrophages play in this process.
Collapse
Affiliation(s)
| | - J A Lindborg
- Department of Neurosciences, Case Western Reserve University, Cleveland OH 44106-4975
| | - J P Niemi
- Department of Neurosciences, Case Western Reserve University, Cleveland OH 44106-4975
| | - R E Zigmond
- Department of Neurosciences, Case Western Reserve University, Cleveland OH 44106-4975
| |
Collapse
|
48
|
Vadivelu S, Stewart TJ, Qu Y, Horn K, Liu S, Li Q, Silver J, McDonald JW. NG2+ progenitors derived from embryonic stem cells penetrate glial scar and promote axonal outgrowth into white matter after spinal cord injury. Stem Cells Transl Med 2015; 4:401-11. [PMID: 25713464 DOI: 10.5966/sctm.2014-0107] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The glial scar resulting from spinal cord injury is rich in chondroitin sulfate proteoglycan (CSPG), a formidable barrier to axonal regeneration. We explored the possibility of breaching that barrier by first examining the scar in a functional in vitro model. We found that embryonic stem cell-derived neural lineage cells (ESNLCs) with prominent expression of nerve glial antigen 2 (NG2) survived, passed through an increasingly inhibitory gradient of CSPG, and expressed matrix metalloproteinase 9 (MMP-9) at the appropriate stage of their development. Outgrowth of axons from ESNLCs followed because the migrating cells sculpted pathways in which CSPG was degraded. The degradative mechanism involved MMP-9 but not MMP-2. To confirm these results in vivo, we transplanted ESNLCs directly into the cavity of a contused spinal cord 9 days after injury. A week later, ESNLCs survived and were expressing both NG2 and MMP-9. Their axons had grown through long distances (>10 mm), although they preferred to traverse white rather than gray matter. These data are consistent with the concept that expression of inhibitory CSPG within the injury scar is an important impediment to regeneration but that NG2+ progenitors derived from ESNLCs can modify the microenvironment to allow axons to grow through the barrier. This beneficial action may be partly due to developmental expression of MMP-9. We conclude that it might eventually be possible to encourage axonal regeneration in the human spinal cord by transplanting ESNLCs or other cells that express NG2.
Collapse
Affiliation(s)
- Sudhakar Vadivelu
- The International Center for Spinal Cord Injury, Hugo W. Moser Research Institute at the Kennedy Krieger Institute, Baltimore, Maryland, USA; Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri, USA; Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA; Department of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Todd J Stewart
- The International Center for Spinal Cord Injury, Hugo W. Moser Research Institute at the Kennedy Krieger Institute, Baltimore, Maryland, USA; Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri, USA; Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA; Department of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Yun Qu
- The International Center for Spinal Cord Injury, Hugo W. Moser Research Institute at the Kennedy Krieger Institute, Baltimore, Maryland, USA; Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri, USA; Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA; Department of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kevin Horn
- The International Center for Spinal Cord Injury, Hugo W. Moser Research Institute at the Kennedy Krieger Institute, Baltimore, Maryland, USA; Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri, USA; Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA; Department of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Su Liu
- The International Center for Spinal Cord Injury, Hugo W. Moser Research Institute at the Kennedy Krieger Institute, Baltimore, Maryland, USA; Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri, USA; Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA; Department of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Qun Li
- The International Center for Spinal Cord Injury, Hugo W. Moser Research Institute at the Kennedy Krieger Institute, Baltimore, Maryland, USA; Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri, USA; Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA; Department of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jerry Silver
- The International Center for Spinal Cord Injury, Hugo W. Moser Research Institute at the Kennedy Krieger Institute, Baltimore, Maryland, USA; Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri, USA; Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA; Department of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - John W McDonald
- The International Center for Spinal Cord Injury, Hugo W. Moser Research Institute at the Kennedy Krieger Institute, Baltimore, Maryland, USA; Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri, USA; Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA; Department of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
49
|
Neuroinflammation triggered by β-glucan/dectin-1 signaling enables CNS axon regeneration. Proc Natl Acad Sci U S A 2015; 112:2581-6. [PMID: 25675510 DOI: 10.1073/pnas.1423221112] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Innate immunity can facilitate nervous system regeneration, yet the underlying cellular and molecular mechanisms are not well understood. Here we show that intraocular injection of lipopolysaccharide (LPS), a bacterial cell wall component, or the fungal cell wall extract zymosan both lead to rapid and comparable intravitreal accumulation of blood-derived myeloid cells. However, when combined with retro-orbital optic nerve crush injury, lengthy growth of severed retinal ganglion cell (RGC) axons occurs only in zymosan-injected mice, and not in LPS-injected mice. In mice deficient for the pattern recognition receptor dectin-1 but not Toll-like receptor-2 (TLR2), zymosan-mediated RGC regeneration is greatly reduced. The combined loss of dectin-1 and TLR2 completely blocks the proregenerative effects of zymosan. In the retina, dectin-1 is expressed by microglia and dendritic cells, but not by RGCs. Dectin-1 is also present on blood-derived myeloid cells that accumulate in the vitreous. Intraocular injection of the dectin-1 ligand curdlan [a particulate form of β(1, 3)-glucan] promotes optic nerve regeneration comparable to zymosan in WT mice, but not in dectin-1(-/-) mice. Particulate β(1, 3)-glucan leads to increased Erk1/2 MAP-kinase signaling and cAMP response element-binding protein (CREB) activation in myeloid cells in vivo. Loss of the dectin-1 downstream effector caspase recruitment domain 9 (CARD9) blocks CREB activation and attenuates the axon-regenerative effects of β(1, 3)-glucan. Studies with dectin-1(-/-)/WT reciprocal bone marrow chimeric mice revealed a requirement for dectin-1 in both retina-resident immune cells and bone marrow-derived cells for β(1, 3)-glucan-elicited optic nerve regeneration. Collectively, these studies identify a molecular framework of how innate immunity enables repair of injured central nervous system neurons.
Collapse
|
50
|
Mammalian target of rapamycin's distinct roles and effectiveness in promoting compensatory axonal sprouting in the injured CNS. J Neurosci 2015; 34:15347-55. [PMID: 25392502 DOI: 10.1523/jneurosci.1935-14.2014] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Mammalian target of rapamycin (mTOR) functions as a master sensor of nutrients and energy, and controls protein translation and cell growth. Deletion of phosphatase and tensin homolog (PTEN) in adult CNS neurons promotes regeneration of injured axons in an mTOR-dependent manner. However, others have demonstrated mTOR-independent axon regeneration in different cell types, raising the question of how broadly mTOR regulates axonal regrowth across different systems. Here we define the role of mTOR in promoting collateral sprouting of spared axons, a key axonal remodeling mechanism by which functions are recovered after CNS injury. Using pharmacological inhibition, we demonstrate that mTOR is dispensable for the robust spontaneous sprouting of corticospinal tract axons seen after pyramidotomy in postnatal mice. In contrast, moderate spontaneous axonal sprouting and induced-sprouting seen under different conditions in young adult mice (i.e., PTEN deletion or degradation of chondroitin proteoglycans; CSPGs) are both reduced upon mTOR inhibition. In addition, to further determine the potency of mTOR in promoting sprouting responses, we coinactivate PTEN and CSPGs, and demonstrate that this combination leads to an additive increase in axonal sprouting compared with single treatments. Our findings reveal a developmental switch in mTOR dependency for inducing axonal sprouting, and indicate that PTEN deletion in adult neurons neither recapitulates the regrowth program of postnatal animals, nor is sufficient to completely overcome an inhibitory environment. Accordingly, exploiting mTOR levels by targeting PTEN combined with CSPG degradation represents a promising strategy to promote extensive axonal plasticity in adult mammals.
Collapse
|