1
|
Kim SJ, Ko WK, Han GH, Lee D, Cho MJ, Sheen SH, Sohn S. Axon guidance gene-targeted siRNA delivery system improves neural stem cell transplantation therapy after spinal cord injury. Biomater Res 2023; 27:101. [PMID: 37840145 PMCID: PMC10577901 DOI: 10.1186/s40824-023-00434-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 09/18/2023] [Indexed: 10/17/2023] Open
Abstract
BACKGROUND Neural stem cells (NSCs) derived from the embryonic spinal cord are excellent candidates for the cellular regeneration of lost neural cells after spinal cord injury (SCI). Semaphorin 3 A (Sema3A) is well known as being implicated in the major axon guidance of the growth cone as a repulsive function during the development of the central nervous system, yet its function in NSC transplantation therapy for SCI has not been investigated. Here, we report for the first time that embryonic spinal cord-derived NSCs significantly express Sema3A in the SCI environment, potentially facilitating inhibition of cell proliferation after transplantation. METHODS siRNA-Sema3A was conjugated with poly-l-lysin-coated gold nanoparticles (AuNPs) through a charge interaction process. NSCs were isolated from embryonic spinal cords of rats. Then, the cells were embedded into a dual-degradable hydrogel with the siRNA- Sema3A loaded-AuNPs and transplanted after complete SCI in rats. RESULTS The knockdown of Sema3A by delivering siRNA nanoparticles via dual-degradable hydrogels led to a significant increase in cell survival and neuronal differentiation of the transplanted NSCs after SCI. Of note, the knockdown of Sema3A increased the synaptic connectivity of transplanted NSC in the injured spinal cord. Moreover, extracellular matrix molecule and functional recovery were significantly improved in Sema3A-inhibited rats compared to those in rats with only NSCs transplanted. CONCLUSIONS These findings demonstrate the important role of Sema3A in NSC transplantation therapy, which may be considered as a future cell transplantation therapy for SCI cases.
Collapse
Affiliation(s)
- Seong Jun Kim
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, 59, Yatap-ro, Bundang- gu, Seongnam-si, 13496, Gyeonggi-do, Republic of Korea
- Department of Biomedical Science, CHA University, 335, Pangyo-ro, Bundang-gu, Seongnam-si, 13488, Gyeonggi-do, Republic of Korea
| | - Wan-Kyu Ko
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, 59, Yatap-ro, Bundang- gu, Seongnam-si, 13496, Gyeonggi-do, Republic of Korea
- Department of Biomedical Science, CHA University, 335, Pangyo-ro, Bundang-gu, Seongnam-si, 13488, Gyeonggi-do, Republic of Korea
| | - Gong Ho Han
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, 59, Yatap-ro, Bundang- gu, Seongnam-si, 13496, Gyeonggi-do, Republic of Korea
- Department of Biomedical Science, CHA University, 335, Pangyo-ro, Bundang-gu, Seongnam-si, 13488, Gyeonggi-do, Republic of Korea
| | - Daye Lee
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, 59, Yatap-ro, Bundang- gu, Seongnam-si, 13496, Gyeonggi-do, Republic of Korea
- Department of Biomedical Science, CHA University, 335, Pangyo-ro, Bundang-gu, Seongnam-si, 13488, Gyeonggi-do, Republic of Korea
| | - Min Jai Cho
- Department of Neurosurgery, Chungbuk National University, 776, 1Sunhawn-ro, Seowon-gu, Cheongju-si, 28644, Republic of Korea
| | - Seung Hun Sheen
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, 59, Yatap-ro, Bundang- gu, Seongnam-si, 13496, Gyeonggi-do, Republic of Korea
| | - Seil Sohn
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, 59, Yatap-ro, Bundang- gu, Seongnam-si, 13496, Gyeonggi-do, Republic of Korea.
- Department of Biomedical Science, CHA University, 335, Pangyo-ro, Bundang-gu, Seongnam-si, 13488, Gyeonggi-do, Republic of Korea.
| |
Collapse
|
2
|
Shafqat A, Albalkhi I, Magableh HM, Saleh T, Alkattan K, Yaqinuddin A. Tackling the glial scar in spinal cord regeneration: new discoveries and future directions. Front Cell Neurosci 2023; 17:1180825. [PMID: 37293626 PMCID: PMC10244598 DOI: 10.3389/fncel.2023.1180825] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 05/08/2023] [Indexed: 06/10/2023] Open
Abstract
Axonal regeneration and functional recovery are poor after spinal cord injury (SCI), typified by the formation of an injury scar. While this scar was traditionally believed to be primarily responsible for axonal regeneration failure, current knowledge takes a more holistic approach that considers the intrinsic growth capacity of axons. Targeting the SCI scar has also not reproducibly yielded nearly the same efficacy in animal models compared to these neuron-directed approaches. These results suggest that the major reason behind central nervous system (CNS) regeneration failure is not the injury scar but a failure to stimulate axon growth adequately. These findings raise questions about whether targeting neuroinflammation and glial scarring still constitute viable translational avenues. We provide a comprehensive review of the dual role of neuroinflammation and scarring after SCI and how future research can produce therapeutic strategies targeting the hurdles to axonal regeneration posed by these processes without compromising neuroprotection.
Collapse
|
3
|
Fibrotic Scar in CNS Injuries: From the Cellular Origins of Fibroblasts to the Molecular Processes of Fibrotic Scar Formation. Cells 2022; 11:cells11152371. [PMID: 35954214 PMCID: PMC9367779 DOI: 10.3390/cells11152371] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/29/2022] [Accepted: 07/30/2022] [Indexed: 02/06/2023] Open
Abstract
Central nervous system (CNS) trauma activates a persistent repair response that leads to fibrotic scar formation within the lesion. This scarring is similar to other organ fibrosis in many ways; however, the unique features of the CNS differentiate it from other organs. In this review, we discuss fibrotic scar formation in CNS trauma, including the cellular origins of fibroblasts, the mechanism of fibrotic scar formation following an injury, as well as the implication of the fibrotic scar in CNS tissue remodeling and regeneration. While discussing the shared features of CNS fibrotic scar and fibrosis outside the CNS, we highlight their differences and discuss therapeutic targets that may enhance regeneration in the CNS.
Collapse
|
4
|
Cooke P, Janowitz H, Dougherty SE. Neuronal Redevelopment and the Regeneration of Neuromodulatory Axons in the Adult Mammalian Central Nervous System. Front Cell Neurosci 2022; 16:872501. [PMID: 35530177 PMCID: PMC9074815 DOI: 10.3389/fncel.2022.872501] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 03/24/2022] [Indexed: 01/09/2023] Open
Abstract
One reason that many central nervous system injuries, including those arising from traumatic brain injury, spinal cord injury, and stroke, have limited recovery of function is that neurons within the adult mammalian CNS lack the ability to regenerate their axons following trauma. This stands in contrast to neurons of the adult mammalian peripheral nervous system (PNS). New evidence, provided by single-cell expression profiling, suggests that, following injury, both mammalian central and peripheral neurons can revert to an embryonic-like growth state which is permissive for axon regeneration. This “redevelopment” strategy could both facilitate a damage response necessary to isolate and repair the acute damage from injury and provide the intracellular machinery necessary for axon regrowth. Interestingly, serotonin neurons of the rostral group of raphe nuclei, which project their axons into the forebrain, display a robust ability to regenerate their axons unaided, counter to the widely held view that CNS axons cannot regenerate without experimental intervention after injury. Furthermore, initial evidence suggests that norepinephrine neurons within the locus coeruleus possess similar regenerative abilities. Several morphological characteristics of serotonin axon regeneration in adult mammals, observable using longitudinal in vivo imaging, are distinct from the known characteristics of unaided peripheral nerve regeneration, or of the regeneration seen in the spinal cord and optic nerve that occurs with experimental intervention. These results suggest that there is an alternative CNS program for axon regeneration that likely differs from that displayed by the PNS.
Collapse
Affiliation(s)
- Patrick Cooke
- Linden Lab, Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Haley Janowitz
- Linden Lab, Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Sarah E Dougherty
- Linden Lab, Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
5
|
Characterization of a Novel Aspect of Tissue Scarring Following Experimental Spinal Cord Injury and the Implantation of Bioengineered Type-I Collagen Scaffolds in the Adult Rat: Involvement of Perineurial-like Cells? Int J Mol Sci 2022; 23:ijms23063221. [PMID: 35328642 PMCID: PMC8954100 DOI: 10.3390/ijms23063221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 03/04/2022] [Accepted: 03/15/2022] [Indexed: 11/17/2022] Open
Abstract
Numerous intervention strategies have been developed to promote functional tissue repair following experimental spinal cord injury (SCI), including the bridging of lesion-induced cystic cavities with bioengineered scaffolds. Integration between such implanted scaffolds and the lesioned host spinal cord is critical for supporting regenerative growth, but only moderate-to-low degrees of success have been reported. Light and electron microscopy were employed to better characterise the fibroadhesive scarring process taking place after implantation of a longitudinally microstructured type-I collagen scaffold into unilateral mid-cervical resection injuries of the adult rat spinal cord. At long survival times (10 weeks post-surgery), sheets of tightly packed cells (of uniform morphology) could be seen lining the inner surface of the repaired dura mater of lesion-only control animals, as well as forming a barrier along the implant–host interface of the scaffold-implanted animals. The highly uniform ultrastructural features of these scarring cells and their anatomical continuity with the local, reactive spinal nerve roots strongly suggest their identity to be perineurial-like cells. This novel aspect of the cellular composition of reactive spinal cord tissue highlights the increasingly complex nature of fibroadhesive scarring involved in traumatic injury, and particularly in response to the implantation of bioengineered collagen scaffolds.
Collapse
|
6
|
Zhang Q, Li Y, Zhuo Y. Synaptic or Non-synaptic? Different Intercellular Interactions with Retinal Ganglion Cells in Optic Nerve Regeneration. Mol Neurobiol 2022; 59:3052-3072. [PMID: 35266115 PMCID: PMC9016027 DOI: 10.1007/s12035-022-02781-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 02/24/2022] [Indexed: 12/31/2022]
Abstract
Axons of adult neurons in the mammalian central nervous system generally fail to regenerate by themselves, and few if any therapeutic options exist to reverse this situation. Due to a weak intrinsic potential for axon growth and the presence of strong extrinsic inhibitors, retinal ganglion cells (RGCs) cannot regenerate their axons spontaneously after optic nerve injury and eventually undergo apoptosis, resulting in permanent visual dysfunction. Regarding the extracellular environment, research to date has generally focused on glial cells and inflammatory cells, while few studies have discussed the potentially significant role of interneurons that make direct connections with RGCs as part of the complex retinal circuitry. In this study, we provide a novel angle to summarize these extracellular influences following optic nerve injury as "intercellular interactions" with RGCs and classify these interactions as synaptic and non-synaptic. By discussing current knowledge of non-synaptic (glial cells and inflammatory cells) and synaptic (mostly amacrine cells and bipolar cells) interactions, we hope to accentuate the previously neglected but significant effects of pre-synaptic interneurons and bring unique insights into future pursuit of optic nerve regeneration and visual function recovery.
Collapse
Affiliation(s)
- Qi Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Yiqing Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou, 510060, China.
| | - Yehong Zhuo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou, 510060, China.
| |
Collapse
|
7
|
Neurogenetic disorders across the lifespan: from aberrant development to degeneration. Nat Rev Neurol 2022; 18:117-124. [PMID: 34987232 PMCID: PMC10132523 DOI: 10.1038/s41582-021-00595-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/23/2021] [Indexed: 02/08/2023]
Abstract
Intellectual disability and autism spectrum disorder (ASD) are common, and genetic testing is increasingly performed in individuals with these diagnoses to inform prognosis, refine management and provide information about recurrence risk in the family. For neurogenetic conditions associated with intellectual disability and ASD, data on natural history in adults are scarce; however, as older adults with these disorders are identified, it is becoming clear that some conditions are associated with both neurodevelopmental problems and neurodegeneration. Moreover, emerging evidence indicates that some neurogenetic conditions associated primarily with neurodegeneration also affect neurodevelopment. In this Perspective, we discuss examples of diseases that have developmental and degenerative overlap. We propose that neurogenetic disorders should be studied continually across the lifespan to understand the roles of the affected genes in brain development and maintenance, and to inform strategies for treatment.
Collapse
|
8
|
Paramasivam A, Mickymaray S, Jayakumar S, Jeraud M, Perumal P, Alassaf A, Aljabr AA, Dasarathy S, Rangasamy SB. Locomotor Behavior Analysis in Spinal Cord Injured Macaca radiata after Predegenerated Peripheral Nerve Grafting-A Preliminary Evidence. Vet Sci 2021; 8:vetsci8120288. [PMID: 34941815 PMCID: PMC8707676 DOI: 10.3390/vetsci8120288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/16/2021] [Accepted: 11/19/2021] [Indexed: 11/16/2022] Open
Abstract
INTRODUCTION Primate animal models are being utilized to explore novel therapies for spinal cord injuries. This study aimed to evaluate the efficiency of the transplantation of predegenerated nerve segments in unilateral spinal cord-hemisected bonnet monkeys' (Macaca radiata) locomotor functions using the complex runways. MATERIALS AND METHODS The bonnet monkeys were initially trained to walk in a bipedal motion on grid and staircase runways. In one group of trained monkeys, surgical hemisection was made in the spinal cord at the T12-L1 level. In the other group, hemisection was induced in the spinal cord, and the ulnar nerve was also transected at the same time (transplant group). After one week, the hemisected cavity was reopened and implanted with predegenerated ulnar nerve segments obtained from the same animal of the transplant group. RESULTS All the operated monkeys showed significant deficits in locomotion on runways at the early postoperative period. The walking ability of operated monkeys was found to be gradually improved, and they recovered nearer to preoperative level at the fourth postoperative month, and there were no marked differences. CONCLUSION The results demonstrate that there were no significant improvements in the locomotion of monkeys on runways after the delayed grafting of nerve segments until one year later. The failure of the predegenerated nerve graft as a possible therapeutic strategy to improve the locomotion of monkeys may be due to a number of factors set in motion by trauma, which could possibly prevent the qualities of regeneration. The exact reason for this ineffectiveness of predegenerated nerve segments and their underlying mechanism is not known.
Collapse
Affiliation(s)
- Anand Paramasivam
- Department of Basic Medical Sciences, College of Dentistry, Majmaah University, Al-Majmaah 11952, Saudi Arabia;
- Department of Physiology, Dr. ALM PGIBMS, University of Madras, Chennai 600113, India; (M.J.); (P.P.); (S.B.R.)
- Department of Medical Education, College of Dentistry, Majmaah University, Majmaah 11952, Saudi Arabia;
- Correspondence:
| | - Suresh Mickymaray
- Department of Biology, College of Science, Majmaah University, Majmaah 11952, Saudi Arabia;
| | - Saikarthik Jayakumar
- Department of Basic Medical Sciences, College of Dentistry, Majmaah University, Al-Majmaah 11952, Saudi Arabia;
- Department of Medical Education, College of Dentistry, Majmaah University, Majmaah 11952, Saudi Arabia;
| | - Mathew Jeraud
- Department of Physiology, Dr. ALM PGIBMS, University of Madras, Chennai 600113, India; (M.J.); (P.P.); (S.B.R.)
- Department of Physiology, Ibn Sina National College for Medical Studies, Jeddah 22421, Saudi Arabia
| | - Periasamy Perumal
- Department of Physiology, Dr. ALM PGIBMS, University of Madras, Chennai 600113, India; (M.J.); (P.P.); (S.B.R.)
- Department of Physiology, Vellore Medical College, Vellore 632002, India
| | - Abdullah Alassaf
- Department of Preventive Dental Sciences, College of Dentistry, Majmaah University, Majmaah 11952, Saudi Arabia;
| | | | - Sridevi Dasarathy
- Department of Neurological Sciences, Rush University Medical Centre, Chicago, IL 60612, USA;
| | - Suresh Babu Rangasamy
- Department of Physiology, Dr. ALM PGIBMS, University of Madras, Chennai 600113, India; (M.J.); (P.P.); (S.B.R.)
- Department of Neurological Sciences, Rush University Medical Centre, Chicago, IL 60612, USA;
| |
Collapse
|
9
|
Dias DO, Kalkitsas J, Kelahmetoglu Y, Estrada CP, Tatarishvili J, Holl D, Jansson L, Banitalebi S, Amiry-Moghaddam M, Ernst A, Huttner HB, Kokaia Z, Lindvall O, Brundin L, Frisén J, Göritz C. Pericyte-derived fibrotic scarring is conserved across diverse central nervous system lesions. Nat Commun 2021; 12:5501. [PMID: 34535655 PMCID: PMC8448846 DOI: 10.1038/s41467-021-25585-5] [Citation(s) in RCA: 121] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 08/17/2021] [Indexed: 12/04/2022] Open
Abstract
Fibrotic scar tissue limits central nervous system regeneration in adult mammals. The extent of fibrotic tissue generation and distribution of stromal cells across different lesions in the brain and spinal cord has not been systematically investigated in mice and humans. Furthermore, it is unknown whether scar-forming stromal cells have the same origin throughout the central nervous system and in different types of lesions. In the current study, we compared fibrotic scarring in human pathological tissue and corresponding mouse models of penetrating and non-penetrating spinal cord injury, traumatic brain injury, ischemic stroke, multiple sclerosis and glioblastoma. We show that the extent and distribution of stromal cells are specific to the type of lesion and, in most cases, similar between mice and humans. Employing in vivo lineage tracing, we report that in all mouse models that develop fibrotic tissue, the primary source of scar-forming fibroblasts is a discrete subset of perivascular cells, termed type A pericytes. Perivascular cells with a type A pericyte marker profile also exist in the human brain and spinal cord. We uncover type A pericyte-derived fibrosis as a conserved mechanism that may be explored as a therapeutic target to improve recovery after central nervous system lesions.
Collapse
Affiliation(s)
- David O Dias
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Jannis Kalkitsas
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Yildiz Kelahmetoglu
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Cynthia P Estrada
- Department of Clinical Neuroscience, Karolinska University Hospital, Solna, Sweden
| | | | - Daniel Holl
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Linda Jansson
- Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Shervin Banitalebi
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Mahmood Amiry-Moghaddam
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Aurélie Ernst
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
- Group Genome Instability in Tumors, German Cancer Research Center, Heidelberg, Germany
| | - Hagen B Huttner
- Department of Neurology, University Hospital Erlangen, Erlangen, Germany
| | - Zaal Kokaia
- Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Olle Lindvall
- Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Lou Brundin
- Department of Clinical Neuroscience, Karolinska University Hospital, Solna, Sweden
| | - Jonas Frisén
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Christian Göritz
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden.
- Ming Wai Lau Centre for Reparative Medicine, Stockholm Node, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
10
|
O'Shea SA, Hickman RA, Cortes E, Vonsattel JP, Fahn S, Okur V, Alcalay RN, Chung WK. Neuropathological Findings in a Case of Parkinsonism and Developmental Delay Associated with a Monoallelic Variant in PLXNA1. Mov Disord 2021; 36:2681-2687. [PMID: 34415653 DOI: 10.1002/mds.28756] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 07/02/2021] [Accepted: 07/19/2021] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND PLXNA1 encodes for Plexin-A, a transmembrane protein expressed in the developing nervous system. Mutations in this gene have been associated with developmental delay but have not been previously associated with the development of parkinsonism. OBJECTIVES To describe the case of a 38-year-old patient with developmental delay who developed parkinsonism later in life. METHODS Post-mortem exome sequencing was performed with confirmation by Sanger sequencing. Brain autopsy was also performed. RESULTS Post-mortem exome sequencing on the proband identified a heterozygous predicted nonsense PLXNA1 variant (c.G3361T:p.Glu1121Ter). Pathology demonstrated arhinencephaly with brainstem heterotopia, diffuse Lewy body disease, and frontotemporal lobar dementia-tau. CONCLUSIONS This case of a patient with developmental delay and parkinsonism with PLXNA1 mutation highlights a need for assessing long-term outcomes of individuals with neurodevelopmental disorders, as well as the need for genetic testing in adults. It also suggests that the link between PLXNA1 and α-synuclein should be explored in the future. © 2021 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Sarah A O'Shea
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, USA.,Department of Neurology, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| | - Richard A Hickman
- Department of Pathology, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| | - Etty Cortes
- Department of Pathology, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA.,Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Jean Paul Vonsattel
- Department of Pathology, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| | - Stanley Fahn
- Department of Neurology, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| | - Volkan Okur
- Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Roy N Alcalay
- Department of Neurology, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| | - Wendy K Chung
- Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| |
Collapse
|
11
|
Unraveling Axon Guidance during Axotomy and Regeneration. Int J Mol Sci 2021; 22:ijms22158344. [PMID: 34361110 PMCID: PMC8347220 DOI: 10.3390/ijms22158344] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 07/28/2021] [Accepted: 07/29/2021] [Indexed: 02/06/2023] Open
Abstract
During neuronal development and regeneration axons extend a cytoskeletal-rich structure known as the growth cone, which detects and integrates signals to reach its final destination. The guidance cues “signals” bind their receptors, activating signaling cascades that result in the regulation of the growth cone cytoskeleton, defining growth cone advance, pausing, turning, or collapse. Even though much is known about guidance cues and their isolated mechanisms during nervous system development, there is still a gap in the understanding of the crosstalk between them, and about what happens after nervous system injuries. After neuronal injuries in mammals, only axons in the peripheral nervous system are able to regenerate, while the ones from the central nervous system fail to do so. Therefore, untangling the guidance cues mechanisms, as well as their behavior and characterization after axotomy and regeneration, are of special interest for understanding and treating neuronal injuries. In this review, we present findings on growth cone guidance and canonical guidance cues mechanisms, followed by a description and comparison of growth cone pathfinding mechanisms after axotomy, in regenerative and non-regenerative animal models.
Collapse
|
12
|
An Extracellular Perspective on CNS Maturation: Perineuronal Nets and the Control of Plasticity. Int J Mol Sci 2021; 22:ijms22052434. [PMID: 33670945 PMCID: PMC7957817 DOI: 10.3390/ijms22052434] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/24/2021] [Accepted: 02/24/2021] [Indexed: 02/07/2023] Open
Abstract
During restricted time windows of postnatal life, called critical periods, neural circuits are highly plastic and are shaped by environmental stimuli. In several mammalian brain areas, from the cerebral cortex to the hippocampus and amygdala, the closure of the critical period is dependent on the formation of perineuronal nets. Perineuronal nets are a condensed form of an extracellular matrix, which surrounds the soma and proximal dendrites of subsets of neurons, enwrapping synaptic terminals. Experimentally disrupting perineuronal nets in adult animals induces the reactivation of critical period plasticity, pointing to a role of the perineuronal net as a molecular brake on plasticity as the critical period closes. Interestingly, in the adult brain, the expression of perineuronal nets is remarkably dynamic, changing its plasticity-associated conditions, including memory processes. In this review, we aimed to address how perineuronal nets contribute to the maturation of brain circuits and the regulation of adult brain plasticity and memory processes in physiological and pathological conditions.
Collapse
|
13
|
Zou Y. Targeting axon guidance cues for neural circuit repair after spinal cord injury. J Cereb Blood Flow Metab 2021; 41:197-205. [PMID: 33167744 PMCID: PMC7812507 DOI: 10.1177/0271678x20961852] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 09/02/2020] [Accepted: 08/28/2020] [Indexed: 12/12/2022]
Abstract
At least two-thirds of spinal cord injury cases are anatomically incomplete, without complete spinal cord transection, although the initial injuries cause complete loss of sensory and motor functions. The malleability of neural circuits and networks allows varied extend of functional restoration in some individuals after successful rehabilitative training. However, in most cases, the efficiency and extent are both limited and uncertain, largely due to the many obstacles of repair. The restoration of function after anatomically incomplete injury is in part made possible by the growth of new axons or new axon branches through the spared spinal cord tissue and the new synaptic connections they make, either along the areas they grow through or in the areas they terminate. This review will discuss new progress on the understanding of the role of axon guidance molecules, particularly the Wnt family proteins, in spinal cord injury and how the knowledge and tools of axon guidance can be applied to increase the potential of recovery. These strategies, combined with others, such as neuroprotection and rehabilitation, may bring new promises. The recovery strategies for anatomically incomplete spinal cord injuries are relevant and may be applicable to traumatic brain injury and stroke.
Collapse
Affiliation(s)
- Yimin Zou
- Neurobiology Section, Biological Sciences
Division, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
14
|
Ueno M, Nakamura Y, Nakagawa H, Niehaus JK, Maezawa M, Gu Z, Kumanogoh A, Takebayashi H, Lu QR, Takada M, Yoshida Y. Olig2-Induced Semaphorin Expression Drives Corticospinal Axon Retraction After Spinal Cord Injury. Cereb Cortex 2020; 30:5702-5716. [PMID: 32564090 DOI: 10.1093/cercor/bhaa142] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 05/02/2020] [Accepted: 05/03/2020] [Indexed: 12/24/2022] Open
Abstract
Axon regeneration is limited in the central nervous system, which hinders the reconstruction of functional circuits following spinal cord injury (SCI). Although various extrinsic molecules to repel axons following SCI have been identified, the role of semaphorins, a major class of axon guidance molecules, has not been thoroughly explored. Here we show that expression of semaphorins, including Sema5a and Sema6d, is elevated after SCI, and genetic deletion of either molecule or their receptors (neuropilin1 and plexinA1, respectively) suppresses axon retraction or dieback in injured corticospinal neurons. We further show that Olig2+ cells are essential for SCI-induced semaphorin expression, and that Olig2 binds to putative enhancer regions of the semaphorin genes. Finally, conditional deletion of Olig2 in the spinal cord reduces the expression of semaphorins, alleviating the axon retraction. These results demonstrate that semaphorins function as axon repellents following SCI, and reveal a novel transcriptional mechanism for controlling semaphorin levels around injured neurons to create zones hostile to axon regrowth.
Collapse
Affiliation(s)
- Masaki Ueno
- Department of System Pathology for Neurological Disorders, Brain Research Institute, Niigata University, Niigata 951-8585, Japan.,Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.,Precursory Research for Embryonic Science and Technology (PRESTO), Japan Science and Technology Agency (JST), Kawaguchi 332-0012, Japan
| | - Yuka Nakamura
- Department of System Pathology for Neurological Disorders, Brain Research Institute, Niigata University, Niigata 951-8585, Japan.,Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Hiroshi Nakagawa
- Systems Neuroscience Section, Primate Research Institute, Kyoto University, Inuyama 484-8506, Japan.,Department of Molecular Neuroscience, WPI Immunology Frontier Research Center, Osaka University, Suita 565-0871, Japan
| | - Jesse K Niehaus
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.,Precursory Research for Embryonic Science and Technology (PRESTO), Japan Science and Technology Agency (JST), Kawaguchi 332-0012, Japan
| | - Mari Maezawa
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Zirong Gu
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.,Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine, Allergy and Rheumatic Diseases, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan
| | - Hirohide Takebayashi
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Japan
| | - Qing Richard Lu
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Masahiko Takada
- Systems Neuroscience Section, Primate Research Institute, Kyoto University, Inuyama 484-8506, Japan
| | - Yutaka Yoshida
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.,Neural Connectivity Development in Physiology and Disease Laboratory, Burke Neurological Institute, White Plains, NY 10605, USA.,Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| |
Collapse
|
15
|
Harris JM, Wang AYD, Boulanger-Weill J, Santoriello C, Foianini S, Lichtman JW, Zon LI, Arlotta P. Long-Range Optogenetic Control of Axon Guidance Overcomes Developmental Boundaries and Defects. Dev Cell 2020; 53:577-588.e7. [PMID: 32516597 PMCID: PMC7375170 DOI: 10.1016/j.devcel.2020.05.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 03/12/2020] [Accepted: 05/11/2020] [Indexed: 01/12/2023]
Abstract
Axons connect neurons together, establishing the wiring architecture of neuronal networks. Axonal connectivity is largely built during embryonic development through highly constrained processes of axon guidance, which have been extensively studied. However, the inability to control axon guidance, and thus neuronal network architecture, has limited investigation of how axonal connections influence subsequent development and function of neuronal networks. Here, we use zebrafish motor neurons expressing a photoactivatable Rac1 to co-opt endogenous growth cone guidance machinery to precisely and non-invasively direct axon growth using light. Axons can be guided over large distances, within complex environments of living organisms, overriding competing endogenous signals and redirecting axons across potent repulsive barriers to construct novel circuitry. Notably, genetic axon guidance defects can be rescued, restoring functional connectivity. These data demonstrate that intrinsic growth cone guidance machinery can be co-opted to non-invasively build new connectivity, allowing investigation of neural network dynamics in intact living organisms.
Collapse
Affiliation(s)
- James M. Harris
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts 02138, USA.,Harvard-MIT Division of Health Sciences and Technology, Harvard Medical School, Boston, MA 02115, USA.,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02138, USA
| | - Andy Yu-Der Wang
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts 02138, USA.,Current Address: Tufts University School of Medicine, Boston, MA 02115, USA
| | - Jonathan Boulanger-Weill
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts 02138, USA
| | - Cristina Santoriello
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts 02138, USA.,Stem Cell Program and Division of Hematology/Oncology, Children’s Hospital and Dana Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Medical School, Harvard Stem Cell Institute, Stem Cell and Regenerative Biology Department, Harvard University, Boston, MA 02115, USA
| | - Stephan Foianini
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts 02138, USA
| | - Jeff W. Lichtman
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts 02138, USA.,Center for Brain Science, Harvard University, Cambridge, Massachusetts 02138, USA
| | - Leonard I. Zon
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts 02138, USA.,Stem Cell Program and Division of Hematology/Oncology, Children’s Hospital and Dana Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Medical School, Harvard Stem Cell Institute, Stem Cell and Regenerative Biology Department, Harvard University, Boston, MA 02115, USA
| | - Paola Arlotta
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts 02138, USA.,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02138, USA.,Lead contact. Correspondence:
| |
Collapse
|
16
|
Neuroprotective Strategies for Retinal Ganglion Cell Degeneration: Current Status and Challenges Ahead. Int J Mol Sci 2020; 21:ijms21072262. [PMID: 32218163 PMCID: PMC7177277 DOI: 10.3390/ijms21072262] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 03/19/2020] [Accepted: 03/20/2020] [Indexed: 12/12/2022] Open
Abstract
The retinal ganglion cells (RGCs) are the output cells of the retina into the brain. In mammals, these cells are not able to regenerate their axons after optic nerve injury, leaving the patients with optic neuropathies with permanent visual loss. An effective RGCs-directed therapy could provide a beneficial effect to prevent the progression of the disease. Axonal injury leads to the functional loss of RGCs and subsequently induces neuronal death, and axonal regeneration would be essential to restore the neuronal connectivity, and to reestablish the function of the visual system. The manipulation of several intrinsic and extrinsic factors has been proposed in order to stimulate axonal regeneration and functional repairing of axonal connections in the visual pathway. However, there is a missing point in the process since, until now, there is no therapeutic strategy directed to promote axonal regeneration of RGCs as a therapeutic approach for optic neuropathies.
Collapse
|
17
|
Möllmert S, Kharlamova MA, Hoche T, Taubenberger AV, Abuhattum S, Kuscha V, Kurth T, Brand M, Guck J. Zebrafish Spinal Cord Repair Is Accompanied by Transient Tissue Stiffening. Biophys J 2019; 118:448-463. [PMID: 31870536 DOI: 10.1016/j.bpj.2019.10.044] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 10/17/2019] [Accepted: 10/24/2019] [Indexed: 01/11/2023] Open
Abstract
Severe injury to the mammalian spinal cord results in permanent loss of function due to the formation of a glial-fibrotic scar. Both the chemical composition and the mechanical properties of the scar tissue have been implicated to inhibit neuronal regrowth and functional recovery. By contrast, adult zebrafish are able to repair spinal cord tissue and restore motor function after complete spinal cord transection owing to a complex cellular response that includes axon regrowth and is accompanied by neurogenesis. The mechanical mechanisms contributing to successful spinal cord repair in adult zebrafish are, however, currently unknown. Here, we employ atomic force microscopy-enabled nanoindentation to determine the spatial distributions of apparent elastic moduli of living spinal cord tissue sections obtained from uninjured zebrafish and at distinct time points after complete spinal cord transection. In uninjured specimens, spinal gray matter regions were stiffer than white matter regions. During regeneration after transection, the spinal cord tissues displayed a significant increase of the respective apparent elastic moduli that transiently obliterated the mechanical difference between the two types of matter before returning to baseline values after the completion of repair. Tissue stiffness correlated variably with cell number density, oligodendrocyte interconnectivity, axonal orientation, and vascularization. This work constitutes the first quantitative mapping of the spatiotemporal changes of spinal cord tissue stiffness in regenerating adult zebrafish and provides the tissue mechanical basis for future studies into the role of mechanosensing in spinal cord repair.
Collapse
Affiliation(s)
| | | | - Tobias Hoche
- Biotechnology Center, Technische Universität Dresden, Dresden, Germany
| | | | - Shada Abuhattum
- Biotechnology Center, Technische Universität Dresden, Dresden, Germany; JPK Instruments, Berlin, Germany; Max Planck Institut for the Science of Light & Max-Planck Institut für Physik und Medizin, Erlangen, Germany
| | - Veronika Kuscha
- Center for Regenerative Therapies, Technische Universität Dresden, Dresden, Germany
| | - Thomas Kurth
- Center for Regenerative Therapies, Technische Universität Dresden, Dresden, Germany
| | - Michael Brand
- Center for Regenerative Therapies, Technische Universität Dresden, Dresden, Germany
| | - Jochen Guck
- Biotechnology Center, Technische Universität Dresden, Dresden, Germany; Max Planck Institut for the Science of Light & Max-Planck Institut für Physik und Medizin, Erlangen, Germany.
| |
Collapse
|
18
|
Chen J, Shifman MI. Inhibition of neogenin promotes neuronal survival and improved behavior recovery after spinal cord injury. Neuroscience 2019; 408:430-447. [PMID: 30943435 DOI: 10.1016/j.neuroscience.2019.03.055] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 03/22/2019] [Accepted: 03/26/2019] [Indexed: 01/09/2023]
Abstract
Following spinal cord trauma, axonal regeneration in the mammalian spinal cord does not occur and functional recovery may be further impeded by retrograde neuronal death. By contrast, lampreys recover after spinal cord injury (SCI) and axons re-connected to their targets in spinal cord. However, the identified reticulospinal (RS) neurons located in the lamprey brain differ in their regenerative capacities - some are good regenerators, and others are bad regenerators - despite the fact that they have analogous projection pathways. Previously, we reported that axonal guidance receptor Neogenin involved in regulation of axonal regeneration after SCI and downregulation of Neogenin synthesis by morpholino oligonucleotides (MO) enhanced the regeneration of RS neurons. Incidentally, the bad regenerating RS neurons often undergo a late retrograde apoptosis after SCI. Here we report that, after SCI, expression of RGMa mRNA was upregulated around the transection site, while its receptor Neogenin continued to be synthesized almost inclusively in the "bad-regenerating" RS neurons. Inhibition of Neogenin by MO prohibited activation of caspases and improved the survival of RS neurons at 10 weeks after SCI. These data provide new evidence in vivo that Neogenin is involved in retrograde neuronal death and failure of axonal regeneration after SCI.
Collapse
Affiliation(s)
- Jie Chen
- Shriners Hospitals Pediatric Research Center (Center for Neural Repair and Rehabilitation), Philadelphia, PA 19140, USA
| | - Michael I Shifman
- Shriners Hospitals Pediatric Research Center (Center for Neural Repair and Rehabilitation), Philadelphia, PA 19140, USA; Department of Neuroscience, Temple University School of Medicine, 3500 North Broad Street, Philadelphia, PA 19140, USA.
| |
Collapse
|
19
|
Alizadeh A, Dyck SM, Karimi-Abdolrezaee S. Traumatic Spinal Cord Injury: An Overview of Pathophysiology, Models and Acute Injury Mechanisms. Front Neurol 2019; 10:282. [PMID: 30967837 PMCID: PMC6439316 DOI: 10.3389/fneur.2019.00282] [Citation(s) in RCA: 725] [Impact Index Per Article: 120.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 03/05/2019] [Indexed: 12/11/2022] Open
Abstract
Traumatic spinal cord injury (SCI) is a life changing neurological condition with substantial socioeconomic implications for patients and their care-givers. Recent advances in medical management of SCI has significantly improved diagnosis, stabilization, survival rate and well-being of SCI patients. However, there has been small progress on treatment options for improving the neurological outcomes of SCI patients. This incremental success mainly reflects the complexity of SCI pathophysiology and the diverse biochemical and physiological changes that occur in the injured spinal cord. Therefore, in the past few decades, considerable efforts have been made by SCI researchers to elucidate the pathophysiology of SCI and unravel the underlying cellular and molecular mechanisms of tissue degeneration and repair in the injured spinal cord. To this end, a number of preclinical animal and injury models have been developed to more closely recapitulate the primary and secondary injury processes of SCI. In this review, we will provide a comprehensive overview of the recent advances in our understanding of the pathophysiology of SCI. We will also discuss the neurological outcomes of human SCI and the available experimental model systems that have been employed to identify SCI mechanisms and develop therapeutic strategies for this condition.
Collapse
Affiliation(s)
- Arsalan Alizadeh
- Regenerative Medicine Program, Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, Spinal Cord Research Center, University of Manitoba, Winnipeg, MB, Canada
| | - Scott Matthew Dyck
- Regenerative Medicine Program, Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, Spinal Cord Research Center, University of Manitoba, Winnipeg, MB, Canada
| | - Soheila Karimi-Abdolrezaee
- Regenerative Medicine Program, Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, Spinal Cord Research Center, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
20
|
Altinova H, Hammes S, Palm M, Gerardo-Nava J, Achenbach P, Deumens R, Hermans E, Führmann T, Boecker A, van Neerven SGA, Bozkurt A, Weis J, Brook GA. Fibroadhesive scarring of grafted collagen scaffolds interferes with implant-host neural tissue integration and bridging in experimental spinal cord injury. Regen Biomater 2019; 6:75-87. [PMID: 30967962 PMCID: PMC6447003 DOI: 10.1093/rb/rbz006] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 01/06/2019] [Indexed: 02/06/2023] Open
Abstract
Severe traumatic spinal cord injury (SCI) results in a devastating and permanent loss of function, and is currently an incurable condition. It is generally accepted that future intervention strategies will require combinational approaches, including bioengineered scaffolds, to support axon growth across tissue scarring and cystic cavitation. Previously, we demonstrated that implantation of a microporous type-I collagen scaffold into an experimental model of SCI was capable of supporting functional recovery in the absence of extensive implant–host neural tissue integration. Here, we demonstrate the reactive host cellular responses that may be detrimental to neural tissue integration after implantation of collagen scaffolds into unilateral resection injuries of the adult rat spinal cord. Immunohistochemistry demonstrated scattered fibroblast-like cell infiltration throughout the scaffolds as well as the presence of variable layers of densely packed cells, the fine processes of which extended along the graft–host interface. Few reactive astroglial or regenerating axonal profiles could be seen traversing this layer. Such encapsulation-type behaviour around bioengineered scaffolds impedes the integration of host neural tissues and reduces the intended bridging role of the implant. Characterization of the cellular and molecular mechanisms underpinning this behaviour will be pivotal in the future design of collagen-based bridging scaffolds intended for regenerative medicine.
Collapse
Affiliation(s)
- Haktan Altinova
- Department of Neurosurgery, RWTH Aachen University Hospital, Aachen, Germany.,Institute of Neuropathology, RWTH Aachen University Hospital, Aachen, Germany.,Police Headquarters Berlin, Medical Commission, Berlin, Germany
| | - Sebastian Hammes
- Institute of Neuropathology, RWTH Aachen University Hospital, Aachen, Germany
| | - Moniek Palm
- Institute of Neuropathology, RWTH Aachen University Hospital, Aachen, Germany
| | - Jose Gerardo-Nava
- Institute of Neuropathology, RWTH Aachen University Hospital, Aachen, Germany
| | - Pascal Achenbach
- Institute of Neuropathology, RWTH Aachen University Hospital, Aachen, Germany
| | - Ronald Deumens
- Institute of Neuropathology, RWTH Aachen University Hospital, Aachen, Germany.,Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
| | - Emmanuel Hermans
- Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
| | - Tobias Führmann
- Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Canada
| | - Arne Boecker
- Department of Hand-, Plastic and Reconstructive Surgery, Burn Centre Trauma Centre, BG Trauma Centre Ludwigshafen, University of Heidelberg, Ludwigshafen, Germany.,Department of Plastic, Reconstructive and Hand Surgery, Burn Centre, RWTH Aachen University Hospital, Aachen, Germany
| | - Sabien Geraldine Antonia van Neerven
- Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium.,Department of Plastic, Reconstructive and Hand Surgery, Burn Centre, RWTH Aachen University Hospital, Aachen, Germany
| | - Ahmet Bozkurt
- Department of Plastic, Reconstructive and Hand Surgery, Burn Centre, RWTH Aachen University Hospital, Aachen, Germany.,Department of Plastic, Aesthetic, Hand and Burn Surgery, Helios University Hospital Wuppertal, University Witten/Herdecke, Wuppertal, Germany
| | - Joachim Weis
- Institute of Neuropathology, RWTH Aachen University Hospital, Aachen, Germany
| | - Gary Anthony Brook
- Institute of Neuropathology, RWTH Aachen University Hospital, Aachen, Germany
| |
Collapse
|
21
|
Körner S, Thau-Habermann N, Kefalakes E, Bursch F, Petri S. Expression of the axon-guidance protein receptor Neuropilin 1 is increased in the spinal cord and decreased in muscle of a mouse model of amyotrophic lateral sclerosis. Eur J Neurosci 2019; 49:1529-1543. [PMID: 30589468 DOI: 10.1111/ejn.14326] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 12/05/2018] [Accepted: 12/13/2018] [Indexed: 12/11/2022]
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a degenerative motor neuron disorder. It is supposed that ALS is at least in part an axonopathy. Neuropilin 1 is an important receptor of the axon repellent Semaphorin 3A and a co-receptor of vascular endothelial growth factor. It is probably involved in neuronal and axonal de-/regeneration and might be of high relevance for ALS pathogenesis and/or disease progression. To elucidate whether the expression of either Neuropilin1 or Semaphorin3A is altered in ALS we investigated these proteins in human brain, spinal cord and muscle tissue of ALS-patients and controls as well as transgenic SOD1G93A and control mice. Neuropilin1 and Semaphorin3A gene and protein expression were assessed by quantitative real-time PCR (qRT-PCR), western blot and immunohistochemistry. Groups were compared using either Student t-test or Mann-Whitney U test. We observed a consistent increase of Neuropilin1 expression in the spinal cord and decrease of Neuropilin1 and Semaphorin3A in muscle tissue of transgenic SOD1G93A mice at the mRNA and protein level. Previous studies have shown that damage of neurons physiologically causes Neuropilin1 and Semaphorin3A increase in the central nervous system and decrease in the peripheral nervous system. Our results indicate that this also occurs in ALS. Pharmacological modulation of expression and function of axon repellents could be a promising future therapeutic option in ALS.
Collapse
Affiliation(s)
- Sonja Körner
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | | | - Ekaterini Kefalakes
- Department of Neurology, Hannover Medical School, Hannover, Germany.,Center for Systems Neuroscience (ZSN), Hannover, Germany
| | - Franziska Bursch
- Department of Neurology, Hannover Medical School, Hannover, Germany.,Center for Systems Neuroscience (ZSN), Hannover, Germany
| | - Susanne Petri
- Department of Neurology, Hannover Medical School, Hannover, Germany.,Center for Systems Neuroscience (ZSN), Hannover, Germany
| |
Collapse
|
22
|
Tran AP, Warren PM, Silver J. The Biology of Regeneration Failure and Success After Spinal Cord Injury. Physiol Rev 2018. [PMID: 29513146 DOI: 10.1152/physrev.00017.2017] [Citation(s) in RCA: 553] [Impact Index Per Article: 79.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Since no approved therapies to restore mobility and sensation following spinal cord injury (SCI) currently exist, a better understanding of the cellular and molecular mechanisms following SCI that compromise regeneration or neuroplasticity is needed to develop new strategies to promote axonal regrowth and restore function. Physical trauma to the spinal cord results in vascular disruption that, in turn, causes blood-spinal cord barrier rupture leading to hemorrhage and ischemia, followed by rampant local cell death. As subsequent edema and inflammation occur, neuronal and glial necrosis and apoptosis spread well beyond the initial site of impact, ultimately resolving into a cavity surrounded by glial/fibrotic scarring. The glial scar, which stabilizes the spread of secondary injury, also acts as a chronic, physical, and chemo-entrapping barrier that prevents axonal regeneration. Understanding the formative events in glial scarring helps guide strategies towards the development of potential therapies to enhance axon regeneration and functional recovery at both acute and chronic stages following SCI. This review will also discuss the perineuronal net and how chondroitin sulfate proteoglycans (CSPGs) deposited in both the glial scar and net impede axonal outgrowth at the level of the growth cone. We will end the review with a summary of current CSPG-targeting strategies that help to foster axonal regeneration, neuroplasticity/sprouting, and functional recovery following SCI.
Collapse
Affiliation(s)
- Amanda Phuong Tran
- Department of Neurosciences, Case Western Reserve University , Cleveland, Ohio ; and School of Biomedical Sciences, University of Leeds , Leeds , United Kingdom
| | - Philippa Mary Warren
- Department of Neurosciences, Case Western Reserve University , Cleveland, Ohio ; and School of Biomedical Sciences, University of Leeds , Leeds , United Kingdom
| | - Jerry Silver
- Department of Neurosciences, Case Western Reserve University , Cleveland, Ohio ; and School of Biomedical Sciences, University of Leeds , Leeds , United Kingdom
| |
Collapse
|
23
|
Repulsive Environment Attenuation during Adult Mouse Optic Nerve Regeneration. Neural Plast 2018; 2018:5851914. [PMID: 30275822 PMCID: PMC6157103 DOI: 10.1155/2018/5851914] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 07/03/2018] [Accepted: 08/02/2018] [Indexed: 01/05/2023] Open
Abstract
The regenerative capacity of CNS tracts has ever been a great hurdle to regenerative medicine. Although recent studies have described strategies to stimulate retinal ganglion cells (RGCs) to regenerate axons through the optic nerve, it still remains to be elucidated how these therapies modulate the inhibitory environment of CNS. Thus, the present work investigated the environmental content of the repulsive axon guidance cues, such as Sema3D and its receptors, myelin debris, and astrogliosis, within the regenerating optic nerve of mice submitted to intraocular inflammation + cAMP combined to conditional deletion of PTEN in RGC after optic nerve crush. We show here that treatment was able to promote axonal regeneration through the optic nerve and reach visual targets at twelve weeks after injury. The Regenerating group presented reduced MBP levels, increased microglia/macrophage number, and reduced astrocyte reactivity and CSPG content following optic nerve injury. In addition, Sema3D content and its receptors are reduced in the Regenerating group. Together, our results provide, for the first time, evidence that several regenerative repulsive signals are reduced in regenerating optic nerve fibers following a combined therapy. Therefore, the treatment used made the CNS microenvironment more permissive to regeneration.
Collapse
|
24
|
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
|
25
|
Ruschel J, Bradke F. Systemic administration of epothilone D improves functional recovery of walking after rat spinal cord contusion injury. Exp Neurol 2018; 306:243-249. [PMID: 29223322 DOI: 10.1016/j.expneurol.2017.12.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 10/28/2017] [Accepted: 12/04/2017] [Indexed: 01/31/2023]
Abstract
Central nervous system (CNS) injuries cause permanent impairments of sensorimotor functions as mature neurons fail to regenerate their severed axons. The poor intrinsic growth capacity of adult CNS neurons and the formation of an inhibitory lesion scar are key impediments to axon regeneration. Systemic administration of the microtubule stabilizing agent epothilone B promotes axon regeneration and recovery of motor function by activating the intrinsic axonal growth machinery and by reducing the inhibitory fibrotic lesion scar. Thus, epothilones hold clinical promise as potential therapeutics for spinal cord injury. Here we tested the efficacy of epothilone D, an epothilone B analog with a superior safety profile. By using liquid chromatography and mass spectrometry (LC/MS), we found adequate CNS penetration and distribution of epothilone D after systemic administration, confirming the suitability of the drug for non-invasive CNS treatment. Systemic administration of epothilone D reduced inhibitory fibrotic scarring, promoted regrowth of injured raphespinal fibers and improved walking function after mid-thoracic spinal cord contusion injury in adult rats. These results confirm that systemic administration of epothilones is a valuable therapeutic strategy for CNS regeneration and repair after injury and provides a further advance for potential clinical translation.
Collapse
Affiliation(s)
- Jörg Ruschel
- German Center for Neurodegenerative Diseases, Sigmund-Freud-Strasse 27, 53127 Bonn, Germany.
| | - Frank Bradke
- German Center for Neurodegenerative Diseases, Sigmund-Freud-Strasse 27, 53127 Bonn, Germany.
| |
Collapse
|
26
|
Eliasen AM, Chin MR, Axelrod AJ, Abagyan R, Siegel D. Cascade reactions leading to the mechanism of action of vinaxanthone and xanthofulvin, natural products that drive nerve repair. Tetrahedron 2018. [DOI: 10.1016/j.tet.2018.02.068] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
27
|
The Extracellular Environment of the CNS: Influence on Plasticity, Sprouting, and Axonal Regeneration after Spinal Cord Injury. Neural Plast 2018; 2018:2952386. [PMID: 29849554 PMCID: PMC5932463 DOI: 10.1155/2018/2952386] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 01/22/2018] [Accepted: 02/06/2018] [Indexed: 11/17/2022] Open
Abstract
The extracellular environment of the central nervous system (CNS) becomes highly structured and organized as the nervous system matures. The extracellular space of the CNS along with its subdomains plays a crucial role in the function and stability of the CNS. In this review, we have focused on two components of the neuronal extracellular environment, which are important in regulating CNS plasticity including the extracellular matrix (ECM) and myelin. The ECM consists of chondroitin sulfate proteoglycans (CSPGs) and tenascins, which are organized into unique structures called perineuronal nets (PNNs). PNNs associate with the neuronal cell body and proximal dendrites of predominantly parvalbumin-positive interneurons, forming a robust lattice-like structure. These developmentally regulated structures are maintained in the adult CNS and enhance synaptic stability. After injury, however, CSPGs and tenascins contribute to the structure of the inhibitory glial scar, which actively prevents axonal regeneration. Myelin sheaths and mature adult oligodendrocytes, despite their important role in signal conduction in mature CNS axons, contribute to the inhibitory environment existing after injury. As such, unlike the peripheral nervous system, the CNS is unable to revert to a “developmental state” to aid neuronal repair. Modulation of these external factors, however, has been shown to promote growth, regeneration, and functional plasticity after injury. This review will highlight some of the factors that contribute to or prevent plasticity, sprouting, and axonal regeneration after spinal cord injury.
Collapse
|
28
|
Dias DO, Göritz C. Fibrotic scarring following lesions to the central nervous system. Matrix Biol 2018; 68-69:561-570. [PMID: 29428230 DOI: 10.1016/j.matbio.2018.02.009] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 02/05/2018] [Accepted: 02/05/2018] [Indexed: 10/18/2022]
Abstract
Following lesions to the central nervous system, scar tissue forms at the lesion site. Injury often severs axons and scar tissue is thought to block axonal regeneration, resulting in permanent functional deficits. While scar-forming astrocytes have been extensively studied, much less attention has been given to the fibrotic, non-glial component of the scar. We here review recent progress in understanding fibrotic scar formation following different lesions to the brain and spinal cord. We specifically highlight recent evidence for pericyte-derived fibrotic scar tissue formation, discussing the origin, recruitment, function and therapeutic relevance of fibrotic scarring.
Collapse
Affiliation(s)
- David Oliveira Dias
- Department of Cell and Molecular Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Christian Göritz
- Department of Cell and Molecular Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden.
| |
Collapse
|
29
|
Jha MK, Kim JH, Song GJ, Lee WH, Lee IK, Lee HW, An SSA, Kim S, Suk K. Functional dissection of astrocyte-secreted proteins: Implications in brain health and diseases. Prog Neurobiol 2017; 162:37-69. [PMID: 29247683 DOI: 10.1016/j.pneurobio.2017.12.003] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Revised: 10/23/2017] [Accepted: 12/08/2017] [Indexed: 02/07/2023]
Abstract
Astrocytes, which are homeostatic cells of the central nervous system (CNS), display remarkable heterogeneity in their morphology and function. Besides their physical and metabolic support to neurons, astrocytes modulate the blood-brain barrier, regulate CNS synaptogenesis, guide axon pathfinding, maintain brain homeostasis, affect neuronal development and plasticity, and contribute to diverse neuropathologies via secreted proteins. The identification of astrocytic proteome and secretome profiles has provided new insights into the maintenance of neuronal health and survival, the pathogenesis of brain injury, and neurodegeneration. Recent advances in proteomics research have provided an excellent catalog of astrocyte-secreted proteins. This review categorizes astrocyte-secreted proteins and discusses evidence that astrocytes play a crucial role in neuronal activity and brain function. An in-depth understanding of astrocyte-secreted proteins and their pathways is pivotal for the development of novel strategies for restoring brain homeostasis, limiting brain injury/inflammation, counteracting neurodegeneration, and obtaining functional recovery.
Collapse
Affiliation(s)
- Mithilesh Kumar Jha
- Department of Pharmacology, Brain Science and Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University School of Medicine, Daegu, Republic of Korea; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jong-Heon Kim
- Department of Pharmacology, Brain Science and Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Gyun Jee Song
- Department of Pharmacology, Brain Science and Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Won-Ha Lee
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
| | - In-Kyu Lee
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Ho-Won Lee
- Department of Neurology, Brain Science and Engineering Institute, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Seong Soo A An
- Department of BioNano Technology, Gachon University, Gyeonggi-do, Republic of Korea
| | - SangYun Kim
- Department of Neurology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Gyeonggi-do, Republic of Korea
| | - Kyoungho Suk
- Department of Pharmacology, Brain Science and Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University School of Medicine, Daegu, Republic of Korea.
| |
Collapse
|
30
|
The Semaphorin 3A inhibitor SM-345431 preserves corneal nerve and epithelial integrity in a murine dry eye model. Sci Rep 2017; 7:15584. [PMID: 29138447 PMCID: PMC5686158 DOI: 10.1038/s41598-017-15682-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 10/31/2017] [Indexed: 11/17/2022] Open
Abstract
Dry eye disease (DED) is a common disorder causing discomfort and ocular fatigue. Corneal nerves are compromised in DED, which may further cause loss of corneal sensation and decreased tear secretion. Semaphorin 3A (Sema3A) is expressed by the corneal epithelium under stress, and is known as an inhibitor of axonal regeneration. Using a murine dry eye model, we found that topical SM-345431, a selective Sema3A inhibitor, preserved corneal sensitivity (2.3 ± 0.3 mm versus 1.4 ± 0.1 mm in vehicle control, p = 0.004) and tear volume (1.1 ± 0.1 mm versus 0.3 ± 0.1 mm in vehicle control, p < 0.001). Fluorescein staining area of the cornea due to damage to barrier function was also reduced (4.1 ± 0.9% in SM-345431 group versus 12.9 ± 2.2% in vehicle control, p < 0.001). The incidence of corneal epithelial erosions was significantly suppressed by SM-345431 (none in SM-345431 group versus six (21%) in vehicle control, p = 0.01). Furthermore, sub-epithelial corneal nerve density and intraepithelial expression of transient receptor potential vanilloid receptor 1 (TRPV1) were significantly preserved with SM-345431. Our results suggest that inhibition of Sema3A may be an effective therapy for DED.
Collapse
|
31
|
Delayed Heterochronic Transplantation following Focal Cortical Lesion Improves Outcome. J Neurosci 2017; 37:6391-6393. [PMID: 28679798 DOI: 10.1523/jneurosci.1021-17.2017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2017] [Revised: 06/04/2017] [Accepted: 06/07/2017] [Indexed: 11/21/2022] Open
|
32
|
Blits B, Boer GJ, Verhaagen J. Pharmacological, Cell, and Gene Therapy Strategies to Promote Spinal Cord Regeneration. Cell Transplant 2017. [DOI: 10.3727/000000002783985521] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
In this review, recent studies using pharmacological treatment, cell transplantation, and gene therapy to promote regeneration of the injured spinal cord in animal models will be summarized. Pharmacological and cell transplantation treatments generally revealed some degree of effect on the regeneration of the injured ascending and descending tracts, but further improvements to achieve a more significant functional recovery are necessary. The use of gene therapy to promote repair of the injured nervous system is a relatively new concept. It is based on the development of methods for delivering therapeutic genes to neurons, glia cells, or nonneural cells. Direct in vivo gene transfer or gene transfer in combination with (neuro)transplantation (ex vivo gene transfer) appeared powerful strategies to promote neuronal survival and axonal regrowth following traumatic injury to the central nervous system. Recent advances in understanding the cellular and molecular mechanisms that govern neuronal survival and neurite outgrowth have enabled the design of experiments aimed at viral vector-mediated transfer of genes encoding neurotrophic factors, growth-associated proteins, cell adhesion molecules, and antiapoptotic genes. Central to the success of these approaches was the development of efficient, nontoxic vectors for gene delivery and the acquirement of the appropriate (genetically modified) cells for neurotransplantation. Direct gene transfer in the nervous system was first achieved with herpes viral and E1-deleted adenoviral vectors. Both vector systems are problematic in that these vectors elicit immunogenic and cytotoxic responses. Adeno-associated viral vectors and lentiviral vectors constitute improved gene delivery systems and are beginning to be applied in neuroregeneration research of the spinal cord. Ex vivo approaches were initially based on the implantation of genetically modified fibroblasts. More recently, transduced Schwann cells, genetically modified pieces of peripheral nerve, and olfactory ensheathing glia have been used as implants into the injured spinal cord.
Collapse
Affiliation(s)
- Bas Blits
- Graduate School Neurosciences Amsterdam, Netherlands Institute for Brain Research, Meibergdreef 33, 1105 AZ Amsterdam-ZO, The Netherlands
| | - Gerard J. Boer
- Graduate School Neurosciences Amsterdam, Netherlands Institute for Brain Research, Meibergdreef 33, 1105 AZ Amsterdam-ZO, The Netherlands
| | - Joost Verhaagen
- Graduate School Neurosciences Amsterdam, Netherlands Institute for Brain Research, Meibergdreef 33, 1105 AZ Amsterdam-ZO, The Netherlands
| |
Collapse
|
33
|
Li Y, Yao D, Zhang J, Liu B, Zhang L, Feng H, Li B. The Effects of Epidermal Neural Crest Stem Cells on Local Inflammation Microenvironment in the Defected Sciatic Nerve of Rats. Front Mol Neurosci 2017; 10:133. [PMID: 28588447 PMCID: PMC5438963 DOI: 10.3389/fnmol.2017.00133] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 04/20/2017] [Indexed: 12/21/2022] Open
Abstract
Cell-based therapy is a promising strategy for the repair of peripheral nerve injuries (PNIs). epidermal neural crest stems cells (EPI-NCSCs) are thought to be important donor cells for repairing PNI in different animal models. Following PNI, inflammatory response is important to regulate the repair process. However, the effects of EPI-NCSCs on regulation of local inflammation microenviroment have not been investigated extensively. In the present study, these effects were studied by using 10 mm defected sciatic nerve, which was bridged with 15 mm artificial nerve composed of EPI-NCSCs, extracellular matrix (ECM) and poly (lactide-co-glycolide) (PLGA). Then the expression of pro- and anti-inflammatory cytokines, polarization of macrophages, regulation of fibroblasts and shwann cells (SCs) were assessed by western blot, immunohistochemistry, immunofluorescence staining at 1, 3, 7 and 21 days after bridging. The structure and the function of the bridged nerve were determined by observation under light microscope and by examination of right lateral foot retraction time (LFRT), sciatic function index (SFI), gastrocnemius wet weight and electrophysiology at 9 weeks. After bridging with EPI-NCSCs, the expression of anti-inflammatory cytokines (IL-4 and IL-13) was increased, but decreased for pro-inflammatory cytokines (IL-6 and TNF-α) compared to the control bridging, which was consistent with increase of M2 macrophages and decrease of M1 macrophages at 7 days after transplantation. Likewise, myelin-formed SCs were significantly increased, but decreased for the activated fibroblasts in their number at 21 days. The recovery of structure and function of nerve bridged with EPI-NCSCs was significantly superior to that of DMEM. These results indicated that EPI-NCSCs could be able to regulate and provide more suitable inflammation microenvironment for the repair of defected sciatic nerve.
Collapse
Affiliation(s)
- Yue Li
- Department of Neurosurgery, Southwest Hospital/State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical UniversityChongqing, China
| | - Dongdong Yao
- Research Institute of Surgery, Daping Hospital/State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical UniversityChongqing, China.,School of Life Sciences/Key Laboratory of Freshwater Fish Reproduction and Development of Education Ministry, Southwest UniversityChongqing, China
| | - Jieyuan Zhang
- Research Institute of Surgery, Daping Hospital/State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical UniversityChongqing, China
| | - Bin Liu
- School of Life Sciences/Key Laboratory of Freshwater Fish Reproduction and Development of Education Ministry, Southwest UniversityChongqing, China
| | - Lu Zhang
- Children's Hospital of Chongqing Medical University/Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Medical UniversityChongqing, China
| | - Hua Feng
- Department of Neurosurgery, Southwest Hospital/State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical UniversityChongqing, China
| | - Bingcang Li
- Research Institute of Surgery, Daping Hospital/State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical UniversityChongqing, China
| |
Collapse
|
34
|
Nagoshi N, Okano H. Applications of induced pluripotent stem cell technologies in spinal cord injury. J Neurochem 2017; 141:848-860. [PMID: 28199003 DOI: 10.1111/jnc.13986] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Revised: 12/30/2016] [Accepted: 01/03/2017] [Indexed: 12/14/2022]
Abstract
Numerous basic research studies have suggested the potential efficacy of neural precursor cell (NPC) transplantation in spinal cord injury (SCI). However, in most such studies, the origin of the cells used was mainly fetal tissue or embryonic stem cells, both of which carry potential ethical concerns with respect to clinical use. The development of induced pluripotent stem cells (iPSCs) opened a new path toward regenerative medicine for SCI. iPSCs can be generated from somatic cells by induction of transcription factors, and induced to differentiate into NPCs with characteristics of cells of the central nervous system. The beneficial effect of iPSC-derived NPC transplantation has been reported from our group and others working in rodent and non-human primate models. These promising results facilitate the application of iPSCs for clinical applications in SCI patients. However, iPSCs also have issues, such as genetic/epigenetic abnormalities and tumorigenesis because of the artificial induction method, that must be addressed prior to clinical use. The selection of somatic cells, generation of integration-free iPSCs, and characterization of differentiated NPCs with thorough quality management are all needed to address these potential risks. To enhance the efficacy of the transplanted iPSC-NPCs, especially at chronic phase of SCI, administration of a chondroitinase or semaphorin3A inhibitor represents a potentially important means of promoting axonal regeneration through the lesion site. The combined use of rehabilitation with such cell therapy approaches is also important, as repetitive training enhances neurite outgrowth of transplanted cells and strengthens neural circuits at central pattern generators. Our group has already evaluated clinical grade iPSC-derived NPCs, and we look forward to initiating clinical testing as the next step toward determining whether this approach is safe and effective for clinical use. This article is part of the mini review series "60th Anniversary of the Japanese Society for Neurochemistry".
Collapse
Affiliation(s)
- Narihito Nagoshi
- Department of Orthopaedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
35
|
Parker K, Berretta A, Saenger S, Sivaramakrishnan M, Shirley SA, Metzger F, Clarkson AN. PEGylated insulin-like growth factor-I affords protection and facilitates recovery of lost functions post-focal ischemia. Sci Rep 2017; 7:241. [PMID: 28325900 PMCID: PMC5428211 DOI: 10.1038/s41598-017-00336-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 02/21/2017] [Indexed: 12/04/2022] Open
Abstract
Insulin-like growth factor-I (IGF-I) is involved in the maturation and maintenance of neurons, and impaired IGF-I signaling has been shown to play a role in various neurological diseases including stroke. The aim of the present study was to investigate the efficacy of an optimized IGF-I variant by adding a 40 kDa polyethylene glycol (PEG) chain to IGF-I to form PEG-IGF-I. We show that PEG-IGF-I has a slower clearance which allows for twice-weekly dosing to maintain steady-state serum levels in mice. Using a photothrombotic model of focal stroke, dosing from 3 hrs post-stroke dose-dependently (0.3–1 mg/kg) decreases the volume of infarction and improves motor behavioural function in both young 3-month and aged 22–24 month old mice. Further, PEG-IGF-I treatment increases GFAP expression when given early (3 hrs post-stroke), increases Synaptophysin expression and increases neurogenesis in young and aged. Finally, neurons (P5–6) cultured in vitro on reactive astrocytes in the presence of PEG-IGF-I showed an increase in neurite length, indicating that PEG-IGF-I can aid in sprouting of new connections. This data suggests a modulatory role of IGF-I in both protective and regenerative processes, and indicates that therapeutic approaches using PEG-IGF-I should be given early and where the endogenous regenerative potential is still high.
Collapse
Affiliation(s)
- Kim Parker
- Department of Anatomy and Brain Health Research Center, University of Otago, Dunedin 9054, New Zealand
| | - Antonio Berretta
- Department of Anatomy and Brain Health Research Center, University of Otago, Dunedin 9054, New Zealand
| | - Stefanie Saenger
- F. Hoffmann-La Roche Ltd., pRED, Pharma Research & Early Development, Roche Innovation Center Basel, Grenzacherstrasse 124, CH-4070, Basel, Switzerland
| | - Manaswini Sivaramakrishnan
- F. Hoffmann-La Roche Ltd., pRED, Pharma Research & Early Development, Roche Innovation Center Basel, Grenzacherstrasse 124, CH-4070, Basel, Switzerland
| | - Simon A Shirley
- Department of Anatomy and Brain Health Research Center, University of Otago, Dunedin 9054, New Zealand
| | - Friedrich Metzger
- F. Hoffmann-La Roche Ltd., pRED, Pharma Research & Early Development, Roche Innovation Center Basel, Grenzacherstrasse 124, CH-4070, Basel, Switzerland
| | - Andrew N Clarkson
- Department of Anatomy and Brain Health Research Center, University of Otago, Dunedin 9054, New Zealand. .,Brain Research New Zealand, University of Otago, Dunedin 9054, New Zealand. .,Faculty of Pharmacy, The University of Sydney, Sydney, Australia.
| |
Collapse
|
36
|
Chen J, Laramore C, Shifman MI. The expression of chemorepulsive guidance receptors and the regenerative abilities of spinal-projecting neurons after spinal cord injury. Neuroscience 2016; 341:95-111. [PMID: 27890825 DOI: 10.1016/j.neuroscience.2016.11.020] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 11/14/2016] [Accepted: 11/16/2016] [Indexed: 12/31/2022]
Abstract
Spinal cord injury (SCI) in mammals leads to permanent loss of function because axons do not regenerate in the central nervous system (CNS). To date, treatments based on neutralizing inhibitory environmental cues, such as the myelin-associated growth inhibitors and chondroitin sulfate proteoglycans, or on adding neurotrophic factors, have had limited success in enhancing regeneration. Published studies suggested that multiple axon guidance cues (repulsive guidance molecule (RGM) family, semaphorins, ephrins, and netrins) persist in adult animals, and that their expression is upregulated after CNS injury. Moreover, many adult CNS neurons continue to express axon guidance receptors. We used the advantages of the lamprey CNS to test the hypotheses that the regenerative abilities of spinal-projecting neurons depend upon their expression of chemorepulsive guidance receptors. After complete spinal transection, lampreys recover behaviorally, and injured axons grow selectively in their correct paths. However, the large identified reticulospinal (RS) neurons in the lamprey brain are heterogeneous in their regenerative abilities - some are high regeneration capacity neurons (probability of axon regeneration >50%), others are low regeneration capacity neurons (<30%). Here we report that the RGM receptor Neogenin is expressed preferentially in the low regeneration capacity RS neurons that regenerate poorly, and that downregulation of Neogenin by morpholino antisense oligonucleotides enhances regeneration of RS axons after SCI. Moreover, lamprey CNS neurons co-express multiple guidance receptors (Neogenin, UNC5 and PlexinA), suggesting that the regenerative abilities of spinal-projecting neurons might reflect the summed influences of the chemorepulsive guidance receptors that they express.
Collapse
Affiliation(s)
- Jie Chen
- Shriners Hospitals Pediatric Research Center (Center for Neural Repair and Rehabilitation), USA
| | - Cindy Laramore
- Shriners Hospitals Pediatric Research Center (Center for Neural Repair and Rehabilitation), USA
| | - Michael I Shifman
- Shriners Hospitals Pediatric Research Center (Center for Neural Repair and Rehabilitation), USA; Department of Neuroscience, Temple University School of Medicine, 3500 North Broad Street, Philadelphia, PA 19140, USA.
| |
Collapse
|
37
|
Altinova H, Möllers S, Deumens R, Gerardo-Nava J, Führmann T, van Neerven SGA, Bozkurt A, Mueller CA, Hoff HJ, Heschel I, Weis J, Brook GA. Functional recovery not correlated with axon regeneration through olfactory ensheathing cell-seeded scaffolds in a model of acute spinal cord injury. Tissue Eng Regen Med 2016; 13:585-600. [PMID: 30603440 DOI: 10.1007/s13770-016-9115-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 01/03/2016] [Accepted: 02/18/2016] [Indexed: 12/18/2022] Open
Abstract
The implantation of bioengineered scaffolds into lesion-induced gaps of the spinal cord is a promising strategy for promoting functional tissue repair because it can be combined with other intervention strategies. Our previous investigations showed that functional improvement following the implantation of a longitudinally microstructured collagen scaffold into unilateral mid-cervical spinal cord resection injuries of adult Lewis rats was associated with only poor axon regeneration within the scaffold. In an attempt to improve graft-host integration as well as functional recovery, scaffolds were seeded with highly enriched populations of syngeneic, olfactory bulb-derived ensheathing cells (OECs) prior to implantation into the same lesion model. Regenerating neurofilament-positive axons closely followed the trajectory of the donor OECs, as well as that of the migrating host cells within the scaffold. However, there was only a trend for increased numbers of regenerating axons above that supported by non-seeded scaffolds or in the untreated lesions. Nonetheless, significant functional recovery in skilled forelimb motor function was observed following the implantation of both seeded and non-seeded scaffolds which could not be correlated to the extent of axon regeneration within the scaffold. Mechanisms other than simple bridging of axon regeneration across the lesion must be responsible for the improved motor function.
Collapse
Affiliation(s)
- Haktan Altinova
- Department of Neurosurgery, Evangelic Hospital Bethel, Bielefeld, Germany.,2Institute of Neuropathology, Uniklinik RWTH Aachen University, Aachen, Germany.,Jülich-Aachen Research Alliance-Translational Brain Medicine (JARA Brain), Jülich, Germany.,4Department of Neurosurgery, Uniklinik RWTH Aachen University, Aachen, Germany
| | - Sven Möllers
- 5Charité Stem Cell Facility, Charité University Hospital, Berlin, Germany
| | - Ronald Deumens
- 2Institute of Neuropathology, Uniklinik RWTH Aachen University, Aachen, Germany.,Jülich-Aachen Research Alliance-Translational Brain Medicine (JARA Brain), Jülich, Germany.,6Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
| | - Jose Gerardo-Nava
- 2Institute of Neuropathology, Uniklinik RWTH Aachen University, Aachen, Germany.,Jülich-Aachen Research Alliance-Translational Brain Medicine (JARA Brain), Jülich, Germany
| | - Tobias Führmann
- 7Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Ontario, Canada
| | | | - Ahmet Bozkurt
- 8Department of Plastic, Reconstructive and Hand Surgery, Burn Centre, Uniklinik RWTH Aachen University, Aachen, Germany.,9Department of Plastic and Aesthetic, Reconstructive and Hand Surgery, Center for Reconstructive Microsurgery and Peripheral Nerve Surgery (ZEMPEN), Agaplesion Markus Hospital Frankfurt, Academic Hospital of Johann Wolfgang von Goethe University, Frankfurt, Germany
| | | | - Hans Joachim Hoff
- Department of Neurosurgery, Evangelic Hospital Bethel, Bielefeld, Germany
| | | | - Joachim Weis
- 2Institute of Neuropathology, Uniklinik RWTH Aachen University, Aachen, Germany.,Jülich-Aachen Research Alliance-Translational Brain Medicine (JARA Brain), Jülich, Germany
| | - Gary Anthony Brook
- 2Institute of Neuropathology, Uniklinik RWTH Aachen University, Aachen, Germany.,Jülich-Aachen Research Alliance-Translational Brain Medicine (JARA Brain), Jülich, Germany
| |
Collapse
|
38
|
Reduced Sympathetic Innervation in Endometriosis is Associated to Semaphorin 3C and 3F Expression. Mol Neurobiol 2016; 54:5131-5141. [PMID: 27558236 DOI: 10.1007/s12035-016-0058-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 08/15/2016] [Indexed: 12/16/2022]
Abstract
Endometriosis is a chronic inflammatory disease and one of the most common causes of pelvic pain. The mechanisms underlying pain emergence or chronic inflammation during endometriosis remain unknown. Several chronic inflammatory diseases including endometriosis show reduced amounts of noradrenergic nerve fibers. The source of the affected innervation is still unclear. Semaphorins represent potential elicitors, due to their known role as axonal guidance cues, and are suggested as nerve repellent factors in different chronic inflammatory diseases. Therefore, semaphorins might influence the progress of neuroinflammatory mechanisms during endometriosis. Here, we analyzed the noradrenergic innervation and the expression of the specific semaphorins and receptors possibly involved in the neuroimmunomodulation in endometriosis. Our studies revealed an affected innervation and a significant increase of semaphorins and their receptors in peritoneal endometriotic tissue. Thereby, the expression of the receptors was identified on the membrane of noradrenergic nerve fibers and vessels. Macrophages and activated fibroblasts were found in higher density levels and additionally express semaphorins in peritoneal endometriotic tissue. Inflammation leads to an increased release of immune cells, which secrete a variety of inflammatory factors capable of affecting innervation. Therefore, our data suggests that the chronic inflammatory condition in endometriosis might contribute to the increase of semaphorins, which could possibly affect the innervation in peritoneal endometriosis.
Collapse
|
39
|
Anderson JE, Do MKQ, Daneshvar N, Suzuki T, Dort J, Mizunoya W, Tatsumi R. The role of semaphorin3A in myogenic regeneration and the formation of functional neuromuscular junctions on new fibres. Biol Rev Camb Philos Soc 2016; 92:1389-1405. [PMID: 27296513 DOI: 10.1111/brv.12286] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 05/10/2016] [Accepted: 05/16/2016] [Indexed: 01/03/2023]
Abstract
Current research on skeletal muscle injury and regeneration highlights the crucial role of nerve-muscle interaction in the restoration of innervation during that process. Activities of muscle satellite or stem cells, recognized as the 'currency' of myogenic repair, have a pivotal role in these events, as shown by ongoing research. More recent investigation of myogenic signalling events reveals intriguing roles for semaphorin3A (Sema3A), secreted by activated satellite cells, in the muscle environment during development and regeneration. For example, Sema3A makes important contributions to regulating the formation of blood vessels, balancing bone formation and bone remodelling, and inflammation, and was recently implicated in the establishment of fibre-type distribution through effects on myosin heavy chain gene expression. This review highlights the active or potential contributions of satellite-cell-derived Sema3A to regulation of the processes of motor neurite ingrowth into a regenerating muscle bed. Successful restoration of functional innervation during muscle repair is essential; this review emphasizes the integrative role of satellite-cell biology in the progressive coordination of adaptive cellular and tissue responses during the injury-repair process in voluntary muscle.
Collapse
Affiliation(s)
- Judy E Anderson
- Department of Biological Sciences, Faculty of Science, University of Manitoba, Winnipeg, R3T 2N2, Canada
| | - Mai-Khoi Q Do
- Department of Animal and Marine Bioresource Sciences, Graduate School of Agriculture, Kyushu University, Higashi-ku Fukuoka, 8128581, Japan
| | - Nasibeh Daneshvar
- Department of Biological Sciences, Faculty of Science, University of Manitoba, Winnipeg, R3T 2N2, Canada
| | - Takahiro Suzuki
- Department of Animal and Marine Bioresource Sciences, Graduate School of Agriculture, Kyushu University, Higashi-ku Fukuoka, 8128581, Japan
| | - Junio Dort
- Department of Biological Sciences, Faculty of Science, University of Manitoba, Winnipeg, R3T 2N2, Canada
| | - Wataru Mizunoya
- Department of Animal and Marine Bioresource Sciences, Graduate School of Agriculture, Kyushu University, Higashi-ku Fukuoka, 8128581, Japan
| | - Ryuichi Tatsumi
- Department of Animal and Marine Bioresource Sciences, Graduate School of Agriculture, Kyushu University, Higashi-ku Fukuoka, 8128581, Japan
| |
Collapse
|
40
|
Quintá HR, Wilson C, Blidner AG, González-Billault C, Pasquini LA, Rabinovich GA, Pasquini JM. Ligand-mediated Galectin-1 endocytosis prevents intraneural H2O2 production promoting F-actin dynamics reactivation and axonal re-growth. Exp Neurol 2016; 283:165-78. [PMID: 27296316 DOI: 10.1016/j.expneurol.2016.06.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 05/08/2016] [Accepted: 06/09/2016] [Indexed: 12/25/2022]
Abstract
UNLABELLED Axonal growth cone collapse following spinal cord injury (SCI) is promoted by semaphorin3A (Sema3A) signaling via PlexinA4 surface receptor. This interaction triggers intracellular signaling events leading to increased hydrogen peroxide levels which in turn promote filamentous actin (F-actin) destabilization and subsequent inhibition of axonal re-growth. In the current study, we demonstrated that treatment with galectin-1 (Gal-1), in its dimeric form, promotes a decrease in hydrogen peroxide (H2O2) levels and F-actin repolimerization in the growth cone and in the filopodium of neuron surfaces. This effect was dependent on the carbohydrate recognition activity of Gal-1, as it was prevented using a Gal-1 mutant lacking carbohydrate-binding activity. Furthermore, Gal-1 promoted its own active ligand-mediated endocytosis together with the PlexinA4 receptor, through mechanisms involving complex branched N-glycans. In summary, our results suggest that Gal-1, mainly in its dimeric form, promotes re-activation of actin cytoskeleton dynamics via internalization of the PlexinA4/Gal-1 complex. This mechanism could explain, at least in part, critical events in axonal regeneration including the full axonal re-growth process, de novo formation of synapse clustering, axonal re-myelination and functional recovery of coordinated locomotor activities in an in vivo acute and chronic SCI model. SIGNIFICANCE STATEMENT Axonal regeneration is a response of injured nerve cells critical for nerve repair in human spinal cord injury. Understanding the molecular mechanisms controlling nerve repair by Galectin-1, may be critical for therapeutic intervention. Our results show that Galectin-1; in its dimeric form, interferes with hydrogen peroxide production triggered by Semaphorin3A. The high levels of this reactive oxygen species (ROS) seem to be the main factor preventing axonal regeneration due to promotion of actin depolymerization at the axonal growth cone. Thus, Galectin-1 administration emerges as a novel therapeutic modality for promoting nerve repair and preventing axonal loss.
Collapse
Affiliation(s)
- Héctor R Quintá
- Departamento de Química Biológica, Instituto de Química y Físico Química Biológica, Universidad de Buenos Aires, Buenos Aires C1113AAD, Argentina
| | - Carlos Wilson
- Laboratory of Cell and Neuronal Dymanics, Faculty of Sciences, Universidad de Chile. Center for Geroscience, Brain Health and Metabolism, Santiago, Chile. The Buck Institute for Research on Aging, Novato, USA
| | - Ada G Blidner
- Laboratorio de Inmunopatología, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET). Buenos Aires C1428, Argentina
| | - Christian González-Billault
- Laboratory of Cell and Neuronal Dymanics, Faculty of Sciences, Universidad de Chile. Center for Geroscience, Brain Health and Metabolism, Santiago, Chile. The Buck Institute for Research on Aging, Novato, USA
| | - Laura A Pasquini
- Departamento de Química Biológica, Instituto de Química y Físico Química Biológica, Universidad de Buenos Aires, Buenos Aires C1113AAD, Argentina
| | - Gabriel A Rabinovich
- Laboratorio de Inmunopatología, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET). Buenos Aires C1428, Argentina; Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, C1428, Argentina
| | - Juana M Pasquini
- Departamento de Química Biológica, Instituto de Química y Físico Química Biológica, Universidad de Buenos Aires, Buenos Aires C1113AAD, Argentina.
| |
Collapse
|
41
|
Abstract
…once the development was ended, the founts of growth and regeneration of the axons and dendrites dried up irrevocably. Santiago Ramón y Cajal Cajal's neurotropic theory postulates that the complexity of the nervous system arises from the collaboration of neurotropic signals from neuronal and non-neuronal cells and that once development has ended, a paucity of neurotropic signals means that the pathways of the central nervous system are "fixed, ended, immutable". While the capacity for regeneration and plasticity of the central nervous system may not be quite as paltry as Cajal proposed, regeneration is severely limited in scope as there is no spontaneous regeneration of long-distance projections in mammals and therefore limited opportunity for functional recovery following spinal cord injury. It is not a far stretch from Cajal to hypothesize that reappropriation of the neurotropic programs of development may be an appropriate strategy for reconstitution of injured circuits. It has become clear, however, that a significant number of the molecular cues governing circuit development become re-active after injury and many assume roles that paradoxically obstruct the functional re-wiring of severed neural connections. Therefore, the problem to address is how individual neural circuits respond to specific molecular cues following injury, and what strategies will be necessary for instigating functional repair or remodeling of the injured spinal cord.
Collapse
Affiliation(s)
- Edmund R Hollis
- Burke Medical Research Institute, White Plains, NY, USA.
- Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
42
|
Körner S, Böselt S, Wichmann K, Thau-Habermann N, Zapf A, Knippenberg S, Dengler R, Petri S. The Axon Guidance Protein Semaphorin 3A Is Increased in the Motor Cortex of Patients With Amyotrophic Lateral Sclerosis. J Neuropathol Exp Neurol 2016; 75:326-333. [PMID: 26921371 DOI: 10.1093/jnen/nlw003] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a degenerative motor neuron disorder that leads to progressive paralysis of skeletal muscles and death by respiratory failure. There is increasing evidence that ALS is at least in part an axonopathy and that mechanisms regulating axonal degeneration and regeneration might be pathogenetically relevant. Semaphorin 3A (Sema3A) is an axon guidance protein; it acts as an axon repellent and prevents axonal regeneration. Increased Sema3A expression has been described in a mouse model of ALS in which it may contribute to motor neuron degeneration. This study aimed to investigate Sema3A mRNA and protein expression in human CNS tissues. We assessed Sema3A expression using quantitative real-time PCR, in situ hybridization, and immunohistochemistry in motor cortex and spinal cord tissue of 8 ALS patients and 6 controls. We found a consistent increase of Sema3A expression in the motor cortex of ALS patients by all 3 methods. In situ hybridization further confirmed that Sema3A expression was present in motor neurons. These findings indicate that upregulation of Sema3A may contribute to axonal degeneration and failure of regeneration in ALS patients. The inhibition of Sema3A therefore might be a promising future therapeutic option for patients with this disease.
Collapse
Affiliation(s)
- Sonja Körner
- From the Department of Neurology, Hannover Medical School, Hannover, Germany (SK, SB, KW, NTH, RD); Department of Medical Statistics, University Medical Center, Göttingen, Germany (AZ); Department of Experimental Pneumology, Hannover Medical School, Hannover, Germany (SK); and Center for Systems Neuroscience (ZSN), Hannover, Germany (RD, SP).
| | - Sebastian Böselt
- From the Department of Neurology, Hannover Medical School, Hannover, Germany (SK, SB, KW, NTH, RD); Department of Medical Statistics, University Medical Center, Göttingen, Germany (AZ); Department of Experimental Pneumology, Hannover Medical School, Hannover, Germany (SK); and Center for Systems Neuroscience (ZSN), Hannover, Germany (RD, SP)
| | - Klaudia Wichmann
- From the Department of Neurology, Hannover Medical School, Hannover, Germany (SK, SB, KW, NTH, RD); Department of Medical Statistics, University Medical Center, Göttingen, Germany (AZ); Department of Experimental Pneumology, Hannover Medical School, Hannover, Germany (SK); and Center for Systems Neuroscience (ZSN), Hannover, Germany (RD, SP)
| | - Nadine Thau-Habermann
- From the Department of Neurology, Hannover Medical School, Hannover, Germany (SK, SB, KW, NTH, RD); Department of Medical Statistics, University Medical Center, Göttingen, Germany (AZ); Department of Experimental Pneumology, Hannover Medical School, Hannover, Germany (SK); and Center for Systems Neuroscience (ZSN), Hannover, Germany (RD, SP)
| | - Antonia Zapf
- From the Department of Neurology, Hannover Medical School, Hannover, Germany (SK, SB, KW, NTH, RD); Department of Medical Statistics, University Medical Center, Göttingen, Germany (AZ); Department of Experimental Pneumology, Hannover Medical School, Hannover, Germany (SK); and Center for Systems Neuroscience (ZSN), Hannover, Germany (RD, SP)
| | - Sarah Knippenberg
- From the Department of Neurology, Hannover Medical School, Hannover, Germany (SK, SB, KW, NTH, RD); Department of Medical Statistics, University Medical Center, Göttingen, Germany (AZ); Department of Experimental Pneumology, Hannover Medical School, Hannover, Germany (SK); and Center for Systems Neuroscience (ZSN), Hannover, Germany (RD, SP)
| | - Reinhard Dengler
- From the Department of Neurology, Hannover Medical School, Hannover, Germany (SK, SB, KW, NTH, RD); Department of Medical Statistics, University Medical Center, Göttingen, Germany (AZ); Department of Experimental Pneumology, Hannover Medical School, Hannover, Germany (SK); and Center for Systems Neuroscience (ZSN), Hannover, Germany (RD, SP)
| | - Susanne Petri
- From the Department of Neurology, Hannover Medical School, Hannover, Germany (SK, SB, KW, NTH, RD); Department of Medical Statistics, University Medical Center, Göttingen, Germany (AZ); Department of Experimental Pneumology, Hannover Medical School, Hannover, Germany (SK); and Center for Systems Neuroscience (ZSN), Hannover, Germany (RD, SP)
| |
Collapse
|
43
|
Berretta A, Gowing EK, Jasoni CL, Clarkson AN. Sonic hedgehog stimulates neurite outgrowth in a mechanical stretch model of reactive-astrogliosis. Sci Rep 2016; 6:21896. [PMID: 26902390 PMCID: PMC4763245 DOI: 10.1038/srep21896] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 02/01/2016] [Indexed: 01/15/2023] Open
Abstract
Although recovery following a stroke is limited, undamaged neurons under the right conditions can establish new connections and take on-board lost functions. Sonic hedgehog (Shh) signaling is integral for developmental axon growth, but its role after injury has not been fully examined. To investigate the effects of Shh on neuronal sprouting after injury, we used an in vitro model of glial scar, whereby cortical astrocytes were mechanically traumatized to mimic reactive astrogliosis observed after stroke. This mechanical trauma impaired neurite outgrowth from post-natal cortical neurons plated on top of reactive astrocytes. Addition of Shh to the media, however, resulted in a concentration-dependent increase in neurite outgrowth. This response was inhibited by cyclopamine and activated by oxysterol 20(S)-hydroxycholesterol, both of which modulate the activity of the Shh co-receptor Smoothened (Smo), demonstrating that Shh-mediated neurite outgrowth is Smo-dependent. In addition, neurite outgrowth was not associated with an increase in Gli-1 transcription, but could be inhibited by PP2, a selective inhibitor of Src family kinases. These results demonstrate that neurons exposed to the neurite growth inhibitory environment associated with a glial scar can be stimulated by Shh, with signaling occurring through a non-canonical pathway, to overcome this suppression and stimulate neurite outgrowth.
Collapse
Affiliation(s)
- Antonio Berretta
- Department of Anatomy, Brain Health Research Centre, University of Otago, Dunedin 9054, New Zealand.
| | - Emma K. Gowing
- Department of Anatomy, Brain Health Research Centre, University of Otago, Dunedin 9054, New Zealand.
| | - Christine L. Jasoni
- Department of Anatomy, Brain Health Research Centre, University of Otago, Dunedin 9054, New Zealand.
| | - Andrew N. Clarkson
- Department of Anatomy, Brain Health Research Centre, University of Otago, Dunedin 9054, New Zealand.
- Brain Research New Zealand, University of Otago, PO Box 913, Dunedin 9054, New Zealand
- Faculty of Pharmacy, The University of Sydney, Sydney, Australia
| |
Collapse
|
44
|
The Chemorepulsive Protein Semaphorin 3A and Perineuronal Net-Mediated Plasticity. Neural Plast 2016; 2016:3679545. [PMID: 27057361 PMCID: PMC4738953 DOI: 10.1155/2016/3679545] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 12/10/2015] [Indexed: 02/08/2023] Open
Abstract
During postnatal development, closure of critical periods coincides with the appearance of extracellular matrix structures, called perineuronal nets (PNN), around various neuronal populations throughout the brain. The absence or presence of PNN strongly correlates with neuronal plasticity. It is not clear how PNN regulate plasticity. The repulsive axon guidance proteins Semaphorin (Sema) 3A and Sema3B are also prominently expressed in the postnatal and adult brain. In the neocortex, Sema3A accumulates in the PNN that form around parvalbumin positive inhibitory interneurons during the closure of critical periods. Sema3A interacts with high-affinity with chondroitin sulfate E, a component of PNN. The localization of Sema3A in PNN and its inhibitory effects on developing neurites are intriguing features and may clarify how PNN mediate structural neural plasticity. In the cerebellum, enhanced neuronal plasticity as a result of an enriched environment correlates with reduced Sema3A expression in PNN. Here, we first review the distribution of Sema3A and Sema3B expression in the rat brain and the biochemical interaction of Sema3A with PNN. Subsequently, we review what is known so far about functional correlates of changes in Sema3A expression in PNN. Finally, we propose a model of how Semaphorins in the PNN may influence local connectivity.
Collapse
|
45
|
Fernández-Klett F, Priller J. The fibrotic scar in neurological disorders. Brain Pathol 2015; 24:404-13. [PMID: 24946078 DOI: 10.1111/bpa.12162] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 05/26/2014] [Indexed: 01/18/2023] Open
Abstract
Tissue fibrosis, or scar formation, is a common response to damage in most organs of the body. The central nervous system (CNS) is special in that fibrogenic cells are restricted to vascular and meningeal niches. However, disruption of the blood-brain barrier and inflammation can unleash stromal cells and trigger scar formation. Astroglia segregate from the inflammatory lesion core, and the so-called "glial scar" composed of hypertrophic astrocytes seals off the intact neural tissue from damage. In the lesion core, a second type of "fibrotic scar" develops, which is sensitive to inflammatory mediators. Genetic fate mapping studies suggest that pericytes and perivascular fibroblasts are activated, but other precursor cells may also be involved in generating a transient fibrous extracellular matrix in the CNS. The stromal cells sense inflammation and attract immune cells, which in turn drive myofibroblast transdifferentiation. We believe that the fibrotic scar represents a major barrier to CNS regeneration. Targeting of fibrosis may therefore prove to be a valuable therapeutic strategy for neurological disorders such as stroke, spinal cord injury and multiple sclerosis.
Collapse
Affiliation(s)
- Francisco Fernández-Klett
- Department of Neuropsychiatry and Laboratory of Molecular Psychiatry, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | |
Collapse
|
46
|
Chin MR, Zlotkowski K, Han M, Patel S, Eliasen AM, Axelrod A, Siegel D. Expedited access to vinaxanthone and chemically edited derivatives possessing neuronal regenerative effects through ynone coupling reactions. ACS Chem Neurosci 2015; 6:542-50. [PMID: 25615693 DOI: 10.1021/cn500237z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The natural product vinaxanthone has demonstrated a remarkable capability to promote nerve growth following injury or transplantation. In rats following total transection of the spinal cord delivery of vinaxanthone enhanced axonal regeneration, remyelination and angiogenesis at the site of injury all leading to an improved reinstatement of motor function. Through the development of a new ynone coupling reaction, chemically edited derivatives of vinaxanthone have been prepared and studied for improved activity. The coupling reaction allows rapid access to new derivatives, wherein n ynone precursors provide n(2) vinaxanthone analogues. These compounds have been tested for their ability to promote neuronal regrowth using laser axotomy, severing axonal connections in Caenorhabditis elegans. This precise microsurgery using C. elegans allows a new in vivo approach for medicinal chemistry based optimization of neuronal growth promoting compounds.
Collapse
Affiliation(s)
- Matthew R. Chin
- Department
of Chemistry, The University of Texas at Austin, Austin, Texas 78712, United States
- Skaggs
School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California 92039, United States
| | - Katherine Zlotkowski
- Department
of Chemistry, The University of Texas at Austin, Austin, Texas 78712, United States
- Skaggs
School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California 92039, United States
| | - Michelle Han
- Department
of Chemistry, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Saagar Patel
- Department
of Chemistry, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Anders M. Eliasen
- Department
of Chemistry, The University of Texas at Austin, Austin, Texas 78712, United States
- Skaggs
School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California 92039, United States
| | - Abram Axelrod
- Department
of Chemistry, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Dionicio Siegel
- Department
of Chemistry, The University of Texas at Austin, Austin, Texas 78712, United States
- Skaggs
School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California 92039, United States
| |
Collapse
|
47
|
Ruschel J, Hellal F, Flynn KC, Dupraz S, Elliott DA, Tedeschi A, Bates M, Sliwinski C, Brook G, Dobrindt K, Peitz M, Brüstle O, Norenberg MD, Blesch A, Weidner N, Bunge MB, Bixby JL, Bradke F. Axonal regeneration. Systemic administration of epothilone B promotes axon regeneration after spinal cord injury. Science 2015; 348:347-52. [PMID: 25765066 DOI: 10.1126/science.aaa2958] [Citation(s) in RCA: 333] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 02/25/2015] [Indexed: 12/14/2022]
Abstract
After central nervous system (CNS) injury, inhibitory factors in the lesion scar and poor axon growth potential prevent axon regeneration. Microtubule stabilization reduces scarring and promotes axon growth. However, the cellular mechanisms of this dual effect remain unclear. Here, delayed systemic administration of a blood-brain barrier-permeable microtubule-stabilizing drug, epothilone B (epoB), decreased scarring after rodent spinal cord injury (SCI) by abrogating polarization and directed migration of scar-forming fibroblasts. Conversely, epothilone B reactivated neuronal polarization by inducing concerted microtubule polymerization into the axon tip, which propelled axon growth through an inhibitory environment. Together, these drug-elicited effects promoted axon regeneration and improved motor function after SCI. With recent clinical approval, epothilones hold promise for clinical use after CNS injury.
Collapse
Affiliation(s)
- Jörg Ruschel
- Axonal Growth and Regeneration, German Center for Neurodegenerative Diseases, Ludwig-Erhard-Allee 2, 53175 Bonn, Germany
| | - Farida Hellal
- Axonal Growth and Regeneration, German Center for Neurodegenerative Diseases, Ludwig-Erhard-Allee 2, 53175 Bonn, Germany
| | - Kevin C Flynn
- Axonal Growth and Regeneration, German Center for Neurodegenerative Diseases, Ludwig-Erhard-Allee 2, 53175 Bonn, Germany
| | - Sebastian Dupraz
- Axonal Growth and Regeneration, German Center for Neurodegenerative Diseases, Ludwig-Erhard-Allee 2, 53175 Bonn, Germany
| | - David A Elliott
- Axonal Growth and Regeneration, German Center for Neurodegenerative Diseases, Ludwig-Erhard-Allee 2, 53175 Bonn, Germany
| | - Andrea Tedeschi
- Axonal Growth and Regeneration, German Center for Neurodegenerative Diseases, Ludwig-Erhard-Allee 2, 53175 Bonn, Germany
| | - Margaret Bates
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, 1095 Northwest 14th Terrace, Miami, FL33136, USA
| | - Christopher Sliwinski
- Spinal Cord Injury Center, Heidelberg University Hospital, Schlierbacher Landstr. 200A, 69118 Heidelberg, Germany
| | - Gary Brook
- Institute for Neuropathology, RWTH Aachen University, Steinbergweg 20, 52074, Aachen, Germany. Jülich-Aachen Research Alliance-Translational Brain Medicine
| | - Kristina Dobrindt
- Institute of Reconstructive Neurobiology, Life&Brain Center, University of Bonn and Hertie Foundation, Sigmund-Freud-Strasse 25, 53127 Bonn, Germany
| | - Michael Peitz
- Institute of Reconstructive Neurobiology, Life&Brain Center, University of Bonn and Hertie Foundation, Sigmund-Freud-Strasse 25, 53127 Bonn, Germany
| | - Oliver Brüstle
- Institute of Reconstructive Neurobiology, Life&Brain Center, University of Bonn and Hertie Foundation, Sigmund-Freud-Strasse 25, 53127 Bonn, Germany
| | - Michael D Norenberg
- Departments of Pathology, Biochemistry and Molecular Biology, University of Miami School of Medicine, Miami, FL 33101, USA
| | - Armin Blesch
- Spinal Cord Injury Center, Heidelberg University Hospital, Schlierbacher Landstr. 200A, 69118 Heidelberg, Germany
| | - Norbert Weidner
- Spinal Cord Injury Center, Heidelberg University Hospital, Schlierbacher Landstr. 200A, 69118 Heidelberg, Germany
| | - Mary Bartlett Bunge
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, 1095 Northwest 14th Terrace, Miami, FL33136, USA
| | - John L Bixby
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, 1095 Northwest 14th Terrace, Miami, FL33136, USA
| | - Frank Bradke
- Axonal Growth and Regeneration, German Center for Neurodegenerative Diseases, Ludwig-Erhard-Allee 2, 53175 Bonn, Germany.
| |
Collapse
|
48
|
Carballo-Molina OA, Velasco I. Hydrogels as scaffolds and delivery systems to enhance axonal regeneration after injuries. Front Cell Neurosci 2015; 9:13. [PMID: 25741236 PMCID: PMC4330895 DOI: 10.3389/fncel.2015.00013] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 01/09/2015] [Indexed: 01/24/2023] Open
Abstract
Damage caused to neural tissue by disease or injury frequently produces a discontinuity in the nervous system (NS). Such damage generates diverse alterations that are commonly permanent, due to the limited regeneration capacity of the adult NS, particularly the Central Nervous System (CNS). The cellular reaction to noxious stimulus leads to several events such as the formation of glial and fibrous scars, which inhibit axonal regeneration in both the CNS and the Peripheral Nervous System (PNS). Although in the PNS there is some degree of nerve regeneration, it is common that the growing axons reinnervate incorrect areas, causing mismatches. Providing a permissive substrate for axonal regeneration in combination with delivery systems for the release of molecules, which enhances axonal growth, could increase regeneration and the recovery of functions in the CNS or the PNS. Currently, there are no effective vehicles to supply growth factors or cells to the damaged/diseased NS. Hydrogels are polymers that are biodegradable, biocompatible and have the capacity to deliver a large range of molecules in situ. The inclusion of cultured neural cells into hydrogels forming three-dimensional structures allows the formation of synapses and neuronal survival. There is also evidence showing that hydrogels constitute an amenable substrate for axonal growth of endogenous or grafted cells, overcoming the presence of axonal regeneration inhibitory molecules, in both the CNS and PNS. Recent experiments suggest that hydrogels can carry and deliver several proteins relevant for improving neuronal survival and axonal growth. Although the use of hydrogels is appealing, its effectiveness is still a matter of discussion, and more results are needed to achieve consistent recovery using different parameters. This review also discusses areas of opportunity where hydrogels can be applied, in order to promote axonal regeneration of the NS.
Collapse
Affiliation(s)
- Oscar A. Carballo-Molina
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de MéxicoMexico, D.F., Mexico
| | - Iván Velasco
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de MéxicoMexico, D.F., Mexico
| |
Collapse
|
49
|
Kilinc D, Blasiak A, O'Mahony JJ, Lee GU. Low piconewton towing of CNS axons against diffusing and surface-bound repellents requires the inhibition of motor protein-associated pathways. Sci Rep 2014; 4:7128. [PMID: 25417891 PMCID: PMC4241520 DOI: 10.1038/srep07128] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 11/03/2014] [Indexed: 12/24/2022] Open
Abstract
Growth cones, dynamic structures at axon tips, integrate chemical and physical stimuli and translate them into coordinated axon behaviour, e.g., elongation or turning. External force application to growth cones directs and enhances axon elongation in vitro; however, direct mechanical stimulation is rarely combined with chemotactic stimulation. We describe a microfluidic device that exposes isolated cortical axons to gradients of diffusing and substrate-bound molecules, and permits the simultaneous application of piconewton (pN) forces to multiple individual growth cones via magnetic tweezers. Axons treated with Y-27632, a RhoA kinase inhibitor, were successfully towed against Semaphorin 3A gradients, which repel untreated axons, with less than 12 pN acting on a small number of neural cell adhesion molecules. Treatment with Y-27632 or monastrol, a kinesin-5 inhibitor, promoted axon towing on substrates coated with chondroitin sulfate proteoglycans, potent axon repellents. Thus, modulating key molecular pathways that regulate contractile stress generation in axons counteracts the effects of repellent molecules and promotes tension-induced growth. The demonstration of parallel towing of axons towards inhibitory environments with minute forces suggests that mechanochemical stimulation may be a promising therapeutic approach for the repair of the damaged central nervous system, where regenerating axons face repellent factors over-expressed in the glial scar.
Collapse
Affiliation(s)
- Devrim Kilinc
- UCD Nanomedicine Centre, School of Chemistry and Chemical Biology, University College Dublin, Belfield, Dublin 4, Ireland
| | - Agata Blasiak
- UCD Nanomedicine Centre, School of Chemistry and Chemical Biology, University College Dublin, Belfield, Dublin 4, Ireland
| | - James J O'Mahony
- UCD Nanomedicine Centre, School of Chemistry and Chemical Biology, University College Dublin, Belfield, Dublin 4, Ireland
| | - Gil U Lee
- UCD Nanomedicine Centre, School of Chemistry and Chemical Biology, University College Dublin, Belfield, Dublin 4, Ireland
| |
Collapse
|
50
|
Zhang HL, Wang J, Tang L. Sema4D knockdown in oligodendrocytes promotes functional recovery after spinal cord injury. Cell Biochem Biophys 2014; 68:489-96. [PMID: 23949850 DOI: 10.1007/s12013-013-9727-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Semaphorin4D (Sema4D) belongs to Semaphorins family and is secreted and membrane-bound protein. Its function on angiogenesis and axon regeneration makes it an ideal therapeutic target for spinal cord injury (SCI). Here we examined Sema4D expression profile by real-time PCR and western blot and found Sema4D was upregulated after SCI. In vitro study showed Sema4D was not only expressed in oligodendrocytes but also in endothelial cells (ECs). Hypoxia can mimic Sema4D upregulation in both cell lines. Moreover, overexpression of Sema4D through lentivirus in ECs promoted tube formation. However, Sema4D overexpression in oligodendrocytes precursor cells (OPCs) inhibited neuron myelination in neuron-oligodendrocyte co-culture system. Therefore, Sema4D knockdown in OPCs was applied in SCI rats. The results indicated that Sema4D knockdown significantly promoted functional recovery with blood-brain barrier score. Taken together, our data suggest that specific Sema4D knockdown in oligodendrocytes without disturbing its angiogenesis effect can be a beneficial strategy for SCI treatment.
Collapse
Affiliation(s)
- Hong-Lei Zhang
- Department of Spine Surgery, Liaocheng People's Hospital Affiliated to Taishan Medical University, Liaocheng, 252000, Shandong Province, China
| | | | | |
Collapse
|