101
|
Cheng P, Kuang F, Ju G. Aescin reduces oxidative stress and provides neuroprotection in experimental traumatic spinal cord injury. Free Radic Biol Med 2016; 99:405-417. [PMID: 27596954 DOI: 10.1016/j.freeradbiomed.2016.09.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Revised: 09/01/2016] [Accepted: 09/01/2016] [Indexed: 12/22/2022]
Abstract
Aescin has many physiological functions that are highly relevant to spinal cord injury (SCI), including anti-inflammation, anti-oxidation, anti-oedema, and enhancing vascular tone. The present study investigated the putative therapeutic value of aescin in SCI, with a focus on its neuroprotective, anti-inflammatory, and anti-oxidative properties. Sodium aescinate (1.0mg/kg body weight) or equivalent volume of saline was administered 30min after injury by intravenous injection, with an additional dose daily for seven consecutive days after moderate SCI in rats. After contusion injury of the 8th thoracic (T8) spinal cord, aescin-treated rats developed less severe hind limb weakness than saline controls, as assayed by the Basso-Beattie-Bresnahan scale, the beam walking test, and a footprint analysis. The improved locomotor outcomes in aescin-treated rats corresponded to markedly decreased immune response, oxidative stress, neuronal loss, axon demyelination, spinal cord swelling, and cell apoptosis, measured around T8 after impact. Our data suggest aescin treatment as a novel, early, neuroprotective approach in SCI. Given the known safety of aescin in clinical applications, the results of this study suggest that it is a good candidate for SCI treatment in humans.
Collapse
Affiliation(s)
- Peng Cheng
- Institute of Neurosciences, PLA Fourth Military Medical University, 169 West Changle Road, Xi'an 710032, China; Department of Neurology, PLA 425th Hospital, 86 Sanya Bay Road, Sanya 572000, China.
| | - Fang Kuang
- Institute of Neurosciences, PLA Fourth Military Medical University, 169 West Changle Road, Xi'an 710032, China
| | - Gong Ju
- Institute of Neurosciences, PLA Fourth Military Medical University, 169 West Changle Road, Xi'an 710032, China.
| |
Collapse
|
102
|
Spinal cord trauma: pathophysiology, classification of spinal cord injury syndromes, treatment principles and controversies. ACTA ACUST UNITED AC 2016. [DOI: 10.1016/j.mporth.2016.07.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
103
|
Volz KR, Evans KD, Kanner CD, Basso DM. Exploring Targeted Contrast-Enhanced Ultrasound to Detect Neural Inflammation. JOURNAL OF DIAGNOSTIC MEDICAL SONOGRAPHY 2016. [DOI: 10.1177/8756479316665865] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Targeted contrast-enhanced ultrasound (TCEUS) is an innovative method of molecular imaging used for detection of inflammatory biomarkers in vivo. By targeting ultrasound contrast to cell adhesion molecules (CAMs), which are known inflammatory markers within neural tissue, a more direct evaluation of neural inflammation can be made. Due to the novel nature of TCEUS, standardized methods of image analysis do not yet exist. Time intensity curve (TIC) shape analysis is currently used in magnetic resonance contrast imaging to determine temporal behavior of perfusion. Therefore, the presented research attempts to determine TIC shape analysis utility in TCEUS imaging by applying it to TCEUS scans targeted to CAMs present in neural inflammation. This was done in an animal model that underwent a traumatic spinal cord injury to induce inflammation ( n = 31). Subjects were divided into four groups, each receiving a TCEUS targeted to a different CAM seven days after surgery (P-selectin, intracellular adhesion molecule 1 [ICAM-1], vascular cell adhesion molecule 1 [VCAM-1], and control). TICs were generated using average pixel intensity within the injured region of the spinal cord. TIC shape analysis found similar curves were produced while targeting P-selectin and VCAM-1, both demonstrating rapid and sustained enhancement. Control injections demonstrated no enhancement. ICAM-1 injections demonstrated limited enhancement and a shape similar to the control.
Collapse
Affiliation(s)
- Kevin R. Volz
- College of Medicine, School of Health and Rehabilitation Science, The Ohio State University, Columbus, OH, USA
| | - Kevin D. Evans
- College of Medicine, School of Health and Rehabilitation Science, The Ohio State University, Columbus, OH, USA
| | - Christopher D. Kanner
- College of Medicine, School of Health and Rehabilitation Science, The Ohio State University, Columbus, OH, USA
| | - D. Michele Basso
- College of Medicine, School of Health and Rehabilitation Science, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
104
|
Zhang H, Wang Y. Identification of molecular pathway changes after spinal cord injury by microarray analysis. J Orthop Surg Res 2016; 11:101. [PMID: 27628653 PMCID: PMC5024485 DOI: 10.1186/s13018-016-0437-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 09/05/2016] [Indexed: 12/17/2022] Open
Abstract
Background Spinal cord injury (SCI) is highly related to the devastating sensory and motor dysfunction. Methods The GSE45006 gene expression profile dataset was downloaded from Gene Expression Omnibus, which was collected from 24 rats including 20 animals with injured T7 spinal cords using an aneurysm clip impact-compression injury model and killed after 1 day, 3 days, 1 week, 2 weeks, and 8 weeks and four sham-operated rats. Differentially expressed genes (DEGs) between the injured rats at each time point and the sham-operated rats were screened. DEGs commonly detected throughout different time points were further identified, followed by comparing the expression level of these DEGs at each time point between the injured spinal cord samples and controls. Pathway enrichment analysis of the common DEGs was performed. Results The difference in the expression level of 416 common DEGs was significant between the injured spinal cord samples and the controls at each time point (P < 0.05), with the most significant difference 1 day after SCI. The common DEGs were enriched in three pathways, namely Fcγ R-mediated phagocytosis, mitogen-activated protein kinase (MAPK) signaling pathway, and chemokine signaling pathway. AKT3 and RAC2 were enriched in all the three pathways; RAP1B in both MAPK signaling pathway and chemokine signaling pathway; and VAV1, LYN, and HCK in both Fcγ R-mediated phagocytosis and chemokine signaling pathway. Conclusions This study has confirmed the occurrence of neuronal death, inflammation, and neuronal regeneration after SCI. AKT3, RAC2, VAV1, RAP18, LYN, and HCK may have critical roles in the pathological responses to SCI.
Collapse
Affiliation(s)
- Haocong Zhang
- Department of Orthopaedics, The General Hospital of PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Yan Wang
- Department of Orthopaedics, The General Hospital of PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China.
| |
Collapse
|
105
|
Breen BA, Kraskiewicz H, Ronan R, Kshiragar A, Patar A, Sargeant T, Pandit A, McMahon SS. Therapeutic Effect of Neurotrophin-3 Treatment in an Injectable Collagen Scaffold Following Rat Spinal Cord Hemisection Injury. ACS Biomater Sci Eng 2016; 3:1287-1295. [DOI: 10.1021/acsbiomaterials.6b00167] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
| | | | | | | | | | - Timothy Sargeant
- Covidien LLC, 60 Middletown Avenue, North Haven, Connecticut 06473, United States
| | | | | |
Collapse
|
106
|
Albadawi H, Chen JW, Oklu R, Wu Y, Wojtkiewicz G, Pulli B, Milner JD, Cambria RP, Watkins MT. Spinal Cord Inflammation: Molecular Imaging after Thoracic Aortic Ischemia Reperfusion Injury. Radiology 2016; 282:202-211. [PMID: 27509542 DOI: 10.1148/radiol.2016152222] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Purpose To evaluate whether noninvasive molecular imaging technologies targeting myeloperoxidase (MPO) can reveal early inflammation associated with spinal cord injury after thoracic aortic ischemia-reperfusion (TAR) in mice. Materials and Methods The study was approved by the institutional animal care and use committee. C57BL6 mice that were 8-10 weeks old underwent TAR (n = 55) or sham (n = 26) surgery. Magnetic resonance (MR) imaging (n = 6) or single photon emission computed tomography (SPECT)/computed tomography (CT) (n = 15) studies targeting MPO activity were performed after intravenous injection of MPO sensors (bis-5-hydroxytryptamide-tetraazacyclododecane [HT]-diethyneletriaminepentaacetic acid [DTPA]-gadolinium or indium 111-bis-5-HT-DTPA, respectively). Immunohistochemistry and flow cytometry were used to identify myeloid cells and neuronal loss. Proinflammatory cytokines, keratinocyte chemoattractant (KC), and interleukin 6 (IL-6) were measured with enzyme-linked immunosorbent assay. Statistical analyses were performed by using nonparametric tests and the Pearson correlation coefficient. P < .05 was considered to indicate a significant difference. Results Myeloid cells infiltrated into the injured cord at 6 and 24 hours after TAR. MR imaging confirmed the presence of ischemic lesions associated with mild MPO-mediated enhancement in the thoracolumbar spine at 24 hours compared with the sham procedure. SPECT/CT imaging of MPO activity showed marked MPO-sensor retention at 6 hours (P = .003) that continued to increase at 24 hours after TAR (P = .0001). The number of motor neurons decreased substantially at 24 hours after TAR (P < .01), which correlated inversely with in vivo inflammatory changes detected at molecular imaging (r = 0.64, P = .0099). MPO was primarily secreted by neutrophils, followed by lymphocyte antigen 6 complexhigh monocytes and/or macrophages. There were corresponding increased levels of proinflammatory cytokines KC (P = .0001) and IL-6 (P = .0001) that mirrored changes in MPO activity. Conclusion MPO is a suitable imaging biomarker for identifying and tracking inflammatory damage in the spinal cord after TAR in a mouse model. © RSNA, 2016 Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Hassan Albadawi
- From the Department of Surgery, Division of Vascular and Endovascular Surgery (H.A., J.D.M., R.P.C., M.T.W.), and Center for System Biology and Institute for Innovation in Imaging, Department of Radiology (J.W.C., Y.W., G.W., B.P.), Massachusetts General Hospital, Harvard Medical School, 70 Blossom St, Edwards 301, Boston, MA 02114; and Department of Radiology, Division of Vascular and Interventional Radiology, Mayo Clinic, Scottsdale, Ariz (R.O.)
| | - John W Chen
- From the Department of Surgery, Division of Vascular and Endovascular Surgery (H.A., J.D.M., R.P.C., M.T.W.), and Center for System Biology and Institute for Innovation in Imaging, Department of Radiology (J.W.C., Y.W., G.W., B.P.), Massachusetts General Hospital, Harvard Medical School, 70 Blossom St, Edwards 301, Boston, MA 02114; and Department of Radiology, Division of Vascular and Interventional Radiology, Mayo Clinic, Scottsdale, Ariz (R.O.)
| | - Rahmi Oklu
- From the Department of Surgery, Division of Vascular and Endovascular Surgery (H.A., J.D.M., R.P.C., M.T.W.), and Center for System Biology and Institute for Innovation in Imaging, Department of Radiology (J.W.C., Y.W., G.W., B.P.), Massachusetts General Hospital, Harvard Medical School, 70 Blossom St, Edwards 301, Boston, MA 02114; and Department of Radiology, Division of Vascular and Interventional Radiology, Mayo Clinic, Scottsdale, Ariz (R.O.)
| | - Yue Wu
- From the Department of Surgery, Division of Vascular and Endovascular Surgery (H.A., J.D.M., R.P.C., M.T.W.), and Center for System Biology and Institute for Innovation in Imaging, Department of Radiology (J.W.C., Y.W., G.W., B.P.), Massachusetts General Hospital, Harvard Medical School, 70 Blossom St, Edwards 301, Boston, MA 02114; and Department of Radiology, Division of Vascular and Interventional Radiology, Mayo Clinic, Scottsdale, Ariz (R.O.)
| | - Gregory Wojtkiewicz
- From the Department of Surgery, Division of Vascular and Endovascular Surgery (H.A., J.D.M., R.P.C., M.T.W.), and Center for System Biology and Institute for Innovation in Imaging, Department of Radiology (J.W.C., Y.W., G.W., B.P.), Massachusetts General Hospital, Harvard Medical School, 70 Blossom St, Edwards 301, Boston, MA 02114; and Department of Radiology, Division of Vascular and Interventional Radiology, Mayo Clinic, Scottsdale, Ariz (R.O.)
| | - Benjamin Pulli
- From the Department of Surgery, Division of Vascular and Endovascular Surgery (H.A., J.D.M., R.P.C., M.T.W.), and Center for System Biology and Institute for Innovation in Imaging, Department of Radiology (J.W.C., Y.W., G.W., B.P.), Massachusetts General Hospital, Harvard Medical School, 70 Blossom St, Edwards 301, Boston, MA 02114; and Department of Radiology, Division of Vascular and Interventional Radiology, Mayo Clinic, Scottsdale, Ariz (R.O.)
| | - John D Milner
- From the Department of Surgery, Division of Vascular and Endovascular Surgery (H.A., J.D.M., R.P.C., M.T.W.), and Center for System Biology and Institute for Innovation in Imaging, Department of Radiology (J.W.C., Y.W., G.W., B.P.), Massachusetts General Hospital, Harvard Medical School, 70 Blossom St, Edwards 301, Boston, MA 02114; and Department of Radiology, Division of Vascular and Interventional Radiology, Mayo Clinic, Scottsdale, Ariz (R.O.)
| | - Richard P Cambria
- From the Department of Surgery, Division of Vascular and Endovascular Surgery (H.A., J.D.M., R.P.C., M.T.W.), and Center for System Biology and Institute for Innovation in Imaging, Department of Radiology (J.W.C., Y.W., G.W., B.P.), Massachusetts General Hospital, Harvard Medical School, 70 Blossom St, Edwards 301, Boston, MA 02114; and Department of Radiology, Division of Vascular and Interventional Radiology, Mayo Clinic, Scottsdale, Ariz (R.O.)
| | - Michael T Watkins
- From the Department of Surgery, Division of Vascular and Endovascular Surgery (H.A., J.D.M., R.P.C., M.T.W.), and Center for System Biology and Institute for Innovation in Imaging, Department of Radiology (J.W.C., Y.W., G.W., B.P.), Massachusetts General Hospital, Harvard Medical School, 70 Blossom St, Edwards 301, Boston, MA 02114; and Department of Radiology, Division of Vascular and Interventional Radiology, Mayo Clinic, Scottsdale, Ariz (R.O.)
| |
Collapse
|
107
|
Li J, Liu Y, Xu H, Fu Q. Nanoparticle-Delivered IRF5 siRNA Facilitates M1 to M2 Transition, Reduces Demyelination and Neurofilament Loss, and Promotes Functional Recovery After Spinal Cord Injury in Mice. Inflammation 2016; 39:1704-17. [DOI: 10.1007/s10753-016-0405-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
108
|
Kumar H, Ropper AE, Lee SH, Han I. Propitious Therapeutic Modulators to Prevent Blood-Spinal Cord Barrier Disruption in Spinal Cord Injury. Mol Neurobiol 2016; 54:3578-3590. [PMID: 27194298 DOI: 10.1007/s12035-016-9910-6] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Accepted: 05/03/2016] [Indexed: 01/09/2023]
Abstract
The blood-spinal cord barrier (BSCB) is a specialized protective barrier that regulates the movement of molecules between blood vessels and the spinal cord parenchyma. Analogous to the blood-brain barrier (BBB), the BSCB plays a crucial role in maintaining the homeostasis and internal environmental stability of the central nervous system (CNS). After spinal cord injury (SCI), BSCB disruption leads to inflammatory cell invasion such as neutrophils and macrophages, contributing to permanent neurological disability. In this review, we focus on the major proteins mediating the BSCB disruption or BSCB repair after SCI. This review is composed of three parts. Section 1. SCI and the BSCB of the review describes critical events involved in the pathophysiology of SCI and their correlation with BSCB integrity/disruption. Section 2. Major proteins involved in BSCB disruption in SCI focuses on the actions of matrix metalloproteinases (MMPs), tumor necrosis factor alpha (TNF-α), heme oxygenase-1 (HO-1), angiopoietins (Angs), bradykinin, nitric oxide (NO), and endothelins (ETs) in BSCB disruption and repair. Section 3. Therapeutic approaches discusses the major therapeutic compounds utilized to date for the prevention of BSCB disruption in animal model of SCI through modulation of several proteins.
Collapse
Affiliation(s)
- Hemant Kumar
- Department of Neurosurgery, CHA University, CHA Bundang Medical Center, Seongnam-si, Gyeonggi-do, 13496, Republic of Korea
| | - Alexander E Ropper
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX, USA
| | - Soo-Hong Lee
- Department of Biomedical Science, CHA University, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea.
| | - Inbo Han
- Department of Neurosurgery, CHA University, CHA Bundang Medical Center, Seongnam-si, Gyeonggi-do, 13496, Republic of Korea.
| |
Collapse
|
109
|
Addition of low-dose valproic acid to saline resuscitation provides neuroprotection and improves long-term outcomes in a large animal model of combined traumatic brain injury and hemorrhagic shock. J Trauma Acute Care Surg 2016; 79:911-9; discussion 919. [PMID: 26680134 DOI: 10.1097/ta.0000000000000789] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Combined traumatic brain injury (TBI) and hemorrhagic shock (HS) is highly lethal. In a nonsurvival model of TBI + HS, addition of high-dose valproic acid (VPA) (300 mg/kg) to hetastarch reduced brain lesion size and associated swelling 6 hours after injury; whether this would have translated into better neurologic outcomes remains unknown. It is also unclear whether lower doses of VPA would be neuroprotective. We hypothesized that addition of low-dose VPA to normal saline (NS) resuscitation would result in improved long-term neurologic recovery and decreased brain lesion size. METHODS TBI was created in anesthetized swine (40-43 kg) by controlled cortical impact, and volume-controlled hemorrhage (40% volume) was induced concurrently. After 2 hours of shock, animals were randomized (n = 5 per group) to NS (3× shed blood) or NS + VPA (150 mg/kg). Six hours after resuscitation, packed red blood cells were transfused, and animals were recovered. Peripheral blood mononuclear cells were analyzed for acetylated histone-H3 at lysine-9. A Neurological Severity Score (NSS) was assessed daily for 30 days. Brain magnetic resonance imaging was performed on Days 3 and 10. Cognitive performance was assessed by training animals to retrieve food from color-coded boxes. RESULTS There was a significant increase in histone acetylation in the NS + VPA-treated animals compared with NS treatment. The NS + VPA group demonstrated significantly decreased neurologic impairment and faster speed of recovery as well as smaller brain lesion size compared with the NS group. Although the final cognitive function scores were similar between the groups, the VPA-treated animals reached the goal significantly faster than the NS controls. CONCLUSION In this long-term survival model of TBI + HS, addition of low-dose VPA to saline resuscitation resulted in attenuated neurologic impairment, faster neurologic recovery, smaller brain lesion size, and a quicker normalization of cognitive functions.
Collapse
|
110
|
Use of the CatWalk Gait Device to Assess Differences in Locomotion between Genders in Rats Inherently and following Spinal Cord Injury. ACTA ACUST UNITED AC 2016. [DOI: 10.1155/2016/6276348] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
A possible cause of differences in recovery after spinal cord injury (SCI) is gender. The effect of gender on locomotor recovery following SCI, however, remains controversial and has produced conflicting results regarding gender’s impact on outcome. A significant shortcoming of previous studies was small sample size. The current work tested what, if any, significant differences existed between genders after SCI with CatWalk Gait Analysis that uses an automated device to measure the foot placement and gait of animals as they voluntarily cross an illuminated glass runway. We hypothesized that, by employing larger sample sizes in a reproducible and clinically relevant contusive SCI paradigm, subtle distinctions in locomotor recovery between sexes, if they exist, would be elucidated. During 13 weeks of functional assessment after SCI, a number of CatWalk parameters, including swing, single stance, and stride length, were significantly affected by gender only as identified by use of ANCOVA analysis, considering age, weight, and baseline performance as covariates. We report here our findings for 197 parameters that were assessed before and after SCI. Evaluating differences in locomotor recovery between sexes after SCI could point to a gender-related advantage and provide novel directions for the development of future therapeutics.
Collapse
|
111
|
Graphene Functionalized Scaffolds Reduce the Inflammatory Response and Supports Endogenous Neuroblast Migration when Implanted in the Adult Brain. PLoS One 2016; 11:e0151589. [PMID: 26978268 PMCID: PMC4792446 DOI: 10.1371/journal.pone.0151589] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 03/01/2016] [Indexed: 01/02/2023] Open
Abstract
Electroactive materials have been investigated as next-generation neuronal tissue engineering scaffolds to enhance neuronal regeneration and functional recovery after brain injury. Graphene, an emerging neuronal scaffold material with charge transfer properties, has shown promising results for neuronal cell survival and differentiation in vitro. In this in vivo work, electrospun microfiber scaffolds coated with self-assembled colloidal graphene, were implanted into the striatum or into the subventricular zone of adult rats. Microglia and astrocyte activation levels were suppressed with graphene functionalization. In addition, self-assembled graphene implants prevented glial scarring in the brain 7 weeks following implantation. Astrocyte guidance within the scaffold and redirection of neuroblasts from the subventricular zone along the implants was also demonstrated. These findings provide new functional evidence for the potential use of graphene scaffolds as a therapeutic platform to support central nervous system regeneration.
Collapse
|
112
|
Snider S, Cavalli A, Colombo F, Gallotti AL, Quattrini A, Salvatore L, Madaghiele M, Terreni MR, Sannino A, Mortini P. A novel composite type I collagen scaffold with micropatterned porosity regulates the entrance of phagocytes in a severe model of spinal cord injury. J Biomed Mater Res B Appl Biomater 2016; 105:1040-1053. [PMID: 26958814 DOI: 10.1002/jbm.b.33645] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 01/29/2016] [Accepted: 02/12/2016] [Indexed: 01/28/2023]
Abstract
Traumatic spinal cord injury (SCI) is a damage to the spinal cord that results in loss or impaired motor and/or sensory function. SCI is a sudden and unexpected event characterized by high morbidity and mortality rate during both acute and chronic stages, and it can be devastating in human, social and economical terms. Despite significant progresses in the clinical management of SCI, there remain no effective treatments to improve neurological outcomes. Among experimental strategies, bioengineered scaffolds have the potential to support and guide injured axons contributing to neural repair. The major aim of this study was to investigate a novel composite type I collagen scaffold with micropatterned porosity in a rodent model of severe spinal cord injury. After segment resection of the thoracic spinal cord we implanted the scaffold in female Sprague-Dawley rats. Controls were injured without receiving implantation. Behavioral analysis of the locomotor performance was monitored up to 55 days postinjury. Two months after injury histopathological analysis were performed to evaluate the extent of scar and demyelination, the presence of connective tissue and axonal regrowth through the scaffold and to evaluate inflammatory cell infiltration at the injured site. We provided evidence that the new collagen scaffold was well integrated with the host tissue, slightly ameliorated locomotor function, and limited the robust recruitment of the inflammatory cells at the injury site during both the acute and chronic stage in spinal cord injured rats. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 105B: 1040-1053, 2017.
Collapse
Affiliation(s)
- Silvia Snider
- Division of Neurosurgery, San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Andrea Cavalli
- Division of Neurosurgery, San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Francesca Colombo
- Division of Neurosurgery, San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Alberto Luigi Gallotti
- Division of Neurosurgery, San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Angelo Quattrini
- Division of Neuroscience and INSPE, San Raffaele Scientific Institute, via Olgettina 60, 20132, Milan, Italy
| | - Luca Salvatore
- Department of Engineering for Innovation, University of Salento, Via per Monteroni, 73100, Lecce, Italy
| | - Marta Madaghiele
- Department of Engineering for Innovation, University of Salento, Via per Monteroni, 73100, Lecce, Italy
| | - Maria Rosa Terreni
- Division of Pathology, San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Alessandro Sannino
- Department of Engineering for Innovation, University of Salento, Via per Monteroni, 73100, Lecce, Italy
| | - Pietro Mortini
- Division of Neurosurgery, San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| |
Collapse
|
113
|
Golipoor Z, Mehraein F, Zafari F, Alizadeh A, Ababzadeh S, Baazm M. Migration of Bone Marrow-Derived Very Small Embryonic-Like Stem Cells toward An Injured Spinal Cord. CELL JOURNAL 2016; 17:639-47. [PMID: 26862523 PMCID: PMC4746414 DOI: 10.22074/cellj.2016.3836] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 10/08/2014] [Indexed: 11/04/2022]
Abstract
OBJECTIVE Bone marrow (BM) is one of the major hematopoietic organs in postnatal life that consists of a heterogeneous population of stem cells which have been previously described. Recently, a rare population of stem cells that are called very small embryonic-like (VSEL) stem cells has been found in the BM. These cells express several developmental markers of pluri- potent stem cells and can be mobilized into peripheral blood (PB) in response to tissue injury. In this study we have attempted to investigate the ability of these cells to migrate toward an injured spinal cord after transplantation through the tail vein in a rat model. MATERIALS AND METHODS In this experimental study, VSELs were isolated from total BM cells using a fluorescent activated cell sorting (FACS) system and sca1 and stage specific embryonic antigen (SSEA-1) antibodies. After isolation, VSELs were cultured for 7 days on C2C12 as the feeder layer. Then, VSELs were labeled with 1,1´-dioctadecyl-3,3,3´,3´- tetramethylindocarbocyanine perchlorate (DiI) and transplanted into the rat spinal cord injury (SCI) model via the tail vein. Finally, we sought to determine the presence of VSELs in the lesion site. RESULTS We isolated a high number of VSELs from the BM. After cultivation, the VSELs colonies were positive for SSEA-1, Oct4 and Sca1. At one month after transplantation, real-time polymerase chain reaction analysis confirmed a significantly increased expres- sion level of Oct4 and SSEA-1 positive cells at the injury site. CONCLUSION VSELs have the capability to migrate and localize in an injured spinal cord after transplantation.
Collapse
Affiliation(s)
- Zoleikha Golipoor
- Department of Anatomy, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Fereshteh Mehraein
- Department of Anatomy, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fariba Zafari
- Department of Anatomy, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Akram Alizadeh
- Department of Tissue Engineering, Faculty of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Shima Ababzadeh
- Department of Anatomy, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Baazm
- Department of Anatomy, Faculty of Medicine, Arak University of Medical Sciences, Arak, Iran
| |
Collapse
|
114
|
Cannabinoids to treat spinal cord injury. Prog Neuropsychopharmacol Biol Psychiatry 2016; 64:190-9. [PMID: 25805333 DOI: 10.1016/j.pnpbp.2015.03.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 03/09/2015] [Accepted: 03/13/2015] [Indexed: 02/06/2023]
Abstract
Spinal cord injury (SCI) is a devastating condition for which there is no standard treatment beyond rehabilitation strategies. In this review, we discuss the current knowledge on the use of cannabinoids to treat this condition. The endocannabinoid system is expressed in the intact spinal cord, and it is dramatically upregulated after lesion. Endogenous activation of this system counteracts secondary damage following SCI, and treatments with endocannabinoids or synthetic cannabinoid receptor agonists promote a better functional outcome in experimental models. The use of cannabinoids in SCI is a new research field and many questions remain open. Here, we discuss caveats and suggest some future directions that may help to understand the role of cannabinoids in SCI and how to take advantage of this system to regain functions after spinal cord damage.
Collapse
|
115
|
Boumaiza S, Oussedik-Oumehdi H, Laraba-Djebari F. Pathophysiological effects of Cerastes cerastes and Vipera lebetina venoms: Immunoneutralization using anti-native and anti-60Co irradiated venoms. Biologicals 2016; 44:1-11. [DOI: 10.1016/j.biologicals.2015.11.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 11/09/2015] [Indexed: 01/02/2023] Open
|
116
|
Wang B, Yuan J, Xu J, Xie J, Wang G, Dong P. Neurotrophin expression and laryngeal muscle pathophysiology following recurrent laryngeal nerve transection. Mol Med Rep 2015; 13:1234-42. [PMID: 26677138 PMCID: PMC4732864 DOI: 10.3892/mmr.2015.4684] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 11/19/2015] [Indexed: 11/05/2022] Open
Abstract
Laryngeal palsy often occurs as a result of recurrent laryngeal or vagal nerve injury during oncological surgery of the head and neck, affecting quality of life and increasing economic burden. Reinnervation following recurrent laryngeal nerve (RLN) injury is difficult despite development of techniques, such as neural anastomosis, nerve grafting and creation of a laryngeal muscle pedicle. In the present study, due to the limited availability of human nerve tissue for research, a rat model was used to investigate neurotrophin expression and laryngeal muscle pathophysiology in RLN injury. Twenty-five male Sprague-Dawley rats underwent right RLN transection with the excision of a 5-mm segment. Vocal fold movements, vocalization, histology and immunostaining were evaluated at different time-points (3, 6, 10 and 16 weeks). Although vocalization was restored, movement of the vocal fold failed to return to normal levels following RLN injury. The expression of brain‑derived neurotrophic factor and glial cell line-derived neurotrophic factor differed in the thyroarytenoid (TA) and posterior cricoarytenoid muscles. The number of axons did not increase to baseline levels over time. Furthermore, normal muscle function was unlikely with spontaneous reinnervation. During regeneration following RLN injury, differences in the expression levels of neurotrophic factors may have resulted in preferential reinnervation of the TA muscles. Data from the present study indicated that neurotrophic factors may be applied for restoring the function of the laryngeal nerve following recurrent injury.
Collapse
Affiliation(s)
- Baoxin Wang
- Department of Otolaryngology, Head and Neck Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, P.R. China
| | - Junjie Yuan
- Department of Orthopedics, Shanghai Fengxian District Central Hospital, Shanghai Jiao Tong University Affiliated Sixth People's Hospital South Campus, Shanghai 200011, P.R. China
| | - Jiafeng Xu
- School of Economics and Finance, Shanghai International Studies University, Shanghai 200083, P.R. China
| | - Jin Xie
- Department of Otolaryngology, Head and Neck Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, P.R. China
| | - Guoliang Wang
- Department of Otolaryngology, Head and Neck Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, P.R. China
| | - Pin Dong
- Department of Otolaryngology, Head and Neck Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, P.R. China
| |
Collapse
|
117
|
An in vivo model of anti-inflammatory activity of subdural dexamethasone following the spinal cord injury. Neurol Neurochir Pol 2015; 50:7-15. [PMID: 26851684 DOI: 10.1016/j.pjnns.2015.10.006] [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: 05/27/2015] [Revised: 09/03/2015] [Accepted: 10/14/2015] [Indexed: 11/21/2022]
Abstract
Current therapies to limit the neural tissue destruction following the spinal cord injury are not effective. Our recent studies indicate that the injury to the white matter of the spinal cord results in a severe inflammatory response where macrophages phagocytize damaged myelin and the fluid-filled cavity of injury extends in size with concurrent and irreversible destruction of the surrounding neural tissue over several months. We previously established that a high dose of 4mg/rat of dexamethasone administered for 1 week via subdural infusion remarkably lowers the numbers of infiltrating macrophages leaving large amounts of un-phagocytized myelin debris and therefore inhibits the severity of inflammation and related tissue destruction. But this dose was potently toxic to the rats. In the present study the lower doses of dexamethasone, 0.125-2.0mg, were administered via the subdural infusion for 2 weeks after an epidural balloon crush of the mid-thoracic spinal cord. The spinal cord cross-sections were analyzed histologically. Levels of dexamethasone used in the current study had no systemic toxic effect and limited phagocytosis of myelin debris by macrophages in the lesion cavity. The subdural infusion with 0.125-2.0mg dexamethasone over 2 week period did not eliminate the inflammatory process indicating the need for a longer period of infusion to do so. However, this treatment has probably lead to inhibition of the tissue destruction by the severe, prolonged inflammatory process.
Collapse
|
118
|
Li F, Cheng B, Cheng J, Wang D, Li H, He X. CCR5 blockade promotes M2 macrophage activation and improves locomotor recovery after spinal cord injury in mice. Inflammation 2015; 38:126-33. [PMID: 25212047 DOI: 10.1007/s10753-014-0014-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Spinal cord injury (SCI) is considered to be primarily associated with loss of motor function and leads to activate diverse cellular mechanisms in the central nervous system to attempt to repair the damaged spinal cord tissue. Chemokine Receptor 5 (CCR5), a major co-receptor for macrophage-tropic human immunodeficiency viruses, is expressed on the surface of monocytes/macrophages, dendritic cells, activated T cells, and NK cells. Recent papers have indicated the important role of CCR5 in SCI, but the mechanism is still unknown. In our current study, CCR5 blockade displayed increased myelin sparring and enhanced SC repair process. The number of CD4(+) T cells, CD8(+) T cells, Ly6G(+) neutrophils and CD11b(+) macrophages were all significantly lower in the anti-CCR5 group than that in the control group after SCI. The IL-4 and IL-13 levels in anti-CCR5 group were markedly higher than that in control group after SCI. Correspondingly, the anti-CCR5-treated group showed increased numbers of Arg1- or CD206-expressing macrophages compared with the control IgG group. Furthermore, CCR5 blockade promoted PPARγ activation, and the increased numbers of M2 macrophages induced by CCR5 blockade were both reversed with additional PPARγ antagonist treatment. In conclusion, our present work provides evidence to support the concept that CCR5 blockade promotes M2 macrophage activation and improves locomotor recovery after SCI in mice.
Collapse
Affiliation(s)
- Fengtao Li
- Orthopedics Department Two, The Second Affiliated Hospital of Xi'an Jiaotong University, Xiwu road NO.157, 710004, Xi An, China
| | | | | | | | | | | |
Collapse
|
119
|
Kelderhouse LE, Robins MT, Rosenbalm KE, Hoylman EK, Mahalingam S, Low PS. Prediction of Response to Therapy for Autoimmune/Inflammatory Diseases Using an Activated Macrophage-Targeted Radioimaging Agent. Mol Pharm 2015; 12:3547-55. [PMID: 26333010 DOI: 10.1021/acs.molpharmaceut.5b00134] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The ability to select patients who will respond to therapy is especially acute for autoimmune/inflammatory diseases, where the costs of therapies can be high and the progressive damage associated with ineffective treatments can be irreversible. In this article we describe a clinical test that will rapidly predict the response of patients with an autoimmune/inflammatory disease to many commonly employed therapies. This test involves quantitative assessment of uptake of a folate receptor-targeted radioimaging agent ((99m)Tc-EC20) by a subset of inflammatory macrophages that accumulate at sites of inflammation. Murine models of four representative inflammatory diseases (rheumatoid arthritis, inflammatory bowel disease, pulmonary fibrosis, and atherosclerosis) show markedly decreased uptake of (99m)Tc-EC20 in inflamed lesions upon initiation of successful therapies, but no decrease in uptake upon administration of ineffective therapies, in both cases long before changes in clinical symptoms can be detected. This predictive capability should reduce costs and minimize morbidities associated with failed autoimmune/inflammatory disease therapies.
Collapse
Affiliation(s)
- Lindsay E Kelderhouse
- Department of Chemistry, Purdue University , West Lafayette, Indiana 47907, United States
| | - Meridith T Robins
- Department of Chemistry, Purdue University , West Lafayette, Indiana 47907, United States
| | - Katelyn E Rosenbalm
- Department of Chemistry, Purdue University , West Lafayette, Indiana 47907, United States
| | - Emily K Hoylman
- Department of Chemistry, Purdue University , West Lafayette, Indiana 47907, United States
| | | | - Philip S Low
- Department of Chemistry, Purdue University , West Lafayette, Indiana 47907, United States
| |
Collapse
|
120
|
Xu X, Li N, Zhu L, Zhou Y, Cheng H. Beneficial effects of local profound hypothermia and the possible mechanism after experimental spinal cord injury in rats. J Spinal Cord Med 2015; 39:220-8. [PMID: 26322652 PMCID: PMC5072505 DOI: 10.1179/2045772315y.0000000051] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
OBJECTIVE The primary focus of this study was to investigate the effects of local profound hypothermia and to explore the possible mechanism in adult rats with spinal cord injury. STUDY DESIGN AND METHODS Spinal cord injury models were established by placing aneurysm clips on T10. An epidural perfusion device was applied to maintain a steady temperature (18 °C) for 120 min with gradual rewarming to 37 °C Total hypothermic duration lasted up to about 170 min. The expression of axon regeneration inhibitors was tested by Western blot and real-time PCR. Luxol Fast Blue (LFB) stain and Bielschowsky silver stain were used to observe spinal cord morphology. Motor function of the hind limbs (BBB score) was monitored for 21 days. RESULTS The expressions of RhoA, ROCK-II, NG2, Neurocan, Brevican, and Nogo-A were downregulated by regional hypothermia (RH) after spinal cord injury. Subsequent observation showed that rats that had received RH had an alleviated demyelinating condition and a greater number of nerve fibers. Furthermore, the RH group achieved higher BBB scores than the spinal cord injury (SCI) group. CONCLUSIONS Recovery of hind limb function in rats can be promoted by local profound hypothermia; this may be caused by the suppression of axon regeneration inhibitors.
Collapse
Affiliation(s)
| | | | | | | | - Huilin Cheng
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| |
Collapse
|
121
|
Endogenous Two-Photon Excited Fluorescence Provides Label-Free Visualization of the Inflammatory Response in the Rodent Spinal Cord. BIOMED RESEARCH INTERNATIONAL 2015; 2015:859084. [PMID: 26355949 PMCID: PMC4555451 DOI: 10.1155/2015/859084] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 07/19/2015] [Accepted: 07/27/2015] [Indexed: 02/07/2023]
Abstract
Activation of CNS resident microglia and invasion of external macrophages plays a central role in spinal cord injuries and diseases. Multiphoton microscopy based on intrinsic tissue properties offers the possibility of label-free imaging and has the potential to be applied in vivo. In this work, we analyzed cellular structures displaying endogenous two-photon excited fluorescence (TPEF) in the pathologic spinal cord. It was compared qualitatively and quantitatively to Iba1 and CD68 immunohistochemical staining in two models: rat spinal cord injury and mouse encephalomyelitis. The extent of tissue damage was retrieved by coherent anti-Stokes Raman scattering (CARS) and second harmonic generation imaging. The pattern of CD68-positive cells representing postinjury activated microglia/macrophages was colocalized to the TPEF signal. Iba1-positive microglia were found in areas lacking any TPEF signal. In peripheral areas of inflammation, we found similar numbers of CD68-positive microglia/macrophages and TPEF-positive structures while the number of Iba1-positive cells was significantly higher. Therefore, we conclude that multiphoton imaging of unstained spinal cord tissue enables retrieving the extent of microglia activation by acquisition of endogenous TPEF. Future application of this technique in vivo will enable monitoring inflammatory responses of the nervous system allowing new insights into degenerative and regenerative processes.
Collapse
|
122
|
Evaluation of Injured Axons Using Two-Photon Excited Fluorescence Microscopy after Spinal Cord Contusion Injury in YFP-H Line Mice. Int J Mol Sci 2015; 16:15785-99. [PMID: 26184175 PMCID: PMC4519925 DOI: 10.3390/ijms160715785] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 07/02/2015] [Accepted: 07/08/2015] [Indexed: 11/17/2022] Open
Abstract
Elucidation of the process of degeneration of injured axons is important for the development of therapeutic modules for the treatment of spinal cord injuries. The aim of this study was to establish a method for time-lapse observation of injured axons in living animals after spinal cord contusion injury. YFP (yellow fluorescent protein)-H transgenic mice, which we used in this study, express fluorescence in their nerve fibers. Contusion damage to the spinal cord at the 11th vertebra was performed by IH (Infinite Horizon) impactor, which applied a pressure of 50 kdyn. The damaged spinal cords were re-exposed during the observation period under anesthesia, and then observed by two-photon excited fluorescence microscopy, which can observe deep regions of tissues including spinal cord axons. No significant morphological change of injured axons was observed immediately after injury. Three days after injury, the number of axons decreased, and residual axons were fragmented. Seven days after injury, only fragments were present in the damaged tissue. No hind-limb movement was observed during the observation period after injury. Despite the immediate paresis of hind-limbs following the contusion injury, the morphological degeneration of injured axons was delayed. This method may help clarification of pathophysiology of axon degeneration and development of therapeutic modules for the treatment of spinal cord injury.
Collapse
|
123
|
Therapeutic Efficacy of E-64-d, a Selective Calpain Inhibitor, in Experimental Acute Spinal Cord Injury. BIOMED RESEARCH INTERNATIONAL 2015; 2015:134242. [PMID: 26240815 PMCID: PMC4512559 DOI: 10.1155/2015/134242] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 05/30/2015] [Accepted: 06/15/2015] [Indexed: 12/18/2022]
Abstract
This study aims to investigate the therapeutic effect of calpain inhibitor E-64-d on SCI and to find a new approach to treat SCI. When an SCI rat model was established, it was immediately administered with E-64-d. RT-PCR and Western blotting were used to determine the protein and mRNA levels of calpain 1 and 68-kD NFP. TUNEL staining and NeuN labeling were performed to analyze neuronal apoptosis in the lesion. Immunohistochemistry assay was carried out to observe the expressions of calpain 1 and GFAP. Cyclooxygenase-2 activity was measured to show the immune response status. Locomotor function was evaluated by inclined plane test and Basso, Beattie, and Bresnahan locomotor rating scale. The results showed that calpain 1 was activated after SCI occurred. Treatment with E-64-d decreased expressions of calpain 1 and GFAP, alleviated neuronal apoptosis, inhibited cyclooxygenase-2 activity, and resulted in the promoted locomotor function. Furthermore, combination of E-64-d and MP had better efficacy than did E-64-d or MP alone. E-64-d is expected to be applied to treat SCI, and its alliance with MP may provide a valid strategy for SCI therapy.
Collapse
|
124
|
Gadani SP, Walsh JT, Lukens JR, Kipnis J. Dealing with Danger in the CNS: The Response of the Immune System to Injury. Neuron 2015; 87:47-62. [PMID: 26139369 PMCID: PMC4491143 DOI: 10.1016/j.neuron.2015.05.019] [Citation(s) in RCA: 213] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Fighting pathogens and maintaining tissue homeostasis are prerequisites for survival. Both of these functions are upheld by the immune system, though the latter is often overlooked in the context of the CNS. The mere presence of immune cells in the CNS was long considered a hallmark of pathology, but this view has been recently challenged by studies demonstrating that immunological signaling can confer pivotal neuroprotective effects on the injured CNS. In this review, we describe the temporal sequence of immunological events that follow CNS injury. Beginning with immediate changes at the injury site, including death of neural cells and release of damage-associated molecular patterns (DAMPs), and progressing through innate and adaptive immune responses, we describe the cascade of inflammatory mediators and the implications of their post-injury effects. We conclude by proposing a revised interpretation of immune privilege in the brain, which takes beneficial neuro-immune communications into account.
Collapse
Affiliation(s)
- Sachin P Gadani
- Center for Brain Immunology and Glia, Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA; Graduate Program in Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA; Medical Scientist Training Program, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - James T Walsh
- Center for Brain Immunology and Glia, Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA; Graduate Program in Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA; Medical Scientist Training Program, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - John R Lukens
- Center for Brain Immunology and Glia, Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA; Graduate Program in Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA.
| | - Jonathan Kipnis
- Center for Brain Immunology and Glia, Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA; Graduate Program in Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA; Medical Scientist Training Program, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA.
| |
Collapse
|
125
|
Le E, Aarabi B, Hersh DS, Shanmuganathan K, Diaz C, Massetti J, Akhtar-Danesh N. Predictors of intramedullary lesion expansion rate on MR images of patients with subaxial spinal cord injury. J Neurosurg Spine 2015; 22:611-21. [DOI: 10.3171/2014.10.spine14576] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT
Studies of preclinical spinal cord injury (SCI) in rodents indicate that expansion of intramedullary lesions (IMLs) seen on MR images may be amenable to neuroprotection. In patients with subaxial SCI and motor-complete American Spinal Injury Association (ASIA) Impairment Scale (AIS) Grade A or B, IML expansion has been shown to be approximately 900 μm/hour. In this study, the authors investigated IML expansion in a cohort of patients with subaxial SCI and AIS Grade A, B, C, or D.
METHODS
Seventy-eight patients who had at least 2 MRI scans within 6 days of SCI were enrolled. Data were analyzed by regression analysis.
RESULTS
In this cohort, the mean age was 45.3 years (SD 18.3 years), 73 patients were injured in a motor vehicle crash, from a fall, or in sport activities, and 77% of them were men. The mean Injury Severity Score (ISS) was 26.7 (SD 16.7), and the AIS grade was A in 23 patients, B in 7, C in 7, and D in 41. The mechanism of injury was distraction in 26 patients, compression in 22, disc/osteophyte complex in 29, and Chance fracture in 1. The mean time between injury onset and the first MRI scan (Interval 1) was 10 hours (SD 8.7 hours), and the mean time to the second MRI scan (Interval 2) was 60 hours (SD 29.6 hours). The mean IML lengths of the first and second MR images were 38.8 mm (SD 20.4 mm) and 51 mm (SD 36.5 mm), respectively. The mean time from the first to the second MRI scan (Interval 3) was 49.9 hours (SD 28.4 hours), and the difference in IML lengths was 12.6 mm (SD 20.7 mm), reflecting an expansion rate of 366 μm/ hour (SD 710 μm/hour). IML expansion in patients with AIS Grades A and B was 918 μm/hour (SD 828 μm/hour), and for those with AIS Grades C and D, it was 21 μm/hour (SD 304 μm/hour). Univariate analysis indicated that AIS Grade A or B versus Grades C or D (p < 0.0001), traction (p= 0.0005), injury morphology (p < 0.005), the surgical approach (p= 0.009), vertebral artery injury (p= 0.02), age (p < 0.05), ISS (p < 0.05), ASIA motor score (p < 0.05), and time to decompression (p < 0.05) were all predictors of lesion expansion. In multiple regression analysis, however, the sole determinant of IML expansion was AIS grade (p < 0.005).
CONCLUSIONS
After traumatic subaxial cervical spine or spinal cord injury, patients with motor-complete injury (AIS Grade A or B) had a significantly higher rate of IML expansion than those with motor-incomplete injury (AIS Grade C or D).
Collapse
Affiliation(s)
| | - Bizhan Aarabi
- 1Department of Neurosurgery and
- 2R Adams Cowley Shock Trauma Center, University of Maryland School of Medicine, Baltimore, Maryland; and
| | | | | | - Cara Diaz
- 2R Adams Cowley Shock Trauma Center, University of Maryland School of Medicine, Baltimore, Maryland; and
| | - Jennifer Massetti
- 2R Adams Cowley Shock Trauma Center, University of Maryland School of Medicine, Baltimore, Maryland; and
| | - Noori Akhtar-Danesh
- 3School of Nursing and Department of Clinical Epidemiology and Biostatistics, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
126
|
Datto JP, Bastidas JC, Miller NL, Shah AK, Arheart KL, Marcillo AE, Dietrich WD, Pearse DD. Female Rats Demonstrate Improved Locomotor Recovery and Greater Preservation of White and Gray Matter after Traumatic Spinal Cord Injury Compared to Males. J Neurotrauma 2015; 32:1146-57. [PMID: 25715192 DOI: 10.1089/neu.2014.3702] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The possibility of a gender-related difference in recovery after spinal cord injury (SCI) remains a controversial subject. Current empirical animal research lacks sizable test groups to definitively determine whether significant differences exist. Evaluating locomotor recovery variances between sexes following a precise, clinically relevant spinal cord contusion model can provide valuable insight into a possible gender-related advantage in outcome post-SCI. In the current study, we hypothesized that by employing larger sample sizes in a reproducible contusive SCI paradigm, subtle distinctions in locomotor recovery between sexes, if they exist, would be elucidated through a broad range of behavioral tests. During 13 weeks of functional assessment after a thoracic (T8) contusive SCI in rat, significant differences owing to gender existed for the Basso, Beattie, and Bresnahan score and CatWalk hindlimb swing, support four, and single stance analyses. Significant differences in locomotor performance were noticeable as early as 4 weeks post-SCI. Stereological tissue-volume analysis determined that females, more so than males, also exhibited greater volumes of preserved gray and white matter within the injured cord segment as well as more spared ventral white matter area at the center of the lesion. The stereological tissue analysis differences favoring females directly correlated with the female rats' greater functional improvement observed at endpoint.
Collapse
Affiliation(s)
- Jeffrey P Datto
- 1 The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine , Miami, Florida
| | - Johana C Bastidas
- 1 The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine , Miami, Florida
| | - Nicole L Miller
- 1 The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine , Miami, Florida
| | - Anna K Shah
- 1 The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine , Miami, Florida
| | - Kristopher L Arheart
- 2 The Departments of Public Health Sciences, University of Miami Miller School of Medicine , Miami, Florida
| | - Alexander E Marcillo
- 1 The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine , Miami, Florida
| | - W Dalton Dietrich
- 1 The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine , Miami, Florida.,3 The Department of Neurological Surgery, University of Miami Miller School of Medicine , Miami, Florida.,4 The Department of Cell Biology, University of Miami Miller School of Medicine , Miami, Florida.,5 The Department of Neurology, University of Miami Miller School of Medicine , Miami, Florida.,6 The Neuroscience Program, University of Miami Miller School of Medicine , Miami, Florida
| | - Damien D Pearse
- 1 The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine , Miami, Florida.,3 The Department of Neurological Surgery, University of Miami Miller School of Medicine , Miami, Florida.,6 The Neuroscience Program, University of Miami Miller School of Medicine , Miami, Florida.,7 The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine , Miami, Florida
| |
Collapse
|
127
|
Chamisha Y, Aroch I, Kuzi S, Srugo I, Bdolah-Abram T, Chai O, Christopher MM, Merbl Y, Rothwell K, Shamir MH. The prognostic value of cerebrospinal fluid characteristics in dogs without deep pain perception due to thoracolumbar disc herniation. Res Vet Sci 2015; 100:189-96. [PMID: 25957960 DOI: 10.1016/j.rvsc.2015.03.017] [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: 11/12/2014] [Revised: 02/03/2015] [Accepted: 03/14/2015] [Indexed: 01/12/2023]
Abstract
Providing a pre-operative prognosis for dogs presented with absent deep pain perception (DPP) is extremely challenging, as the overall recovery rates widely vary. This study assesses the possible correlation between the severity of spinal cord injury and CSF cytology in 31 paraplegic dogs presented with absent DPP due to acute thoracolumbar intervertebral disc herniation (TL-IVDH). All dogs underwent surgical decompression immediately following diagnosis. CSF TNCC, macrophage percentage and macrophage to monocyte (MΦ:M) ratio were significantly higher in dogs that failed to regain DPP within 10 days post-operatively and in dogs that failed to regain ambulation at the end of the study period (P< 0.05). MΦ:M of 0.73 and higher corresponded to a sensitivity of 54% and specificity of 100% for prediction of a negative long-term outcome. CSF TNCC, macrophage percentage and MΦ:M ratio effectively predicted regaining DPP and the long-term outcome in dogs that lost DPP due to acute TL-IVDH.
Collapse
Affiliation(s)
- Y Chamisha
- Department of Neurology and Neurosurgery, Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 76100, Israel
| | - I Aroch
- Department of Small Animal Internal Medicine, Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 76100, Israel
| | - S Kuzi
- Department of Small Animal Internal Medicine, Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 76100, Israel
| | - I Srugo
- Department of Neurology and Neurosurgery, Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 76100, Israel
| | - T Bdolah-Abram
- Teaching Services Unit, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9190501, Israel
| | - O Chai
- Department of Neurology and Neurosurgery, Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 76100, Israel
| | - M M Christopher
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| | - Y Merbl
- Department of Neurology and Neurosurgery, Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 76100, Israel
| | - K Rothwell
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 76100, Israel
| | - M H Shamir
- Department of Neurology and Neurosurgery, Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 76100, Israel.
| |
Collapse
|
128
|
Siddiqui AM, Khazaei M, Fehlings MG. Translating mechanisms of neuroprotection, regeneration, and repair to treatment of spinal cord injury. PROGRESS IN BRAIN RESEARCH 2015; 218:15-54. [PMID: 25890131 DOI: 10.1016/bs.pbr.2014.12.007] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
One of the big challenges in neuroscience that remains to be understood is why the central nervous system is not able to regenerate to the extent that the peripheral nervous system does. This is especially problematic after traumatic injuries, like spinal cord injury (SCI), since the lack of regeneration leads to lifelong deficits and paralysis. Treatment of SCI has improved during the last several decades due to standardized protocols for emergency medical response teams and improved medical, surgical, and rehabilitative treatments. However, SCI continues to result in profound impairments for the individual. There are many processes that lead to the pathophysiology of SCI, such as ischemia, vascular disruption, neuroinflammation, oxidative stress, excitotoxicity, demyelination, and cell death. Current treatments include surgical decompression, hemodynamic control, and methylprednisolone. However, these early treatments are associated with modest functional recovery. Some treatments currently being investigated for use in SCI target neuroprotective (riluzole, minocycline, G-CSF, FGF-2, and polyethylene glycol) or neuroregenerative (chondroitinase ABC, self-assembling peptides, and rho inhibition) strategies, while many cell therapies (embryonic stem cells, neural stem cells, induced pluripotent stem cells, mesenchymal stromal cells, Schwann cells, olfactory ensheathing cells, and macrophages) have also shown promise. However, since SCI has multiple factors that determine the progress of the injury, a combinatorial therapeutic approach will most likely be required for the most effective treatment of SCI.
Collapse
Affiliation(s)
- Ahad M Siddiqui
- Department of Genetics and Development, Toronto Western Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Mohamad Khazaei
- Department of Genetics and Development, Toronto Western Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Michael G Fehlings
- Department of Genetics and Development, Toronto Western Research Institute, University Health Network, Toronto, Ontario, Canada; Department of Surgery, University of Toronto, Toronto, Ontario, Canada; Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
129
|
Improved fracture healing in patients with concomitant traumatic brain injury: proven or not? Mediators Inflamm 2015; 2015:204842. [PMID: 25873754 PMCID: PMC4385630 DOI: 10.1155/2015/204842] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 01/19/2015] [Indexed: 01/08/2023] Open
Abstract
Over the last 3 decades, scientific evidence advocates an association between traumatic brain injury (TBI) and accelerated fracture healing. Multiple clinical and preclinical studies have shown an enhanced callus formation and an increased callus volume in patients, respectively, rats with concomitant TBI. Over time, different substances (cytokines, hormones, etc.) were in focus to elucidate the relationship between TBI and fracture healing. Until now, the mechanism behind this relationship is not fully clarified and a consensus on which substance plays the key role could not be attained in the literature. In this review, we will give an overview of current concepts and opinions on this topic published in the last decade and both clinical and pathophysiological theories will be discussed.
Collapse
|
130
|
Aksoy H, Sancar M, Sen A, Okuyan B, Bitis L, Uras F, Akakin D, Cevik O, Kultur S, İzzettin FV. The effect of topical ethanol extract ofCotinus coggygriaScop. on cutaneous wound healing in rats. Nat Prod Res 2015; 30:452-5. [DOI: 10.1080/14786419.2015.1019349] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
131
|
Honokiol downregulates Kruppel-like factor 4 expression, attenuates inflammation, and reduces histopathology after spinal cord injury in rats. Spine (Phila Pa 1976) 2015; 40:363-8. [PMID: 25774462 DOI: 10.1097/brs.0000000000000758] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
STUDY DESIGN Randomized experimental study. OBJECTIVE To investigate the neuroprotective effect of honokiol (HNK) on rats subjected to traumatic spinal cord injury (SCI) and the molecular mechanisms. SUMMARY OF BACKGROUND DATA Inflammation contributes to the secondary injury to the spinal cord. Honokiol has been used as a neuroprotective agent because of its strong antioxidant and anti-inflammatory properties. Kruppel-like factor 4 (Klf4) is a newly identified critical target for the anti-inflammatory effect of HNK. Whether HNK can inhibit inflammatory response in rat model of SCI through mediating the expression of Klf4 has yet to be elucidated. METHODS Eighty-four adult female Sprague-Dawley rats were randomly divided into 4 groups as sham, SCI, SCI + Vehicle (0.1% propylene glycol in saline, intraperitoneally), and SCI + HNK (20 mg/kg, intraperitoneally) groups. The influences of HNK on the proinflammatory cytokines, microglial activation, neutrophil infiltration, histological changes, and improvement in motor function were assessed. RESULTS In the SCI group, proinflammatory cytokines, microglial activation, neutrophil infiltration, and Klf4 expression levels were increased compared with the sham group (P < 0.001). HNK intervention downregulated the expression of Klf4, reduced the production of proinflammatory cytokines, inhibited microglial activation, and neutrophil infiltration (P < 0.05). Furthermore, HNK also reduced histopathology and improved functional outcome after traumatic SCI. CONCLUSION HNK reduces secondary tissue damage and improves locomotor function recovery after SCI through suppressing inflammatory response, and can be used as a potential therapeutic agent for SCI. LEVEL OF EVIDENCE NA.
Collapse
|
132
|
Börcek AÖ, Çivi S, Öcal Ö, Gülbahar Ö. Effects of tumor necrosis factor alpha blocker adalimumab in experimental spinal cord injury. J Korean Neurosurg Soc 2015; 57:73-6. [PMID: 25733985 PMCID: PMC4345196 DOI: 10.3340/jkns.2015.57.2.73] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Revised: 08/04/2014] [Accepted: 08/24/2014] [Indexed: 11/27/2022] Open
Abstract
Objective Tumor necrosis factor alpha (TNF-α) have proven effects in pathogenesis of neuroinflammation after spinal cord injury (SCI). Current study is designed to evaluate the effects of an anti-TNF-α agent, adalimumab, on spinal cord clip compression injury in rats. Methods Thirty two male adult Wistar rats were divided into four groups (sham, trauma, infliximab, and adalimumab groups) and SCI was introduced using an aneurysm clip. Animals in treatment groups received 5 mg/kg subcutaneous adalimumab and infliximab right after the trauma. Malondialdehyde (MDA) levels were studied in traumatized spinal cord tissues 72 hours after the injury as a marker of lipid peroxidation. Results Animals that received anti-TNF-α agents are found to have significantly decreased MDA levels. MDA levels were significantly different between the trauma and infliximab groups (p<0.01) and trauma and adalimumab groups (p=0.022). There was no significant difference in neurological evaluation of the rats using Tarlov scale. Conclusion These results suggest that, like infliximab, adalimumab has favorable effects on lipid peroxidation induced by spinal cord trauma in rats.
Collapse
Affiliation(s)
- Alp Özgün Börcek
- Department of Neurosurgery, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Soner Çivi
- Department of Neurosurgery, Medicana Private Hospital, Konya, Turkey
| | - Özgür Öcal
- Department of Neurosurgery, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Özlem Gülbahar
- Department of Biochemistry, Gazi University Faculty of Medicine, Ankara, Turkey
| |
Collapse
|
133
|
Liu NK, Xu XM. Neuroprotection and its molecular mechanism following spinal cord injury. Neural Regen Res 2015; 7:2051-62. [PMID: 25624837 PMCID: PMC4296426 DOI: 10.3969/j.issn.1673-5374.2012.26.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Accepted: 07/10/2012] [Indexed: 12/27/2022] Open
Abstract
Acute spinal cord injury initiates a complex cascade of molecular events termed ‘secondary injury’, which leads to progressive degeneration ranging from early neuronal apoptosis at the lesion site to delayed degeneration of intact white matter tracts, and, ultimately, expansion of the initial injury. These secondary injury processes include, but are not limited to, inflammation, free radical-induced cell death, glutamate excitotoxicity, phospholipase A2 activation, and induction of extrinsic and intrinsic apoptotic pathways, which are important targets in developing neuroprotective strategies for treatment of spinal cord injury. Recently, a number of studies have shown promising results on neuroprotection and recovery of function in rodent models of spinal cord injury using treatments that target secondary injury processes including inflammation, phospholipase A2 activation, and manipulation of the PTEN-Akt/mTOR signaling pathway. The present review outlines our ongoing research on the molecular mechanisms of neuroprotection in experimental spinal cord injury and briefly summarizes our earlier findings on the therapeutic potential of pharmacological treatments in spinal cord injury.
Collapse
Affiliation(s)
- Nai-Kui Liu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery & Goodman Campbell Brain and Spine, Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Xiao-Ming Xu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery & Goodman Campbell Brain and Spine, Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
134
|
Schroeder J, Kueper J, Leon K, Liebergall M. Stem cells for spine surgery. World J Stem Cells 2015; 7:186-194. [PMID: 25621119 PMCID: PMC4300930 DOI: 10.4252/wjsc.v7.i1.186] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Revised: 10/08/2014] [Accepted: 10/29/2014] [Indexed: 02/06/2023] Open
Abstract
In the past few years, stem cells have become the focus of research by regenerative medicine professionals and tissue engineers. Embryonic stem cells, although capable of differentiating into cell lineages of all three germ layers, are limited in their utilization due to ethical issues. In contrast, the autologous harvest and subsequent transplantation of adult stem cells from bone marrow, adipose tissue or blood have been experimentally utilized in the treatment of a wide variety of diseases ranging from myocardial infarction to Alzheimer’s disease. The physiologic consequences of stem cell transplantation and its impact on functional recovery have been studied in countless animal models and select clinical trials. Unfortunately, the bench to bedside translation of this research has been slow. Nonetheless, stem cell therapy has received the attention of spinal surgeons due to its potential benefits in the treatment of neural damage, muscle trauma, disk degeneration and its potential contribution to bone fusion.
Collapse
|
135
|
Lee JY, Choi HY, Na WH, Ju BG, Yune TY. Ghrelin inhibits BSCB disruption/hemorrhage by attenuating MMP-9 and SUR1/TrpM4 expression and activation after spinal cord injury. Biochim Biophys Acta Mol Basis Dis 2014; 1842:2403-12. [DOI: 10.1016/j.bbadis.2014.09.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 09/06/2014] [Accepted: 09/17/2014] [Indexed: 12/11/2022]
|
136
|
Zhou X, He X, Ren Y. Function of microglia and macrophages in secondary damage after spinal cord injury. Neural Regen Res 2014; 9:1787-95. [PMID: 25422640 PMCID: PMC4239768 DOI: 10.4103/1673-5374.143423] [Citation(s) in RCA: 205] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/20/2014] [Indexed: 01/10/2023] Open
Abstract
Spinal cord injury (SCI) is a devastating type of neurological trauma with limited therapeutic opportunities. The pathophysiology of SCI involves primary and secondary mechanisms of injury. Among all the secondary injury mechanisms, the inflammatory response is the major contributor and results in expansion of the lesion and further loss of neurologic function. Meanwhile, the inflammation directly and indirectly dominates the outcomes of SCI, including not only pain and motor dysfunction, but also preventingneuronal regeneration. Microglia and macrophages play very important roles in secondary injury. Microglia reside in spinal parenchyma and survey the microenvironment through the signals of injury or infection. Macrophages are derived from monocytes recruited to injured sites from the peripheral circulation. Activated resident microglia and monocyte-derived macrophages induce and magnify immune and inflammatory responses not only by means of their secretory moleculesand phagocytosis, but also through their influence on astrocytes, oligodendrocytes and demyelination. In this review, we focus on the roles of microglia and macrophages in secondary injury and how they contribute to the sequelae of SCI.
Collapse
Affiliation(s)
- Xiang Zhou
- Department of Orthopedic Surgery, the Second Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Xijing He
- Department of Orthopedic Surgery, the Second Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Yi Ren
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL, USA
| |
Collapse
|
137
|
Hooshmand MJ, Galvan MD, Partida E, Anderson AJ. Characterization of recovery, repair, and inflammatory processes following contusion spinal cord injury in old female rats: is age a limitation? IMMUNITY & AGEING 2014; 11:15. [PMID: 25512759 PMCID: PMC4265993 DOI: 10.1186/1742-4933-11-15] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Accepted: 10/12/2014] [Indexed: 01/14/2023]
Abstract
Background Although the incidence of spinal cord injury (SCI) is steadily rising in the elderly human population, few studies have investigated the effect of age in rodent models. Here, we investigated the effect of age in female rats on spontaneous recovery and repair after SCI. Young (3 months) and aged (18 months) female rats received a moderate contusion SCI at T9. Behavioral recovery was assessed, and immunohistocemical and stereological analyses performed. Results Aged rats demonstrated greater locomotor deficits compared to young, beginning at 7 days post-injury (dpi) and lasting through at least 28 dpi. Unbiased stereological analyses revealed a selective increase in percent lesion area and early (2 dpi) apoptotic cell death caudal to the injury epicenter in aged versus young rats. One potential mechanism for these differences in lesion pathogenesis is the inflammatory response; we therefore assessed humoral and cellular innate immune responses. No differences in either acute or chronic serum complement activity, or acute neutrophil infiltration, were observed between age groups. However, the number of microglia/macrophages present at the injury epicenter was increased by 50% in aged animals versus young. Conclusions These data suggest that age affects recovery of locomotor function, lesion pathology, and microglia/macrophage response following SCI.
Collapse
Affiliation(s)
- Mitra J Hooshmand
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine, 2001 Sue and Bill Gross Stem Cell Research, Irvine, CA 92697-4292, USA.,Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA 92697, USA
| | - Manuel D Galvan
- Reeve-Irvine Research Center, University of California Irvine, Irvine, CA 92697, USA.,Anatomy and Neurobiology, University of California Irvine, Irvine, CA 92697, USA
| | - Elizabeth Partida
- Reeve-Irvine Research Center, University of California Irvine, Irvine, CA 92697, USA
| | - Aileen J Anderson
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine, 2001 Sue and Bill Gross Stem Cell Research, Irvine, CA 92697-4292, USA.,Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA 92697, USA
| |
Collapse
|
138
|
Lee JY, Choi HY, Ahn HJ, Ju BG, Yune TY. Matrix metalloproteinase-3 promotes early blood-spinal cord barrier disruption and hemorrhage and impairs long-term neurological recovery after spinal cord injury. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:2985-3000. [PMID: 25325922 DOI: 10.1016/j.ajpath.2014.07.016] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 06/21/2014] [Accepted: 07/15/2014] [Indexed: 11/16/2022]
Abstract
After spinal cord injury (SCI), blood-spinal cord barrier (BSCB) disruption by matrix metalloproteinases (MMPs) leads to BSCB permeability and blood cell infiltration, contributing to permanent neurological disability. Herein, we report that MMP-3 plays a critical role in BSCB disruption after SCI in mice. MMP-3 was induced in infiltrated neutrophils and blood vessels after SCI, and NF-κB as a transcription factor was involved in MMP-3 expression. BSCB permeability and blood cell infiltration after injury were more reduced in Mmp3 knockout (KO) mice than in wild-type (WT) mice, which was significantly inhibited by Mmp3 siRNA or a general inhibitor of MMPs, N-isobutyl-N-(4-methoxyphenylsulfonyl)glycyl hydroxamic acid. The level of tight junction proteins, such as occludin and zonula occludens-1, which decreased after SCI, was also higher in Mmp3 KO than in WT mice. Exogenously, MMP-3 injection into the normal spinal cord also induced BSCB permeability. Furthermore, MMP-9 activation after injury was mediated by MMP-3 activation. Finally, improved functional recovery was observed in Mmp3 KO mice compared with WT mice after injury. These results demonstrated the role of MMP-3 in BSCB disruption after SCI for the first time and suggest that the regulation of MMP-3 can be considered a therapeutic target to inhibit BSCB disruption and hemorrhage, and thereby enhance functional recovery after acute SCI.
Collapse
Affiliation(s)
- Jee Youn Lee
- Age-Related and Brain Diseases Research Center, School of Medicine, Kyung Hee University, Seoul, Republic of Korea; Neurodegeneration Control Research Center, School of Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Hae Young Choi
- Age-Related and Brain Diseases Research Center, School of Medicine, Kyung Hee University, Seoul, Republic of Korea; Neurodegeneration Control Research Center, School of Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Hyun-Jong Ahn
- Department of Microbiology, School of Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Bong Gun Ju
- Department of Life Science, Sogang University, Seoul, Republic of Korea
| | - Tae Young Yune
- Age-Related and Brain Diseases Research Center, School of Medicine, Kyung Hee University, Seoul, Republic of Korea; Neurodegeneration Control Research Center, School of Medicine, Kyung Hee University, Seoul, Republic of Korea; Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul, Republic of Korea.
| |
Collapse
|
139
|
WONG JK, ZOU H. Reshaping the chromatin landscape after spinal cord injury. FRONTIERS IN BIOLOGY 2014; 9:356-366. [PMID: 25554728 PMCID: PMC4280023 DOI: 10.1007/s11515-014-1329-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The pathophysiology underlying spinal cord injury is complex. Mechanistic understanding of the adaptive responses to injury is critical for targeted therapy aimed at reestablishing lost connections between proximal and distal neurons. After injury, cell-type specific gene transcription programs govern distinct cellular behaviors, and chromatin regulators play a central role in shaping the chromatin landscape to adjust transcriptional profiles in a context-dependent manner. In this review, we summarize recent progress on the pleiotropic roles of chromatin regulators in mediating the diverse adaptive behaviors of neurons and glial cells after spinal cord injury, and wherever possible, discuss the underlying mechanisms and genomic targets. We specifically draw attention to the perspective that takes into consideration the impact of epigenetic modulation on axon growth potential, together with its effect on wound-healing properties of glial cells. Epigenetic modulation of chromatin state represents an emerging therapeutic direction to promote neural repair and axon regeneration after spinal cord injury.
Collapse
Affiliation(s)
- Jamie K. WONG
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Hongyan ZOU
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Neurosurgery, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
140
|
Chakrabarti M, Haque A, Banik NL, Nagarkatti P, Nagarkatti M, Ray SK. Estrogen receptor agonists for attenuation of neuroinflammation and neurodegeneration. Brain Res Bull 2014; 109:22-31. [PMID: 25245209 DOI: 10.1016/j.brainresbull.2014.09.004] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Revised: 09/09/2014] [Accepted: 09/10/2014] [Indexed: 01/05/2023]
Abstract
Recent results from laboratory investigations and clinical trials indicate important roles for estrogen receptor (ER) agonists in protecting the central nervous system (CNS) from noxious consequences of neuroinflammation and neurodegeneration. Neurodegenerative processes in several CNS disorders including spinal cord injury (SCI), multiple sclerosis (MS), Parkinson's disease (PD), and Alzheimer's disease (AD) are associated with activation of microglia and astrocytes, which drive the resident neuroinflammatory response. During neurodegenerative processes, activated microglia and astrocytes cause deleterious effects on surrounding neurons. The inhibitory activity of ER agonists on microglia activation might be a beneficial therapeutic option for delaying the onset or progression of neurodegenerative injuries and diseases. Recent studies suggest that ER agonists can provide neuroprotection by modulation of cell survival mechanisms, synaptic reorganization, regenerative responses to axonal injury, and neurogenesis process. The anti-inflammatory and neuroprotective actions of ER agonists are mediated mainly via two ERs known as ERα and ERβ. Although some studies have suggested that ER agonists may be deleterious to some neuronal populations, the potential clinical benefits of ER agonists for augmenting cognitive function may triumph over the associated side effects. Also, understanding the modulatory activities of ER agonists on inflammatory pathways will possibly lead to the development of selective anti-inflammatory molecules with neuroprotective roles in different CNS disorders such as SCI, MS, PD, and AD in humans. Future studies should be concentrated on finding the most plausible molecular pathways for enhancing protective functions of ER agonists in treating neuroinflammatory and neurodegenerative injuries and diseases in the CNS.
Collapse
Affiliation(s)
- Mrinmay Chakrabarti
- University of South Carolina School of Medicine, Department of Pathology, Microbiology, and Immunology, Columbia, SC 29209, USA
| | - Azizul Haque
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Naren L Banik
- Department of Neurosurgery and Neurology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Prakash Nagarkatti
- University of South Carolina School of Medicine, Department of Pathology, Microbiology, and Immunology, Columbia, SC 29209, USA
| | - Mitzi Nagarkatti
- University of South Carolina School of Medicine, Department of Pathology, Microbiology, and Immunology, Columbia, SC 29209, USA
| | - Swapan K Ray
- University of South Carolina School of Medicine, Department of Pathology, Microbiology, and Immunology, Columbia, SC 29209, USA.
| |
Collapse
|
141
|
Bowes AL, Yip PK. Modulating inflammatory cell responses to spinal cord injury: all in good time. J Neurotrauma 2014; 31:1753-66. [PMID: 24934600 DOI: 10.1089/neu.2014.3429] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Spinal cord injury can have a range of debilitating effects, permanently impacting a patient's quality of life. Initially thought to be an immune privileged site, the spinal cord is able to mount a timely and well organized inflammatory response to injury. Intricate immune cell interactions are triggered, typically consisting of a staggered multiphasic immune cell response, which can become deregulated if left unchecked. Although several immunomodulatory compounds have yielded success in experimental rodent spinal cord injury models, their translation to human clinical studies needs further consideration. Because temporal differences between rodent and human inflammatory responses to spinal cord injury do exist, drug delivery timing will be a crucial component in recovery from spinal cord injury. Given too early, immunomodulatory therapies may impede beneficial inflammatory reactions to the injured spinal cord or even miss the opportunity to dampen delayed harmful autoimmune processes. Therefore, this review aims to summarize the temporal inflammatory response to spinal cord injury, as well as detailing specific immune cell functions. By clearly defining the chronological order of inflammatory events after trauma, immunomodulatory drug delivery timing can be better optimized. Further, we compare spinal cord injury-induced inflammatory responses in rodent and human studies, enabling clinicians to consider these differences when initiating clinical trials. Improved understanding of the cellular immune response after spinal cord injury would enhance the efficacy of immunomodulatory agents, enabling combined therapies to be considered.
Collapse
Affiliation(s)
- Amy L Bowes
- Centre for Neuroscience and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London , London, United Kingdom
| | | |
Collapse
|
142
|
Peterson SL, Anderson AJ. Complement and spinal cord injury: traditional and non-traditional aspects of complement cascade function in the injured spinal cord microenvironment. Exp Neurol 2014; 258:35-47. [PMID: 25017886 DOI: 10.1016/j.expneurol.2014.04.028] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Revised: 04/14/2014] [Accepted: 04/28/2014] [Indexed: 12/21/2022]
Abstract
The pathology associated with spinal cord injury (SCI) is caused not only by primary mechanical trauma, but also by secondary responses of the injured CNS. The inflammatory response to SCI is robust and plays an important but complex role in the progression of many secondary injury-associated pathways. Although recent studies have begun to dissect the beneficial and detrimental roles for inflammatory cells and proteins after SCI, many of these neuroimmune interactions are debated, not well understood, or completely unexplored. In this regard, the complement cascade is a key component of the inflammatory response to SCI, but is largely underappreciated, and our understanding of its diverse interactions and effects in this pathological environment is limited. In this review, we discuss complement in the context of SCI, first in relation to traditional functions for complement cascade activation, and then in relation to novel roles for complement proteins in a variety of models.
Collapse
Affiliation(s)
- Sheri L Peterson
- Sue & Bill Gross Stem Cell Center, University of California, Irvine, Irvine, CA 92697, USA; Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA 92697, USA; Department of Anatomy & Neurobiology, University of California, Irvine, Irvine, CA 92697, USA
| | - Aileen J Anderson
- Sue & Bill Gross Stem Cell Center, University of California, Irvine, Irvine, CA 92697, USA; Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA 92697, USA; Department of Anatomy & Neurobiology, University of California, Irvine, Irvine, CA 92697, USA; Department of Physical Medicine and Rehabilitation, University of California, Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
143
|
Petrosyan HA, Alessi V, Singh V, Hunanyan AS, Levine JM, Arvanian VL. Transduction efficiency of neurons and glial cells by AAV-1, -5, -9, -rh10 and -hu11 serotypes in rat spinal cord following contusion injury. Gene Ther 2014; 21:991-1000. [DOI: 10.1038/gt.2014.74] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 05/09/2014] [Accepted: 07/10/2014] [Indexed: 12/18/2022]
|
144
|
Immune modulatory therapies for spinal cord injury – Past, present and future. Exp Neurol 2014; 258:91-104. [DOI: 10.1016/j.expneurol.2014.01.025] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2013] [Revised: 01/21/2014] [Accepted: 01/30/2014] [Indexed: 01/18/2023]
|
145
|
Bloom O. Non-mammalian model systems for studying neuro-immune interactions after spinal cord injury. Exp Neurol 2014; 258:130-40. [PMID: 25017894 PMCID: PMC4099969 DOI: 10.1016/j.expneurol.2013.12.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Revised: 12/24/2013] [Accepted: 12/26/2013] [Indexed: 01/09/2023]
Abstract
Mammals exhibit poor recovery after injury to the spinal cord, where the loss of neurons and neuronal connections can be functionally devastating. In contrast, it has long been appreciated that many non-mammalian vertebrate species exhibit significant spontaneous functional recovery after spinal cord injury (SCI). Identifying the biological responses that support an organism's inability or ability to recover function after SCI is an important scientific and medical question. While recent advances have been made in understanding the responses to SCI in mammals, we remain without an effective clinical therapy for SCI. A comparative biological approach to understanding responses to SCI in non-mammalian vertebrates will yield important insights into mechanisms that promote recovery after SCI. Presently, mechanistic studies aimed at elucidating responses, both intrinsic and extrinsic to neurons, that result in different regenerative capacities after SCI across vertebrates are just in their early stages. There are several inhibitory mechanisms proposed to impede recovery from SCI in mammals, including reactive gliosis and scarring, myelin associated proteins, and a suboptimal immune response. One hypothesis to explain the robust regenerative capacity of several non-mammalian vertebrates is a lack of some or all of these inhibitory signals. This review presents the current knowledge of immune responses to SCI in several non-mammalian species that achieve anatomical and functional recovery after SCI. This subject is of growing interest, as studies increasingly show both beneficial and detrimental roles of the immune response following SCI in mammals. A long-term goal of biomedical research in all experimental models of SCI is to understand how to promote functional recovery after SCI in humans. Therefore, understanding immune responses to SCI in non-mammalian vertebrates that achieve functional recovery spontaneously may identify novel strategies to modulate immune responses in less regenerative species and promote recovery after SCI.
Collapse
Affiliation(s)
- Ona Bloom
- The Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, NY 11030, USA; The Hofstra North Shore-LIJ School of Medicine, Hempstead Turnpike, Hempstead, NY 11549, USA.
| |
Collapse
|
146
|
Fu R, Shen Q, Xu P, Luo JJ, Tang Y. Phagocytosis of microglia in the central nervous system diseases. Mol Neurobiol 2014; 49:1422-34. [PMID: 24395130 PMCID: PMC4012154 DOI: 10.1007/s12035-013-8620-6] [Citation(s) in RCA: 440] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 12/15/2013] [Indexed: 12/20/2022]
Abstract
Microglia, the resident macrophages of the central nervous system, rapidly activate in nearly all kinds of neurological diseases. These activated microglia become highly motile, secreting inflammatory cytokines, migrating to the lesion area, and phagocytosing cell debris or damaged neurons. During the past decades, the secretory property and chemotaxis of microglia have been well-studied, while relatively less attention has been paid to microglial phagocytosis. So far there is no obvious concordance with whether it is beneficial or detrimental in tissue repair. This review focuses on phagocytic phenotype of microglia in neurological diseases such as Alzheimer's disease, multiple sclerosis, Parkinson's disease, traumatic brain injury, ischemic and other brain diseases. Microglial morphological characteristics, involved receptors and signaling pathways, distribution variation along with time and space changes, and environmental factors that affecting phagocytic function in each disease are reviewed. Moreover, a comparison of contributions between macrophages from peripheral circulation and the resident microglia to these pathogenic processes will also be discussed.
Collapse
Affiliation(s)
- Ruying Fu
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Number 107, Yan Jiang Xi Road, Guangzhou, 510120 Guangdong Province China
| | - Qingyu Shen
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Number 107, Yan Jiang Xi Road, Guangzhou, 510120 Guangdong Province China
- Department of Neurology, Zengcheng People’s Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Pengfei Xu
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Number 107, Yan Jiang Xi Road, Guangzhou, 510120 Guangdong Province China
| | - Jin Jun Luo
- Department of Neurology, School of Medicine, Temple University, Philadelphia, PA USA
| | - Yamei Tang
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Number 107, Yan Jiang Xi Road, Guangzhou, 510120 Guangdong Province China
- Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
147
|
|
148
|
Affiliation(s)
- Phillip G Popovich
- Center for Brain and Spinal Cord Repair, Department of Neuroscience, Wexner Medical Center at The Ohio State University, USA
| |
Collapse
|
149
|
Song Y, Li S, Song B, Zhang Y, Gao W, Li N, Fan K, Ma J. The pathological changes in the spinal cord after dural tear with and without autologous fascia repair. EUROPEAN SPINE JOURNAL : OFFICIAL PUBLICATION OF THE EUROPEAN SPINE SOCIETY, THE EUROPEAN SPINAL DEFORMITY SOCIETY, AND THE EUROPEAN SECTION OF THE CERVICAL SPINE RESEARCH SOCIETY 2014; 23:1531-40. [PMID: 24801575 DOI: 10.1007/s00586-014-3326-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Revised: 04/17/2014] [Accepted: 04/18/2014] [Indexed: 10/25/2022]
Abstract
PURPOSE Dural tear is one of the common complications of spinal surgery leading to cerebrospinal fluid leakage followed by serial secondary symptoms. However, little is known about pathological changes of the spinal cord after dural tear. In the present study, we aimed to study the pathological changes in the spinal cord after dural tear with and without autologous fascia repair. METHODS Sixty Sprague-Dawley rats were used for dural tear and autologous fascia graft repair models. Three days and 1 week after surgery, the pathological changes in the spinal cord were analyzed by immunohistochemistry, Western blot, enzyme-linked immunosorbent assay and spinal somatosensory evoked potentials test. RESULTS Neuroinflammation was found in the parenchyma of the spinal cord characterized by gliosis, increased expression of inflammatory factors and infiltration of exogenesis immunocells in the rats without repair, which impaired the sensory conduction function of the spinal cord at the early stage of injury. Repairing with autologous fascia could attenuate neuroinflammation and help to maintain normal sensory conduction function of the spinal cord. CONCLUSION Dural tear could cause a series of inflammatory reactions in the spinal cord and further impair its sensory conduction function at the early stage of injury. Repairing with autologous fascia was a necessary and effective way to prevent the neuroinflammation and to maintain the normal function of the spinal cord.
Collapse
Affiliation(s)
- Yi Song
- Graduate School of Dalian Medical University, Dalian, 116044, Liaoning, China,
| | | | | | | | | | | | | | | |
Collapse
|
150
|
Raposo C, Schwartz M. Glial scar and immune cell involvement in tissue remodeling and repair following acute CNS injuries. Glia 2014; 62:1895-904. [PMID: 24756949 DOI: 10.1002/glia.22676] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Revised: 03/30/2014] [Accepted: 04/01/2014] [Indexed: 12/17/2022]
Abstract
Inadequate axonal regeneration is a common phenomenon occurring following acute injury to the central nervous system (CNS), and is often associated with permanent neurological deficits. The injured axons attempting to regenerate face the inhospitable environment of the CNS scar, which can hinder axonal growth and sprouting. In addition, in response to the insult, intense activation and infiltration of immune cells take place. Both the scar tissue and immune response, which have received a bad reputation in the context of CNS repair are essential for the overall recovery from CNS injuries, but are not optimally controlled. The glial scar contributes to protection of the spared neural tissues by establishing a boundary between damaged and salvageable tissue, and by educating the immune cells to promote the healing of the CNS tissue. In turn, the immune cells, and in particular the infiltrating macrophages, exert several functions at the lesion site, including resolution of the microglial response, control of scar tissue degradation, and production of growth factors; thereby, promoting neuronal survival, axonal regeneration, and tissue remodeling. As axonal regeneration and tissue remodeling are viewed as critical steps for the overall functional recovery following CNS injury, a detailed understanding of the mechanisms underlying the timely formation and degradation of the CNS scar, and its crosstalk with the inflammatory response, are of great importance, both biologically and clinically.
Collapse
Affiliation(s)
- Catarina Raposo
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | | |
Collapse
|