1
|
Yu CG, Bondada V, Joshi A, Reneer DV, Telling GC, Saatman KE, Geddes JW. Calpastatin Overexpression Protects against Excitotoxic Hippocampal Injury and Traumatic Spinal Cord Injury. J Neurotrauma 2020; 37:2268-2276. [PMID: 32718209 DOI: 10.1089/neu.2020.7122] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Small molecule inhibitors of calcium-dependent proteases, calpains (CAPNs), protect against neurodegeneration induced by a variety of insults including excitotoxicity and spinal cord injury (SCI). Many of these compounds, however, also inhibit other proteases, which has made it difficult to evaluate the contribution of calpains to neurodegeneration. Calpastatin is a highly specific endogenous inhibitor of classical calpains, including CAPN1 and CAPN2. In the present study, we utilized transgenic mice that overexpress human calpastatin under the prion promoter (PrP-hCAST) to evaluate the hypothesis that calpastatin overexpression protects against excitotoxic hippocampal injury and contusive SCI. The PrP-hCAST organotypic hippocampal slice cultures showed reduced neuronal death and reduced calpain-dependent proteolysis (α-spectrin breakdown production, 145 kDa) at 24 h after N-methyl-D-aspartate (NMDA) injury compared with the wild-type (WT) cultures (n = 5, p < 0.05). The PrP-hCAST mice (n = 13) displayed a significant improvement in locomotor function at one and three weeks after contusive SCI compared with the WT controls (n = 9, p < 0.05). Histological assessment of lesion volume and tissue sparing, performed on the same animals used for behavioral analysis, revealed that calpastatin overexpression resulted in a 30% decrease in lesion volume (p < 0.05) and significant increases in tissue sparing, white matter sparing, and gray matter sparing at four weeks post-injury compared with WT animals. Calpastatin overexpression reduced α-spectrin breakdown by 51% at 24 h post-injury, compared with WT controls (p < 0.05, n = 3/group). These results provide support for the hypothesis that sustained calpain-dependent proteolysis contributes to pathological deficits after excitotoxic injury and traumatic SCI.
Collapse
Affiliation(s)
- Chen Guang Yu
- Department of Neuroscience and Spinal Cord and Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - Vimala Bondada
- Department of Neuroscience and Spinal Cord and Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - Aashish Joshi
- Spinal Cord and Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - Dexter V Reneer
- Spinal Cord and Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - Glenn C Telling
- Department of Microbiology, Immunology & Pathology, Colorado State University College of Veterinary Medicine and Biomedical Science, Fort Collins, Colorado, USA
| | - Kathryn E Saatman
- Department of Physiology, Spinal Cord and Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - James W Geddes
- Department of Neuroscience and Spinal Cord and Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| |
Collapse
|
2
|
Yu CG, Bondada V, Ghoshal S, Singh R, Pistilli CK, Dayaram K, Iqbal H, Sands M, Davis KL, Bondada S, Geddes JW. Repositioning Flubendazole for Spinal Cord Injury. J Neurotrauma 2019; 36:2618-2630. [PMID: 30747048 DOI: 10.1089/neu.2018.6160] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
We previously reported the serendipitous observation that fenbendazole, a benzimidazole anthelmintic, improved functional and pathological outcomes following thoracic spinal cord contusion injury in mice when administered pre-injury. Fenbendazole is widely used in veterinary medicine. However, it is not approved for human use and it was uncertain if only post-injury administration would offer similar benefits. In the present study we evaluated post-injury administration of a closely related, human anthelmintic drug, flubendazole, using a rat spinal cord contusion injury model. Flubendazole, administered i.p. 5 or 10 mg/kg day, beginning 3 h post-injury and daily thereafter for 2 or 4 weeks, resulted in improved locomotor function after contusion spinal cord injury (SCI) compared with vehicle-treated controls. Histological analysis of spinal cord sections showed that such treatment with flubendazole also reduced lesion volume and improved total tissue sparing, white matter sparing, and gray matter sparing. Flubendazole inhibited the activation of glial fibrillary acidic protein (GFAP); suppressed cyclin B1 expression and Bruton tyrosine kinase activation, markers of B cell activation/proliferation and inflammation; and reduced B cell autoimmune response. Together, these results suggest the use of the benzimidazole anthelmintic flubendazole as a potential therapeutic for SCI.
Collapse
Affiliation(s)
- Chen Guang Yu
- Spinal Cord and Brain Injury Research Center, Department of Neuroscience, University of Kentucky College of Medicine, Lexington, Kentucky
| | - Vimala Bondada
- Spinal Cord and Brain Injury Research Center, Department of Neuroscience, University of Kentucky College of Medicine, Lexington, Kentucky
| | - Sarbani Ghoshal
- Spinal Cord and Brain Injury Research Center, Department of Neuroscience, University of Kentucky College of Medicine, Lexington, Kentucky
| | - Ranjana Singh
- Spinal Cord and Brain Injury Research Center, Department of Neuroscience, University of Kentucky College of Medicine, Lexington, Kentucky
| | - Christina K Pistilli
- Spinal Cord and Brain Injury Research Center, Department of Neuroscience, University of Kentucky College of Medicine, Lexington, Kentucky
| | - Kavi Dayaram
- Spinal Cord and Brain Injury Research Center, Department of Neuroscience, University of Kentucky College of Medicine, Lexington, Kentucky
| | - Hina Iqbal
- Spinal Cord and Brain Injury Research Center, Department of Neuroscience, University of Kentucky College of Medicine, Lexington, Kentucky
| | - Madison Sands
- Spinal Cord and Brain Injury Research Center, Department of Neuroscience, University of Kentucky College of Medicine, Lexington, Kentucky
| | - Kate L Davis
- Spinal Cord and Brain Injury Research Center, Department of Neuroscience, University of Kentucky College of Medicine, Lexington, Kentucky
| | - Subarrao Bondada
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky College of Medicine, Lexington, Kentucky
| | - James W Geddes
- Spinal Cord and Brain Injury Research Center, Department of Neuroscience, University of Kentucky College of Medicine, Lexington, Kentucky
| |
Collapse
|
3
|
Hu J, Chen L, Huang X, Wu K, Ding S, Wang W, Wang B, Smith C, Ren C, Ni H, ZhuGe Q, Yang J. Calpain inhibitor MDL28170 improves the transplantation-mediated therapeutic effect of bone marrow-derived mesenchymal stem cells following traumatic brain injury. Stem Cell Res Ther 2019; 10:96. [PMID: 30876457 PMCID: PMC6420775 DOI: 10.1186/s13287-019-1210-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 01/16/2019] [Accepted: 03/06/2019] [Indexed: 01/14/2023] Open
Abstract
Background Studies have shown that transplantation of bone marrow-derived mesenchymal stem cells (BMSCs) protects against brain damage. However, the low survival number of transplanted BMSCs remains a pertinent challenge and can be attributed to the unfavorable microenvironment of the injured brain. It is well known that calpain activation plays a critical role in traumatic brain injury (TBI)-mediated inflammation and cell death; previous studies showed that inhibiting calpain activation is neuroprotective after TBI. Thus, we investigated whether preconditioning with the calpain inhibitor, MDL28170, could enhance the survival of BMSCs transplanted at 24 h post TBI to improve neurological function. Methods TBI rat model was induced by the weight-drop method, using the gravitational forces of a free falling weight to produce a focal brain injury. MDL28170 was injected intracranially at the lesion site at 30 min post TBI, and the secretion levels of neuroinflammatory factors were assessed 24 h later. BMSCs labeled with green fluorescent protein (GFP) were locally administrated into the lesion site of TBI rat brains at 24 h post TBI. Immunofluorescence and histopathology were performed to evaluate the BMSC survival and the TBI lesion volume. Modified neurological severity scores were chosen to evaluate the functional recovery. The potential mechanisms by which MDL28170 is involved in the regulation of inflammation signaling pathway and cell apoptosis were determined by western blot and immunofluorescence staining. Results Overall, we found that a single dose of MDL28170 at acute phase of TBI improved the microenvironment by inhibiting the inflammation, facilitated the survival of grafted GFP-BMSCs, and reduced the grafted cell apoptosis, leading to the reduction of lesion cavity. Furthermore, a significant neurological function improvement was observed when BMSCs were transplanted into a MDL28170-preconditioned TBI brains compared with the one without MDL28170-precondition group. Conclusions Taken together, our data suggest that MDL28170 improves BMSC transplantation microenvironment and enhances the neurological function restoration after TBI via increased survival rate of BMSCs. We suggest that the calpain inhibitor, MDL28170, could be pursued as a new combination therapeutic strategy to advance the effects of transplanted BMSCs in cell-based regenerative medicine. Electronic supplementary material The online version of this article (10.1186/s13287-019-1210-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jiangnan Hu
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China. .,Department of Pharmaceutical Sciences, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA.
| | - Lefu Chen
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Xujun Huang
- Department of Intensive Care Unit (ICU), Hengdian Wenrong Hospital, Jinhua, 322100, China
| | - Ke Wu
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Saidan Ding
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Weikan Wang
- Department of Pharmaceutical Sciences, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
| | - Brian Wang
- Department of Pharmaceutical Sciences, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
| | - Charity Smith
- Department of Pharmaceutical Sciences, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
| | - Changhong Ren
- Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Haoqi Ni
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Qichuan ZhuGe
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| | - Jianjing Yang
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| |
Collapse
|
4
|
Shi D, He T, Tang W, Li H, Wang C, Zheng M, Hu J, Song X, Ding Y, Chen YY, Shen Y, Jin H, Wang LL. Local application of MDL28170-loaded PCL film improves functional recovery by preserving survival of motor neurons after traumatic spinal cord injury. Neurosci Lett 2019; 694:161-167. [PMID: 30528875 DOI: 10.1016/j.neulet.2018.12.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Revised: 11/30/2018] [Accepted: 12/03/2018] [Indexed: 11/27/2022]
Abstract
Neuronal death and organization degeneration can happen inordinately after spinal cord injury (SCI), which lead to nerve dysfunction. We aimed to determine whether local application of a cell permeable calpain I inhibitor (MDL28170) can promote SCI recovery by increasing neuronal cell viability. MDL28170-loaded polycaprolactone (PCL) film was fabricated. Scanning electron microscopy showed the surface of PCL film was smooth with holes (diameter at μM level). The PCL film was non-toxic, biological compatibility, and had good neuron adhension and slow release characteristic. MDL28170 increased VSC4.1 motor neurons' viability under tunicamycin (an endoplasmic reticulum stress) induced injury. In a traumatic SCI rat model, MDL28170-loaded PCL film reduced the area of lesion cavity, and promoted recovery of locomotor behavior. Moreover, the expression of GAP-43 was upregulated after MDL28170-loaded PCL film treatment. Thus, our findings demonstrated that localized delivery of MDL28170 could promote SCI recovery by inhibiting endoplasmic reticulum stress, preserving survival of the motor neurons, which may point out a promising therapeutic target for treating SCI patient.
Collapse
Affiliation(s)
- Dongling Shi
- Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311400, China
| | - Teng He
- Department of Basic Medicine Sciences, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Weijian Tang
- Department of Basic Medicine Sciences, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Heyangzi Li
- Department of Basic Medicine Sciences, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Chao Wang
- Department of Basic Medicine Sciences, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Mingzhi Zheng
- Department of Pharmacology, Hangzhou Medical College, Hangzhou, 310053, China
| | - Jue Hu
- Department of Pharmacology, Hangzhou Medical College, Hangzhou, 310053, China
| | - Xinghui Song
- Core Facilities, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Yuemin Ding
- Department of Clinical Medicine, School of Medicine, Zhejiang University City College, Hangzhou, 310015, China
| | - Ying-Ying Chen
- Department of Basic Medicine Sciences, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Yueliang Shen
- Department of Basic Medicine Sciences, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Hongfeng Jin
- Department of Cardiology, Zhejiang Hospital, Hangzhou, 310030, China.
| | - Lin-Lin Wang
- Department of Basic Medicine Sciences, School of Medicine, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
5
|
Guest JD, Moore SW, Aimetti AA, Kutikov AB, Santamaria AJ, Hofstetter CP, Ropper AE, Theodore N, Ulich TR, Layer RT. Internal decompression of the acutely contused spinal cord: Differential effects of irrigation only versus biodegradable scaffold implantation. Biomaterials 2018; 185:284-300. [DOI: 10.1016/j.biomaterials.2018.09.025] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 09/04/2018] [Accepted: 09/16/2018] [Indexed: 12/13/2022]
|
6
|
Tamtaji OR, Mirhosseini N, Reiter RJ, Azami A, Asemi Z. Melatonin, a calpain inhibitor in the central nervous system: Current status and future perspectives. J Cell Physiol 2018; 234:1001-1007. [DOI: 10.1002/jcp.27084] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 06/29/2018] [Indexed: 12/22/2022]
Affiliation(s)
- Omid Reza Tamtaji
- Physiology Research Center Kashan University of Medical Sciences Kashan Iran
| | | | - Russel J. Reiter
- Department of Cellular and Structural Biology University of Texas Health Science Center San Antonio Texas
| | - Abolfazl Azami
- Anatomical Sciences Research Center Kashan University of Medical Sciences Kashan Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases Kashan University of Medical Sciences Kashan Iran
| |
Collapse
|
7
|
Narang A, Qiao F, Atkinson C, Zhu H, Yang X, Kulik L, Holers VM, Tomlinson S. Natural IgM antibodies that bind neoepitopes exposed as a result of spinal cord injury , drive secondary injury by activating complement. J Neuroinflammation 2017. [PMID: 28629465 PMCID: PMC5477255 DOI: 10.1186/s12974-017-0894-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Background Natural IgM antibodies (Abs) function as innate immune sensors of injury via recognition of neoepitopes expressed on damaged cells, although how this recognition systems function following spinal cord injury (SCI) exposes various neoepitopes and their precise nature remains largely unknown. Here, we investigated the role of two natural IgM monoclonal Abs (mAbs), B4 and C2, that recognize post-ischemic neoepitopes following ischemia and reperfusion in other tissues. Methods Identification of post-SCI expressed neoepitopes was examined using previously characterized monoclonal Abs (B4 and C2 mAbs). The role of post-SCI neoepitopes and their recognition by natural IgM Abs in propagating secondary injury was examined in Ab-deficient Rag1−/− or wild type C57BL/6 mice using Ab reconstitution experiments and neoepitope-targeted therapeutic studies, respectively. Results Administration of B4 or C2 mAb following murine SCI increased lesion size and worsened functional outcome in otherwise protected Ab-deficient Rag1−/− mice. Injury correlated with colocalized deposition of IgM and C3d in injured spinal cords from both mAb reconstituted Rag1−/− mice and untreated wild-type mice. Depletion of peritoneal B1 B cells, a source of natural Abs, reduced circulating levels of IgM with B4 (annexin-IV) and C2 (subset of phospholipids) reactivity, reduced IgM and complement deposition in the spinal cord, and protected against SCI. We therefore investigated whether the B4 neoepitope represents a therapeutic target for complement inhibition. B4-Crry, a fusion protein consisting of a single-chain Ab derived from B4 mAb, linked to the complement inhibitor Crry, significantly protected against SCI. B4-Crry exhibited a dual function in that it inhibited both the binding of pathogenic IgM and blocked complement activation in the spinal cord. Conclusions This study identifies important neoepitopes expressed within the spinal cord after injury. These neoepitopes are recognized by clonally specific natural IgM Abs that activate complement and drive pathology. We demonstrate that these neoepitopes represent novel targets for the therapeutic delivery of a complement inhibitor, and possibly other payload, to the injured spinal cord. Electronic supplementary material The online version of this article (doi:10.1186/s12974-017-0894-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Aarti Narang
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Ave, CRI 213, Charleston, SC, 29425, USA
| | - Fei Qiao
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Ave, CRI 213, Charleston, SC, 29425, USA
| | - Carl Atkinson
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Ave, CRI 213, Charleston, SC, 29425, USA
| | - Hong Zhu
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Ave, CRI 213, Charleston, SC, 29425, USA
| | - Xiaofeng Yang
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Ave, CRI 213, Charleston, SC, 29425, USA
| | - Liudmila Kulik
- Departments of Medicine and Immunology, University of Colorado School of Medicine, Denver, CO, USA
| | - V Michael Holers
- Departments of Medicine and Immunology, University of Colorado School of Medicine, Denver, CO, USA
| | - Stephen Tomlinson
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Ave, CRI 213, Charleston, SC, 29425, USA. .,Ralph H Johnson VA Medical Center, Charleston, SC, USA.
| |
Collapse
|
8
|
Haque A, Capone M, Matzelle D, Cox A, Banik NL. Targeting Enolase in Reducing Secondary Damage in Acute Spinal Cord Injury in Rats. Neurochem Res 2017; 42:2777-2787. [PMID: 28508172 DOI: 10.1007/s11064-017-2291-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 04/24/2017] [Accepted: 05/06/2017] [Indexed: 12/12/2022]
Abstract
Spinal cord injury (SCI) is a complex debilitating condition leading to permanent life-long neurological deficits. The complexity of SCI suggests that a concerted multi-targeted therapeutic approach is warranted to optimally improve function. Damage to spinal cord is complicated by an increased detrimental response from secondary injury factors mediated by activated glial cells and infiltrating macrophages. While elevation of enolase especially neuron specific enolase (NSE) in glial and neuronal cells is believed to trigger inflammatory cascades in acute SCI, alteration of NSE and its subsequent effects in acute SCI remains unknown. This study measured NSE expression levels and key inflammatory mediators after acute SCI and investigated the role of ENOblock, a novel small molecule inhibitor of enolase, in a male Sprague-Dawley (SD) rat SCI model. Serum NSE levels as well as cytokines/chemokines and metabolic factors were evaluated in injured animals following treatment with vehicle alone or ENOblock using Discovery assay. Spinal cord samples were also analyzed for NSE and MMPs 2 and 9 as well as glial markers by Western blotting. The results indicated a significant decrease in serum inflammatory cytokines/chemokines and NSE, alterations of metabolic factors and expression of MMPs in spinal cord tissues after treatment with ENOblock (100 µg/kg, twice). These results support the hypothesis that activation of glial cells and inflammation status can be modulated by regulation of NSE expression and activity. Analysis of SCI tissue samples by immunohistochemistry confirmed that ENOblock decreased gliosis which may have occurred through reduction of elevated NSE in rats. Overall, elevation of NSE is deleterious as it promotes extracellular degradation and production of inflammatory cytokines/chemokines and metabolic factors which activates glia and damages neurons. Thus, reduction of NSE by ENOblock may have potential therapeutic implications in acute SCI.
Collapse
Affiliation(s)
- Azizul Haque
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, BSB-201, Charleston, SC, 29425, USA.
| | - Mollie Capone
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, BSB-201, Charleston, SC, 29425, USA
- Ralph H. Johnson Veterans Administration Medical Center, Charleston, SC, USA
| | - Denise Matzelle
- Department of Neurosurgery, Medical University of South Carolina, Charleston, USA
- Ralph H. Johnson Veterans Administration Medical Center, Charleston, SC, USA
| | - April Cox
- FirstString Research, Mt. Pleasant, SC, USA
| | - Naren L Banik
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, BSB-201, Charleston, SC, 29425, USA
- Department of Neurosurgery, Medical University of South Carolina, Charleston, USA
- Ralph H. Johnson Veterans Administration Medical Center, Charleston, SC, USA
| |
Collapse
|
9
|
Jung SY, Seo TB, Kim DY. Treadmill exercise facilitates recovery of locomotor function through axonal regeneration following spinal cord injury in rats. J Exerc Rehabil 2016; 12:284-92. [PMID: 27656624 PMCID: PMC5031384 DOI: 10.12965/jer.1632698.349] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 07/31/2016] [Indexed: 11/29/2022] Open
Abstract
Spinal cord injury (SCI) disrupts both axonal pathways and segmental spinal cord circuity, resulting in permanent neurological deficits. Physical exercise is known to increase the expression of neurotrophins for improving the injured spinal cord. In the present study, we investigated the effects of treadmill exercise on locomotor function in relation with brain-derived neurotrophic factor (BDNF) expression after SCI. The rats were divided into five groups: control group, sham operation group, sham operation and exercise group, SCI group, and SCI and exercise group. The laminectomy was performed at the T9–T10 level. The exposed dorsal surface of the spinal cord received contusion injury (10 g × 25 mm) using the impactor. Treadmill exercise was performed 6 days per a week for 6 weeks. In order to evaluate the locomotor function of animals, Basso-Beattie-Bresnahan (BBB) locomotor scale was conducted once a week for 6 weeks. We examined BDNF expression and axonal sprouting in the injury site of the spinal cord using Western blot analysis and immunofluorescence staining. SCI induced loss of locomotor function with decreased BDNF expression in the injury site. Treadmill exercise increased the score of BBB locomotor scale and reduced cavity formation in the injury site. BDNF expression and axonal sprouting within the trabecula were further facilitated by treadmill exercise in SCI-exposed rats. The present study provides the evidence that treadmill exercise may facilitate recovery of locomotor function through axonal regeneration via BDNF expression following SCI.
Collapse
Affiliation(s)
- Sun-Young Jung
- Department of Physical Therapy, Hosan University, Gyeongsan, Korea
| | - Tae-Beom Seo
- Division of Sports Science and Engineering, Korea Institute of Sports Science, Seoul, Korea
| | - Dae-Young Kim
- Department of Sports Health Care, College of Humanities & Social Sciences, Inje University, Gimhae, Korea
| |
Collapse
|
10
|
Calpain inhibitor attenuates ER stress-induced apoptosis in injured spinal cord after bone mesenchymal stem cells transplantation. Neurochem Int 2016; 97:15-25. [DOI: 10.1016/j.neuint.2016.04.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Revised: 04/09/2016] [Accepted: 04/28/2016] [Indexed: 12/25/2022]
|
11
|
Moghieb A, Bramlett HM, Das JH, Yang Z, Selig T, Yost RA, Wang MS, Dietrich WD, Wang KKW. Differential Neuroproteomic and Systems Biology Analysis of Spinal Cord Injury. Mol Cell Proteomics 2016; 15:2379-95. [PMID: 27150525 PMCID: PMC4937511 DOI: 10.1074/mcp.m116.058115] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2016] [Revised: 04/08/2016] [Indexed: 12/13/2022] Open
Abstract
Acute spinal cord injury (SCI) is a devastating condition with many consequences and no known effective treatment. Although it is quite easy to diagnose traumatic SCI, the assessment of injury severity and projection of disease progression or recovery are often challenging, as no consensus biomarkers have been clearly identified. Here rats were subjected to experimental moderate or severe thoracic SCI. At 24h and 7d postinjury, spinal cord segment caudal to injury center versus sham samples was harvested and subjected to differential proteomic analysis. Cationic/anionic-exchange chromatography, followed by 1D polyacrylamide gel electrophoresis, was used to reduce protein complexity. A reverse phase liquid chromatography-tandem mass spectrometry proteomic platform was then utilized to identify proteome changes associated with SCI. Twenty-two and 22 proteins were up-regulated at 24 h and 7 day after SCI, respectively; whereas 19 and 16 proteins are down-regulated at 24 h and 7 day after SCI, respectively, when compared with sham control. A subset of 12 proteins were identified as candidate SCI biomarkers - TF (Transferrin), FASN (Fatty acid synthase), NME1 (Nucleoside diphosphate kinase 1), STMN1 (Stathmin 1), EEF2 (Eukaryotic translation elongation factor 2), CTSD (Cathepsin D), ANXA1 (Annexin A1), ANXA2 (Annexin A2), PGM1 (Phosphoglucomutase 1), PEA15 (Phosphoprotein enriched in astrocytes 15), GOT2 (Glutamic-oxaloacetic transaminase 2), and TPI-1 (Triosephosphate isomerase 1), data are available via ProteomeXchange with identifier PXD003473. In addition, Transferrin, Cathepsin D, and TPI-1 and PEA15 were further verified in rat spinal cord tissue and/or CSF samples after SCI and in human CSF samples from moderate/severe SCI patients. Lastly, a systems biology approach was utilized to determine the critical biochemical pathways and interactome in the pathogenesis of SCI. Thus, SCI candidate biomarkers identified can be used to correlate with disease progression or to identify potential SCI therapeutic targets.
Collapse
Affiliation(s)
- Ahmed Moghieb
- From the ‡Program for Neurotrauma, Neuroproteomics & Biomarkers Research, §The Departments of Psychiatry, and ‖Chemistry, University of Florida, Gainesville, Florida 32611
| | - Helen M Bramlett
- **Department of Neurological Surgery, ‡‡The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, 1095 NW 14th Terrace LPLC 3-18, Miami, Florida, 33136
| | - Jyotirmoy H Das
- From the ‡Program for Neurotrauma, Neuroproteomics & Biomarkers Research, §§Washington University School of Medicine, St. Louis, Missouri 63110
| | - Zhihui Yang
- From the ‡Program for Neurotrauma, Neuroproteomics & Biomarkers Research, §The Departments of Psychiatry, and
| | - Tyler Selig
- From the ‡Program for Neurotrauma, Neuroproteomics & Biomarkers Research
| | - Richard A Yost
- ‖Chemistry, University of Florida, Gainesville, Florida 32611
| | - Michael S Wang
- **Department of Neurological Surgery, ‡‡The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, 1095 NW 14th Terrace LPLC 3-18, Miami, Florida, 33136
| | - W Dalton Dietrich
- **Department of Neurological Surgery, ‡‡The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, 1095 NW 14th Terrace LPLC 3-18, Miami, Florida, 33136
| | - Kevin K W Wang
- From the ‡Program for Neurotrauma, Neuroproteomics & Biomarkers Research, §The Departments of Psychiatry, and ¶Neuroscience,
| |
Collapse
|
12
|
Vana PG, LaPorte HM, Kennedy RH, Gamelli RL, Majetschak M. Effects of a caspase and a calpain inhibitor on resting energy expenditures in normal and hypermetabolic rats: a pilot study. Physiol Res 2016; 65:537-41. [PMID: 27070748 DOI: 10.33549/physiolres.933201] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Several diseases induce hypermetabolism, which is characterized by increases in resting energy expenditures (REE) and whole body protein loss. Exaggerated protein degradation is thought to be the driving force underlying this response. The effects of caspase and calpain inhibitors on REE in physiological and hypermetabolic conditions, however, are unknown. Thus, we studied whether MDL28170 (calpain inhibitor) or z-VAD-fmk (caspase inhibitor) affect REE under physiological conditions and during hypermetabolism post-burn. Rats were treated five times weekly and observed for 6 weeks. Treatment was started 2 h (early) or 48 h (late) after burn. In normal rats, MDL28170 transiently increased REE to 130 % of normal during week 2-4. z-VAD-fmk reduced REE by 20-25 % throughout the observation period. Within 14 days after burns, REE increased to 130+/-5 %. Whereas MDL28170/early treatment did not affect REE, MDL28170/late transiently increased REE to 180+/-10 % of normal by week 4 post-burn. In contrast, with z-VAD-fmk/early REE remained between 90-110 % of normal post-burn. z-VAD-fmk/late did not affect burn-induced increases in REE. These data suggest that caspase cascades contribute to the development of hypermetabolism and that burn-induced hypermetabolism can be pharmacologically modulated. Our data point towards caspase cascades as possible therapeutic targets to attenuate hypermetabolism after burns, and possibly in other catabolic disease processes.
Collapse
Affiliation(s)
- P G Vana
- Burn and Shock Trauma Research Institute, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, USA.
| | | | | | | | | |
Collapse
|
13
|
Ramos RCDV, Alegrete N. O papel da farmacoterapia na modificação do estado neurológico de traumatizados vértebro‐medulares. Rev Bras Ortop 2015. [DOI: 10.1016/j.rbo.2014.09.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
14
|
do Vale Ramos RC, Alegrete N. The role of pharmacotherapy in modifying the neurological status of patients with spinal and spinal cord injuries. Rev Bras Ortop 2015; 50:617-24. [PMID: 27218071 PMCID: PMC4866940 DOI: 10.1016/j.rboe.2015.09.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 09/02/2014] [Indexed: 01/03/2023] Open
Abstract
The aim here was to conduct a review of the literature on pharmacological therapies for modifying the neurological status of patients with spinal cord injuries. The PubMed database was searched for articles with the terms "spinal cord injury AND methylprednisolone/GM1/apoptosis inhibitor/calpain inhibitor/naloxone/tempol/tirilazad", in Portuguese or in English, published over the last five years. Older studies were included because of their historical importance. The pharmacological groups were divided according to their capacity to interfere with the physiopathological mechanisms of secondary injuries. Use of methylprednisolone needs to be carefully weighed up: other anti-inflammatory agents have shown benefits in humans or in animals. GM1 does not seem to have greater efficacy than methylprednisolone, but longer-term studies are needed. Many inhibitors of apoptosis have shown benefits in in vitro studies or in animals. Naloxone has not shown benefits. Tempol inhibits the main consequences of oxidation at the level of the spinal cord and other antioxidant drugs seem to have an effect superior to that of methylprednisolone. There is an urgent need to find new treatments that improve the neurological status of patients with spinal cord injuries. The benefits from treatment with methylprednisolone have been questioned, with concerns regarding its safety. Other drugs have been studied, and some of these may provide promising alternatives. Additional studies are needed in order to reach conclusions regarding the benefits of these agents in clinical practice.
Collapse
|
15
|
Targeting ERK1/2-calpain 1-NF-κB signal transduction in secondary tissue damage and astrogliosis after spinal cord injury. ACTA ACUST UNITED AC 2015. [DOI: 10.1007/s11515-015-1373-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
16
|
Xu JY, Jiang Y, Liu W, Huang YG. Calpain inhibitor reduces cancer-induced bone pain possibly through inhibition of osteoclastogenesis in rat cancer-induced bone pain model. Chin Med J (Engl) 2015; 128:1102-7. [PMID: 25881607 PMCID: PMC4832953 DOI: 10.4103/0366-6999.155109] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Background: Calpain, a calcium-dependent cysteine protease, has been demonstrated to regulate osteoclastogenesis, which is considered one of the major reasons for cancer-induced bone pain (CIBP). In the present study, calpain inhibitor was applied in a rat CIBP model to determine whether it could reduce CIBP through regulation of osteoclastogenesis activity. Methods: A rat CIBP model was established with intratibial injection of Walker 256 cells. Then, the efficacy of intraperitoneal administered calpain inhibitor III (MDL28170, 1 mg/kg) on mechanical withdrawal threshold (MWT) of bilateral hind paws was examined on postoperative days (PODs) 2, 5, 8, 11, and 14. On POD 14, the calpain inhibitor's effect on tumor bone tartrate-resistant acid phosphatase (TRAP) stain and radiology was also carefully investigated. Results: Pain behavioral tests in rats showed that the calpain inhibitor effectively attenuated MWTs of both the surgical side and contralateral side hind paws on POD 5, 8, and 11 (P < 0.05). TRAP-positive cell count of the surgical side bone was significantly decreased in the calpain inhibitor group compared with the vehicle group (P < 0.05). However, bone resorption and destruction measured by radiographs showed no difference between the two groups. Conclusions: Calpain inhibitor can effectively reduce CIBP of both the surgical side and nonsurgical side after tumor injection in a rat CIBP model. It may be due to the inhibition of receptor activator of nuclear factor-kappa B ligand-induced osteoclastogenesis. Whether a calpain inhibitor could be a novel therapeutic target to treat CIBP needs further investigation.
Collapse
Affiliation(s)
| | | | - Wei Liu
- Department of Anesthesiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, China
| | | |
Collapse
|
17
|
White-Schenk D, Shi R, Leary JF. Nanomedicine strategies for treatment of secondary spinal cord injury. Int J Nanomedicine 2015; 10:923-38. [PMID: 25673988 PMCID: PMC4321603 DOI: 10.2147/ijn.s75686] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Neurological injury, such as spinal cord injury, has a secondary injury associated with it. The secondary injury results from the biological cascade after the primary injury and affects previous uninjured, healthy tissue. Therefore, the mitigation of such a cascade would benefit patients suffering a primary injury and allow the body to recover more quickly. Unfortunately, the delivery of effective therapeutics is quite limited. Due to the inefficient delivery of therapeutic drugs, nanoparticles have become a major field of exploration for medical applications. Based on their material properties, they can help treat disease by delivering drugs to specific tissues, enhancing detection methods, or a mixture of both. Incorporating nanomedicine into the treatment of neuronal injury and disease would likely push nanomedicine into a new light. This review highlights the various pathological issues involved in secondary spinal cord injury, current treatment options, and the improvements that could be made using a nanomedical approach.
Collapse
Affiliation(s)
- Désirée White-Schenk
- Interdisciplinary Biomedical Sciences Program, Purdue University, West Lafayette, IN, USA ; Birck Nanotechnology Center, Discovery Park, Purdue University, West Lafayette, IN, USA
| | - Riyi Shi
- Interdisciplinary Biomedical Sciences Program, Purdue University, West Lafayette, IN, USA ; Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA ; Department of Basic Medical Sciences, Lynn School of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
| | - James F Leary
- Interdisciplinary Biomedical Sciences Program, Purdue University, West Lafayette, IN, USA ; Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA ; Department of Basic Medical Sciences, Lynn School of Veterinary Medicine, Purdue University, West Lafayette, IN, USA ; Birck Nanotechnology Center, Discovery Park, Purdue University, West Lafayette, IN, USA
| |
Collapse
|
18
|
Kumar P, Choonara YE, Pillay V. In silico affinity profiling of neuroactive polyphenols for post-traumatic calpain inactivation: a molecular docking and atomistic simulation sensitivity analysis. Molecules 2014; 20:135-68. [PMID: 25546626 PMCID: PMC6272800 DOI: 10.3390/molecules20010135] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2014] [Accepted: 12/16/2014] [Indexed: 11/16/2022] Open
Abstract
Calcium-activated nonlysosomal neutral proteases, calpains, are believed to be early mediators of neuronal damage associated with neuron death and axonal degeneration after traumatic neural injuries. In this study, a library of biologically active small molecular weight calpain inhibitors was used for model validation and inhibition site recognition. Subsequently, two natural neuroactive polyphenols, curcumin and quercetin, were tested for their sensitivity and activity towards calpain's proteolytic sequence and compared with the known calpain inhibitors via detailed molecular mechanics (MM), molecular dynamics (MD), and docking simulations. The MM and MD energy profiles (SJA6017 < AK275 < AK295 < PD151746 < quercetin < leupeptin < PD150606 < curcumin < ALLN < ALLM < MDL-28170 < calpeptin) and the docking analysis (AK275 < AK295 < PD151746 < ALLN < PD150606 < curcumin < leupeptin < quercetin < calpeptin < SJA6017 < MDL-28170 < ALLM) demonstrated that polyphenols conferred comparable calpain inhibition profiling. The modeling paradigm used in this study provides the first detailed account of corroboration of enzyme inhibition efficacy of calpain inhibitors and the respective calpain-calpain inhibitor molecular complexes' energetic landscape and in addition stimulates the polyphenol bioactive paradigm for post-SCI intervention with implications reaching to experimental in vitro, in cyto, and in vivo studies.
Collapse
Affiliation(s)
- Pradeep Kumar
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 7 York Road, Parktown 2193, South Africa.
| | - Yahya E Choonara
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 7 York Road, Parktown 2193, South Africa.
| | - Viness Pillay
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 7 York Road, Parktown 2193, South Africa.
| |
Collapse
|
19
|
Zhang Z, Xu J, Bai Y, Hou T. Effects of Methylprednisolone on the Expression and Activity of Calpain Following Ischemia-Reperfusion Spinal Cord Injury in Rats. NEUROSCIENCE AND MEDICINE 2014; 05:23-31. [DOI: 10.4236/nm.2014.51005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/28/2023]
|
20
|
Yu CG, Singh R, Crowdus C, Raza K, Kincer J, Geddes JW. Fenbendazole improves pathological and functional recovery following traumatic spinal cord injury. Neuroscience 2013; 256:163-9. [PMID: 24183965 DOI: 10.1016/j.neuroscience.2013.10.039] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Revised: 10/17/2013] [Accepted: 10/19/2013] [Indexed: 01/16/2023]
Abstract
During a study of spinal cord injury (SCI), mice in our colony were treated with the anthelmintic fenbendazole to treat pinworms detected in other mice not involved in the study. As this was not part of the original experimental design, we subsequently compared pathological and functional outcomes of SCI in female C57BL/6 mice who received fenbendazole (150 ppm, 8 mg/kg body weight/day) for 4 weeks prior to moderate contusive SCI (50 kdyn force) as compared to mice on the same diet without added fenbendazole. The fenbendazole-treated mice exhibited improved locomotor function, determined using the Basso mouse scale, as well as improved tissue sparing following contusive SCI. Fenbendazole may exert protective effects through multiple possible mechanisms, one of which is inhibition of the proliferation of B lymphocytes, thereby reducing antibody responses. Autoantibodies produced following SCI contribute to the axon damage and locomotor deficits. Fenbendazole pretreatment reduced the injury-induced CD45R-positive B cell signal intensity and IgG immunoreactivity at the lesion epicenter 6 weeks after contusive SCI in mice, consistent with a possible effect on the immune response to the injury. Fenbendazole and related benzimadole antihelmintics are FDA approved, exhibit minimal toxicity, and represent a novel group of potential therapeutics targeting secondary mechanisms following SCI.
Collapse
Affiliation(s)
- C G Yu
- Spinal Cord and Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, KY 40536, USA; Department of Anatomy and Neurobiology, University of Kentucky College of Medicine, Lexington, KY 40536, USA.
| | - R Singh
- Spinal Cord and Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, KY 40536, USA; Department of Anatomy and Neurobiology, University of Kentucky College of Medicine, Lexington, KY 40536, USA.
| | - C Crowdus
- Spinal Cord and Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, KY 40536, USA; Department of Anatomy and Neurobiology, University of Kentucky College of Medicine, Lexington, KY 40536, USA.
| | - K Raza
- Spinal Cord and Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, KY 40536, USA.
| | - J Kincer
- Division of Laboratory Animal Resources, University of Kentucky College of Medicine, Lexington, KY 40536, USA.
| | - J W Geddes
- Spinal Cord and Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, KY 40536, USA; Department of Anatomy and Neurobiology, University of Kentucky College of Medicine, Lexington, KY 40536, USA.
| |
Collapse
|
21
|
Jeffery N, Levine J, Olby N, Stein V. Intervertebral Disk Degeneration in Dogs: Consequences, Diagnosis, Treatment, and Future Directions. J Vet Intern Med 2013; 27:1318-33. [DOI: 10.1111/jvim.12183] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2013] [Revised: 06/25/2013] [Accepted: 08/01/2013] [Indexed: 01/25/2023] Open
Affiliation(s)
- N.D. Jeffery
- Department of Veterinary Clinical Sciences; College of Veterinary Medicine; Iowa State University; Ames IA
| | - J.M. Levine
- Department of Small Animal Clinical Sciences; College of Veterinary Medicine and Biomedical Sciences; Texas A&M University; College Station TX
| | - N.J. Olby
- College of Veterinary Medicine; North Carolina State University; Raleigh NC
| | - V.M. Stein
- Department of Small Animal Medicine and Surgery; University of Veterinary Medicine Hannover; Hannover Germany
| |
Collapse
|
22
|
Yu CG, Li Y, Raza K, Yu XX, Ghoshal S, Geddes JW. Calpain 1 knockdown improves tissue sparing and functional outcomes after spinal cord injury in rats. J Neurotrauma 2013; 30:427-33. [PMID: 23102374 PMCID: PMC4169127 DOI: 10.1089/neu.2012.2561] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
To evaluate the hypothesis that calpain 1 knockdown would reduce pathological damage and functional deficits after spinal cord injury (SCI), we developed lentiviral vectors encoding calpain 1 shRNA and eGFP as a reporter (LV-CAPN1 shRNA). The ability of LV-CAPN1 shRNA to knockdown calpain 1 was confirmed in rat NRK cells using Northern and Western blot analysis. To investigate the effects on spinal cord injury, LV-CAPN1shRNA or LV-mismatch control shRNA (LV-control shRNA) were administered by convection enhanced diffusion at spinal cord level T10 in Long-Evans female rats (200-250 g) 1 week before contusion SCI, 180 kdyn force, or sham surgery at the same thoracic level. Intraspinal administration of the lentiviral particles resulted in transgene expression, visualized by eGFP, in spinal tissue at 2 weeks after infection. Calpain 1 protein levels were reduced by 54% at T10 2 weeks after shRNA-mediated knockdown (p<0.05, compared with the LV-control group, n=3 per group) while calpain 2 levels were unchanged. Intraspinal administration of LV-CAPN1shRNA 1 week before contusion SCI resulted in a significant improvement in locomotor function over 6 weeks postinjury, compared with LV-control administration (p<0.05, n=10 per group). Histological analysis of spinal cord sections indicated that pre-injury intraspinal administration of LV-CAPN1shRNA significantly reduced spinal lesion volume and improved total tissue sparing, white matter sparing, and gray matter sparing (p<0.05, n=10 per group). Together, results support the hypothesis that calpain 1 activation contributes to the tissue damage and impaired locomotor function after SCI, and that calpain1 represents a potential therapeutic target.
Collapse
Affiliation(s)
- Chen Guang Yu
- Spinal Cord and Brain Injury Research Center and Department of Anatomy and Neurobiology, University of Kentucky College of Medicine, Lexington, Kentucky 40536-0509, USA.
| | | | | | | | | | | |
Collapse
|
23
|
Veeravalli KK, Dasari VR, Rao JS. Regulation of proteases after spinal cord injury. J Neurotrauma 2012; 29:2251-62. [PMID: 22709139 DOI: 10.1089/neu.2012.2460] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Spinal cord injury is a major medical problem worldwide. Unfortunately, we still do not have suitable therapeutic agents for the treatment of spinal cord injury and prevention of its devastating consequences. Scientists and physicians are baffled by the challenges of controlling progressive neurodegeneration in spinal cord injury, which has not been healed with any currently-available treatments. Although extensive work has been carried out to better understand the pathophysiology of spinal cord injury, our current understanding of the repair mechanisms of secondary injury processes is still meager. Several investigators reported the crucial role played by various proteases after spinal cord injury. Understanding the beneficial and harmful roles these proteases play after spinal cord injury will allow scientists to plan and design appropriate treatment strategies to improve functional recovery after spinal cord injury. This review will focus on various proteases such as matrix metalloproteinases, cysteine proteases, and serine proteases and their inhibitors in the context of spinal cord injury.
Collapse
Affiliation(s)
- Krishna Kumar Veeravalli
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, Illinois 61605, USA
| | | | | |
Collapse
|
24
|
Schoch KM, Evans HN, Brelsfoard JM, Madathil SK, Takano J, Saido TC, Saatman KE. Calpastatin overexpression limits calpain-mediated proteolysis and behavioral deficits following traumatic brain injury. Exp Neurol 2012; 236:371-82. [PMID: 22572592 DOI: 10.1016/j.expneurol.2012.04.022] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Revised: 03/23/2012] [Accepted: 04/23/2012] [Indexed: 02/06/2023]
Abstract
Traumatic brain injury (TBI) results in abrupt, initial cell damage leading to delayed neuronal death. The calcium-activated proteases, calpains, are known to contribute to this secondary neurodegenerative cascade. Although the specific inhibitor of calpains, calpastatin, is present within neurons, normal levels of calpastatin are unable to fully prevent the damaging proteolytic activity of calpains after injury. In this study, increased calpastatin expression was achieved using transgenic mice that overexpress the human calpastatin (hCAST) construct under control of a calcium-calmodulin-dependent kinase II α promoter. Naïve hCAST transgenic mice exhibited enhanced neuronal calpastatin expression and significantly reduced protease activity. Acute calpain-mediated spectrin proteolysis in the cortex and hippocampus induced by controlled cortical impact brain injury was significantly attenuated in calpastatin overexpressing mice. Aspects of posttraumatic motor and cognitive behavioral deficits were also lessened in hCAST transgenic mice compared to their wildtype littermates. However, volumetric analyses of neocortical contusion revealed no histological neuroprotection at either acute or long-term time points. Partial hippocampal neuroprotection observed at a moderate injury severity was lost after severe TBI. This study underscores the effectiveness of calpastatin overexpression in reducing calpain-mediated proteolysis and behavioral impairment after TBI, supporting the therapeutic potential for calpain inhibition. In addition, the reduction in spectrin proteolysis without accompanied neocortical neuroprotection suggests the involvement of other factors that are critical for neuronal survival after contusion brain injury.
Collapse
Affiliation(s)
- Kathleen M Schoch
- Spinal Cord and Brain Injury Research Center, University of Kentucky, B416 Biomedical and Biological Sciences Research Building, 741 South Limestone Street, Lexington, KY 40536-0509, USA.
| | | | | | | | | | | | | |
Collapse
|
25
|
Priestley JV, Michael-Titus AT, Tetzlaff W. Limiting spinal cord injury by pharmacological intervention. HANDBOOK OF CLINICAL NEUROLOGY 2012; 109:463-484. [PMID: 23098731 DOI: 10.1016/b978-0-444-52137-8.00029-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
The direct primary mechanical trauma to neurons, glia and blood vessels that occurs with spinal cord injury (SCI) is followed by a complex cascade of biochemical and cellular changes which serve to increase the size of the injury site and the extent of cellular and axonal loss. The aim of neuroprotective strategies in SCI is to limit the extent of this secondary cell loss by inhibiting key components of the evolving injury cascade. In this review we will briefly outline the pathophysiological events that occur in SCI, and then review the wide range of neuroprotective agents that have been evaluated in preclinical SCI models. Agents will be considered under the following categories: antioxidants, erythropoietin and derivatives, lipids, riluzole, opioid antagonists, hormones, anti-inflammatory agents, statins, calpain inhibitors, hypothermia, and emerging strategies. Several clinical trials of neuroprotective agents have already taken place and have generally had disappointing results. In attempting to identify promising new treatments, we will therefore highlight agents with (1) low known risks or established clinical use, (2) behavioral data gained in clinically relevant animal models, (3) efficacy when administered after the injury, and (4) robust effects seen in more than one laboratory and/or more than one model of SCI.
Collapse
|
26
|
Striatal inhibition of calpains prevents levodopa-induced neurochemical changes and abnormal involuntary movements in the hemiparkinsonian rat model. Neurobiol Dis 2011; 45:645-55. [PMID: 22037042 DOI: 10.1016/j.nbd.2011.10.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Revised: 10/04/2011] [Accepted: 10/11/2011] [Indexed: 11/21/2022] Open
Abstract
Pharmacological dopamine replacement with l-3,4-dihydroxyphenylalanine (L-DOPA) remains the most effective approach to treat the motor symptoms of Parkinson's disease (PD). However, as the disease progresses, the therapeutic response to L-DOPA gradually becomes erratic and is associated with the emergence of dyskinesia in the majority of patients. The pathogenesis of L-DOPA-induced dyskinesia (LID) is still unknown. In the current study, using the 6-hydroxydopamine (6-OHDA)-lesioned rat model of PD, we demonstrated that the calcium-dependent proteins calpains and cdk5 of the striatum play a critical role in the behavioral and molecular changes evoked by L-DOPA therapy. We first confirmed that L-DOPA reversed PD symptoms, assessed by the cylinder, stepping and vibrissae-elicited reaching tests in this animal model, and elicited robust abnormal involuntary movements (AIMs) reminiscent of LID. Interestingly, intrastriatal infusion of the calpains inhibitor MDL28170, and to a lower extent the cdk5 inhibitor roscovitine, reduced the severity and amplitude of AIMs without affecting L-DOPA's antiparkinsonian effects. Notably, the calpains and cdk5 inhibitors totally reversed the striatal molecular changes attributed to L-DOPA therapy, such as ERK1/2 and dynamin phosphorylation. Another fascinating observation was that L-DOPA therapy, in combination with intrastriatal infusion of MDL28170, augmented tyrosine hydroxylase levels in the striatum of lesioned rats without affecting the number of dopaminergic cells in the substantia nigra. These findings disclose a novel mechanism underlying the maladaptive alterations induced by L-DOPA therapy in the 6-OHDA rat model of PD.
Collapse
|
27
|
Hill CE, Guller Y, Raffa SJ, Hurtado A, Bunge MB. A calpain inhibitor enhances the survival of Schwann cells in vitro and after transplantation into the injured spinal cord. J Neurotrauma 2011; 27:1685-95. [PMID: 20568964 DOI: 10.1089/neu.2010.1272] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Despite the diversity of cells available for transplantation into sites of spinal cord injury (SCI), and the known ability of transplanted cells to integrate into host tissue, functional improvement associated with cellular transplantation has been limited. One factor potentially limiting the efficacy of transplanted cells is poor cell survival. Recently we demonstrated rapid and early death of Schwann cells (SCs) within the first 24 h after transplantation, by both necrosis and apoptosis, which results in fewer than 20% of the cells surviving beyond 1 week. To enhance SC transplant survival, in vitro and in vivo models to rapidly screen compounds for their ability to promote SC survival are needed. The current study utilized in vitro models of apoptosis and necrosis, and based on withdrawal of serum and mitogens and the application of hydrogen peroxide, we screened several inhibitors of apoptosis and necrosis. Of the compounds tested, the calpain inhibitor MDL28170 enhanced SC survival both in vitro in response to oxidative stress induced by application of H2O2, and in vivo following delayed transplantation into the moderately contused spinal cord. The results support the use of calpain inhibitors as a promising new treatment for promoting the survival of transplanted cells. They also suggest that in vitro assays for cell survival may be useful for establishing new compounds that can then be tested in vivo for their ability to promote transplanted SC survival.
Collapse
Affiliation(s)
- Caitlin E Hill
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida, USA.
| | | | | | | | | |
Collapse
|
28
|
Ray SK, Samantaray S, Smith JA, Matzelle DD, Das A, Banik NL. Inhibition of cysteine proteases in acute and chronic spinal cord injury. Neurotherapeutics 2011; 8:180-6. [PMID: 21373949 PMCID: PMC3101838 DOI: 10.1007/s13311-011-0037-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Spinal cord injury (SCI) is a serious neurological disorder that debilitates mostly young people. Unfortunately, we still do not have suitable therapeutic agents for treatment of SCI and prevention of its devastating consequences. However, we have gained a good understanding of pathological mechanisms that cause neurodegeneration leading to paralysis or even death following SCI. Primary injury to the spinal cord initiates the secondary injury process that includes various deleterious factors for ultimate activation of different cysteine proteases for degradation of cellular key cytoskeleton and other crucial proteins for delayed death of neurons and glial cells at the site of SCI and its penumbra in different animal models. An important aspect of SCI is the increase in intracellular free Ca(2+) concentration within a short time of primary injury. Various studies in different laboratories demonstrate that the most important cysteine protease for neurodegeneration in SCI is calpain, which absolutely requires intracellular free Ca(2+) for its activation. Furthermore, other cysteine proteases, such as caspases and cathepsin B also make a contribution to neurodegeneration in SCI. Therefore, inhibition of cysteine proteases is an important goal in prevention of neurodegeneration in SCI. Studies showed that individual inhibitors of cysteine proteases provided significant neuroprotection in animal models of SCI. Recent studies suggest that physiological hormones, such as estrogen and melatonin, can be successfully used for prevention of neurodegeneration and preservation of motor function in acute SCI as well as in chronic SCI in rats.
Collapse
Affiliation(s)
- Swapan K. Ray
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina 29209 USA
| | - Supriti Samantaray
- Division of Neurology, Department of Neurosciences, Medical University of South Carolina, 96 Jonathan Lucas Street, Suite 309 CSB, Charleston, South Carolina 29425 USA
| | - Joshua A. Smith
- Division of Neurology, Department of Neurosciences, Medical University of South Carolina, 96 Jonathan Lucas Street, Suite 309 CSB, Charleston, South Carolina 29425 USA
| | - Denise D. Matzelle
- Division of Neurology, Department of Neurosciences, Medical University of South Carolina, 96 Jonathan Lucas Street, Suite 309 CSB, Charleston, South Carolina 29425 USA
| | - Arabinda Das
- Division of Neurology, Department of Neurosciences, Medical University of South Carolina, 96 Jonathan Lucas Street, Suite 309 CSB, Charleston, South Carolina 29425 USA
| | - Naren L. Banik
- Division of Neurology, Department of Neurosciences, Medical University of South Carolina, 96 Jonathan Lucas Street, Suite 309 CSB, Charleston, South Carolina 29425 USA
| |
Collapse
|
29
|
Thompson SN, Carrico KM, Mustafa AG, Bains M, Hall ED. A pharmacological analysis of the neuroprotective efficacy of the brain- and cell-permeable calpain inhibitor MDL-28170 in the mouse controlled cortical impact traumatic brain injury model. J Neurotrauma 2011; 27:2233-43. [PMID: 20874056 DOI: 10.1089/neu.2010.1474] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The cytoskeletal and neuronal protective effects of early treatment with the blood-brain barrier- and cell-permeable calpain inhibitor MDL-28170 was examined in the controlled cortical impact (CCI) traumatic brain injury (TBI) model in male CF-1 mice. This was preceded by a dose-response and pharmacodynamic evaluation of IV or IP doses of MDL-28170 with regard to ex vivo inhibition of calpain 2 activity in harvested brain homogenates. From these data, we tested the effects of an optimized MDL-28170 dosing regimen on calpain-mediated degradation of the neuronal cytoskeletal protein α-spectrin in cortical or hippocampal tissue of mice 24 h after CCI-TBI (1.0 mm depth, 3.5 m/sec velocity). With treatment initiated at 15 min post-TBI, α-spectrin degradation was significantly reduced by 40% in hippocampus and 44% in cortex. This effect was still observed with a 1-h but not a 3-h post-TBI delay. The cytoskeletal protection is most likely taking place in neurons surrounding the area of mainly necrotic degeneration, since MDL-28170 did not reduce hemispheric lesion volume as measured by the aminocupric silver staining method. This lack of effect on lesion volume has been seen with other calpain inhibitors, which suggests that pharmacological calpain inhibition by itself, while able to reduce axonal injury, may not be able to produce a measurable reduction in lesion volume. This is in contrast to certain other neuroprotective mechanistic approaches such as the mitochondrial protectant cyclosporine A, which produces at least a partial decrease in lesion volume in the same model. Accordingly, the combination of a calpain inhibitor with a compound such as cyclosporine A may be needed to achieve the optimal degree of post-TBI neuroprotection.
Collapse
Affiliation(s)
- Stephanie N Thompson
- University of Kentucky Spinal Cord & Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, Kentucky 40536-0509, USA
| | | | | | | | | |
Collapse
|
30
|
MDL-28170 has no analgesic effect on CCI induced neuropathic pain in mice. Molecules 2010; 15:3038-47. [PMID: 20657463 PMCID: PMC6263360 DOI: 10.3390/molecules15053038] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2010] [Revised: 04/13/2010] [Accepted: 04/26/2010] [Indexed: 11/16/2022] Open
Abstract
The calpain inhibitor MDL-28710 blocks the early local pro-inflammatory cytokine gene expression in mice after chronic constriction nerve injury (CCI). One-hundred-thirteen wild type mice of C57Bl/6J background received CCI of the right sciatic nerve. Mechanical paw withdrawal thresholds and thermal withdrawal latencies were investigated at baseline and at 1, 3, and 7 days after CCI. Three application regimens were used for MDL-28170: a) single injection 40 min before CCI; b) serial injections of MDL-28170 40 min before and up to day three after CCI; c) sustained application via intraperitoneal osmotic pumps. The control animals received the vehicle DMSO/PEG 400. The tolerable dose of MDL-28170 for mice was 30 mg/kg body weight, higher doses were lethal within the first hours after application. Mechanical withdrawal thresholds and thermal withdrawal latencies were reduced after CCI and did not normalize after single or serial injections, nor with application of MDL-28170 via osmotic pumps. Although the calpain inhibitor MDL-28170 inhibits the early local cytokine upregulation in the sciatic nerve after CCI, pain behavior is not altered. This finding implies that local cytokine upregulation after nerve injury alone is only one factor in the induction and maintenance of neuropathic pain.
Collapse
|
31
|
Yu CG, Yezierski RP, Joshi A, Raza K, Li Y, Geddes JW. Involvement of ERK2 in traumatic spinal cord injury. J Neurochem 2010; 113:131-42. [PMID: 20067580 DOI: 10.1111/j.1471-4159.2010.06579.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Activation of extracellular signal-regulated protein kinase 1/2 (ERK1/2) are implicated in the pathophysiology of spinal cord injury (SCI). However, the specific functions of individual ERK isoforms in neurodegeneration are largely unknown. We investigated the hypothesis that ERK2 activation may contribute to pathological and functional deficits following SCI and that ERK2 knockdown using RNA interference may provide a novel therapeutic strategy for SCI. Lentiviral ERK2 shRNA and siRNA were utilized to knockdown ERK2 expression in the spinal cord following SCI. Pre-injury intrathecal administration of ERK2 siRNA significantly reduced excitotoxic injury-induced activation of ERK2 (p < 0.001) and caspase 3 (p < 0.01) in spinal cord. Intraspinal administration of lentiviral ERK2 shRNA significantly reduced ERK2 expression in the spinal cord (p < 0.05), but did not alter ERK1 expression. Administration of the lentiviral ERK2 shRNA vector 1 week prior to severe spinal cord contusion injury resulted in a significant improvement in locomotor function (p < 0.05), total tissue sparing (p < 0.05), white matter sparing (p < 0.05), and gray matter sparing (p < 0.05) 6 weeks following severe contusive SCI. Our results suggest that ERK2 signaling is a novel target associated with the deleterious consequences of spinal injury.
Collapse
Affiliation(s)
- Chen-Guang Yu
- Spinal Cord and Brain Injury Research Center and Department of Anatomy and Neurobiology, University of Kentucky College of Medicine, Lexington, Kentucky 40536-0509, USA.
| | | | | | | | | | | |
Collapse
|
32
|
Gold MS, Kobeissy FH, Wang KKW, Merlo LJ, Bruijnzeel AW, Krasnova IN, Cadet JL. Methamphetamine- and trauma-induced brain injuries: comparative cellular and molecular neurobiological substrates. Biol Psychiatry 2009; 66:118-27. [PMID: 19345341 PMCID: PMC2810951 DOI: 10.1016/j.biopsych.2009.02.021] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2008] [Revised: 02/19/2009] [Accepted: 02/22/2009] [Indexed: 12/21/2022]
Abstract
The use of methamphetamine (METH) is a growing public health problem, because its abuse is associated with long-term biochemical and structural effects on the human brain. Neurodegeneration is often observed in humans, because of mechanical injuries (e.g., traumatic brain injury [TBI]) and ischemic damage (strokes). In this review, we discuss recent findings documenting the fact that the psychostimulant drug METH can cause neuronal damage in several brain regions. The accumulated evidence from our laboratories and those of other investigators indicates that acute administration of METH leads to activation of calpain and caspase proteolytic systems. These systems are also involved in causing neuronal damage secondary to traumatic and ischemic brain injuries. Protease activation is accompanied by proteolysis of endogenous neuronal structural proteins (alphaII-spectrin protein and microtubule-associated protein-tau), evidenced by the appearance of their breakdown products after these injuries. When taken together, these observations suggest that METH exposure, like TBI, can cause substantial damage to the brain by causing both apoptotic and necrotic cell death in the brains of METH addicts who use large doses of the drug during their lifetimes. Finally, because METH abuse is accompanied by functional and structural changes in the brain similar to those in TBI, METH addicts might experience greater benefit if their treatment involved greater emphasis on rehabilitation in conjunction with potential neuroprotective pharmacological agents such as calpain and caspase inhibitors similar to those used in TBI.
Collapse
Affiliation(s)
- Mark S Gold
- Center for Neuroproteomics and Biomarkers Research, McKnight Brain Institute of the University of Florida, Gainesville, Florida 32610, USA
| | | | | | | | | | | | | |
Collapse
|
33
|
Yune TY, Lee JY, Cui CM, Kim HC, Oh TH. Neuroprotective effect of Scutellaria baicalensis on spinal cord injury in rats. J Neurochem 2009; 110:1276-87. [PMID: 19519665 DOI: 10.1111/j.1471-4159.2009.06214.x] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Inflammation has been known to play an important role in the pathogenesis after spinal cord injury (SCI). Microglia are activated after injury and produce a variety of proinflammatory factors such as tumor necrosis factor-alpha, interleukin-1beta, cyclooxygenase-2, and reactive oxygen species leading to apoptosis of neurons and oligodendrocytes. In this study, we examined the neuroprotective effects of total ethanol extract of Scutellaria baicalensis (EESB), after SCI. Using primary microglial cultures, EESB treatment significantly inhibited lipopolysaccharide-induced expression of such inflammatory mediators as tumor necrosis factor-alpha, IL-1beta, IL-6, cyclooxygenase-2, and inducible nitric oxide synthase. Furthermore, reactive oxygen species and nitric oxide production were significantly attenuated by EESB treatment. For in vivo study, rats that had received a moderate spinal cord contusion injury at T9 received EESB orally at a dose of 100 mg/kg. EESB inhibited expression of proinflammatory factors and protein carbonylation and nitration after SCI. EESB also inhibited microglial activation at 4 h after injury. Furthermore, EESB significantly inhibited apoptotic cell death of neurons and oligodendrocytes and improved functional recovery after SCI. Lesion cavity and myelin loss were also reduced following EESB treatment. Thus, our data suggest that EESB significantly improve functional recovery by inhibiting inflammation and oxidative stress after injury.
Collapse
Affiliation(s)
- Tae Y Yune
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul, Korea
| | | | | | | | | |
Collapse
|
34
|
Reed TT, Owen J, Pierce WM, Sebastian A, Sullivan PG, Butterfield DA. Proteomic identification of nitrated brain proteins in traumatic brain-injured rats treated postinjury with gamma-glutamylcysteine ethyl ester: Insights into the role of elevation of glutathione as a potential therapeutic strategy for traumatic brain injury. J Neurosci Res 2009; 87:408-17. [DOI: 10.1002/jnr.21872] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
35
|
Neurite consolidation is an active process requiring constant repression of protrusive activity. EMBO J 2008; 28:248-60. [PMID: 19096364 DOI: 10.1038/emboj.2008.265] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2008] [Accepted: 11/25/2008] [Indexed: 01/05/2023] Open
Abstract
During development, neurons extend projections that pathfind to reach their appropriate targets. These projections are composed of two distinct domains: a highly dynamic growth cone and a stable neurite shaft, which is considered to be consolidated. Although the regulation of these domains is critical to the appropriate formation of neural networks, the molecular mechanisms that regulate neurite shape remain poorly understood. Here, we show that calpain protease activity localizes to the neurite shaft, where it is essential for the repression of protrusive activity by limiting cortactin levels and inhibiting actin polymerization. Correspondingly, inhibition of calpain by branching factors induces the formation of new growth cones along the neurite shaft through cAMP elevation. These findings demonstrate that neurite consolidation is an active process requiring constant repression of protrusive activity. We also show that sprouting is, at least in part, accomplished by turning off the mechanism of consolidation.
Collapse
|
36
|
Yu CG, Joshi A, Geddes JW. Intraspinal MDL28170 microinjection improves functional and pathological outcome following spinal cord injury. J Neurotrauma 2008; 25:833-40. [PMID: 18627259 DOI: 10.1089/neu.2007.0490] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Although calpain (calcium-activated cysteine protease) inhibition represents a rational therapeutic target for spinal cord injury (SCI), few studies have reported improved functional outcomes with post-injury administration of calpain inhibitors. This reflects the weak potency and limited aqueous solubility of current calpain inhibitors. Previously, we demonstrated that intraspinal microinjection of the calpain inhibitor MDL28170 resulted in greater inhibition of calpain activity as compared to systemic administration of the same compound. In the present study, we evaluated the ability of intraspinal MDL28170 microinjection to spare spinal tissue and locomotor dysfunction following SCI. Contusion SCI was produced in female Long-Evans rats using the Infinite Horizon impactor at the 200-kdyn force setting. Open-field locomotion was evaluated until 6 weeks post-injury. Histological assessment of tissue sparing was performed at 6 weeks after SCI. The results demonstrate that MDL28170, administered with a single post-injury intraspinal microinjection (50 nmoles), significantly improves both locomotor function and pathological outcome measures following SCI.
Collapse
Affiliation(s)
- Chen-Guang Yu
- Spinal Cord and Brain Injury Research Center, Department of Anatomy and Neurobiology, University of Kentucky, Lexington, Kentucky, USA
| | | | | |
Collapse
|
37
|
Yune TY, Lee JY, Jiang MH, Kim DW, Choi SY, Oh TH. Systemic administration of PEP-1-SOD1 fusion protein improves functional recovery by inhibition of neuronal cell death after spinal cord injury. Free Radic Biol Med 2008; 45:1190-200. [PMID: 18722523 DOI: 10.1016/j.freeradbiomed.2008.07.016] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2008] [Revised: 06/19/2008] [Accepted: 07/15/2008] [Indexed: 01/08/2023]
Abstract
Spinal cord injury (SCI) produces excessive levels of reactive oxygen species (ROS) that induce apoptosis of neurons. Cu,Zn-superoxide dismutase (SOD1) is a key antioxidant enzyme that detoxifies intracellular ROS, thereby protecting cells from oxidative damage. PEP-1 is a peptide carrier capable of delivering full-length native peptides or proteins into cells. In the study described here, we fused a human SOD1 gene with PEP-1 in a bacterial expression vector to produce a genetic in-frame PEP-1-SOD1 fusion protein; we then investigated the neuroprotective effect of the fusion protein after SCI. The expressed and purified PEP-1-SOD1 was efficiently delivered into cultured cells and spinal cords in vivo, and the delivered fusion protein was biologically active. Systemic administration of PEP-1-SOD1 significantly decreased levels of ROS and protein carbonylation and nitration in spinal motor neurons after injury. PEP-1-SOD1 treatment also significantly inhibited mitochondrial cytochrome c release and activation of caspase-9 and caspase-3 in spinal cords after injury. Furthermore, PEP-1-SOD1 treatment significantly reduced ROS-induced apoptosis of motor neurons and improved functional recovery after SCI. These results suggest that PEP-1-SOD1 may provide a novel strategy for the therapeutic delivery of antioxidant enzymes that protect neurons from ROS after SCI.
Collapse
Affiliation(s)
- Tae Young Yune
- Age-Related and Brain Diseases Research Center, Kyung Hee University, Seoul 130-701, Korea.
| | | | | | | | | | | |
Collapse
|
38
|
Experimental reovirus-induced acute flaccid paralysis and spinal motor neuron cell death. J Neuropathol Exp Neurol 2008; 67:231-9. [PMID: 18344914 DOI: 10.1097/nen.0b013e31816564f0] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Acute flaccid paralysis (AFP) describes the loss of motor function in 1 or more limbs commonly associated with viral infection and destruction of motor neurons in the anterior horns of the spinal cord. Therapy is limited, and the development of effective treatments is hampered by a lack of experimental models. Reovirus infection of neonatal mice provides a model for the study of CNS viral infection pathogenesis. Injection of the Reovirus serot Type 3 strains Abney (T3A) or Dearing (T3D) into the hindlimb of 1-day-old mice resulted in the development of AFP in more than 90% of infected mice. Acute flaccid paralysis began in the ipsilateral hindlimb at 8 to 10 days postinfection and progressed to paraplegia 24 hours later. Paralysis correlated with injury, neuron loss, and spread of viral antigen first to the ipsilateral and then to the contralateral anterior horns. As demonstrated by the activation of caspase 3 and its colocalization with viral antigen in the anterior horn and concomitant cleavage of poly-(adenosine diphosphate-ribose) polymerase, AFP was associated with apoptosis. Calpain activity and inducible nitric oxide synthase expression were both elevated in the spinal cords of paralyzed animals. This study represents the first detailed characterization of a novel and highly efficient experimental model of virus-induced AFP that will facilitate evaluation of therapeutic strategies targeting virus-induced paralysis.
Collapse
|