1
|
Takahashi M, DiBattista JD, Jarman S, Newman SJ, Wakefield CB, Harvey ES, Bunce M. Partitioning of diet between species and life history stages of sympatric and cryptic snappers (Lutjanidae) based on DNA metabarcoding. Sci Rep 2020; 10:4319. [PMID: 32152406 PMCID: PMC7062689 DOI: 10.1038/s41598-020-60779-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 02/17/2020] [Indexed: 01/23/2023] Open
Abstract
Lutjanus erythropterus and L. malabaricus are sympatric, sister taxa that are important to fisheries throughout the Indo-Pacific. Their juveniles are morphologically indistinguishable (i.e. cryptic). A DNA metabarcoding dietary study was undertaken to assess the diet composition and partitioning between the juvenile and adult life history stages of these two lutjanids. Major prey taxa were comprised of teleosts and crustaceans for all groups except adult L. erythropterus, which instead consumed soft bodied invertebrates (e.g. tunicates, comb jellies and medusae) as well as teleosts, with crustaceans being notably absent. Diet composition was significantly different among life history stages and species, which may be associated with niche habitat partitioning or differences in mouth morphology within adult life stages. This study provides the first evidence of diet partitioning between cryptic juveniles of overlapping lutjanid species, thus providing new insights into the ecological interactions, habitat associations, and the specialised adaptations required for the coexistence of closely related species. This study has improved our understanding of the differential contributions of the juvenile and adult diets of these sympatric species within food webs. The diet partitioning reported in this study was only revealed by the taxonomic resolution provided by the DNA metabarcoding approach and highlights the potential utility of this method to refine the dietary components of reef fishes more generally.
Collapse
Affiliation(s)
- Miwa Takahashi
- School of Molecular and Life Sciences, Curtin University, Kent Street, Bentley, Perth, WA, 6102, Australia.
| | - Joseph D DiBattista
- School of Molecular and Life Sciences, Curtin University, Kent Street, Bentley, Perth, WA, 6102, Australia
- Australian Museum Research Institute, Australian Museum, 1 William Street, Sydney, NSW, 2010, Australia
| | - Simon Jarman
- School of Molecular and Life Sciences, Curtin University, Kent Street, Bentley, Perth, WA, 6102, Australia
- School of Biological Sciences, University of Western Australia, 35 Stirling Highway, Perth, WA, 6009, Australia
| | - Stephen J Newman
- Western Australian Fisheries and Marine Research Laboratories, Department of Primary Industries and Regional Development, Government of Western Australia, P.O. Box 20, North Beach, WA, 6920, Australia
| | - Corey B Wakefield
- Western Australian Fisheries and Marine Research Laboratories, Department of Primary Industries and Regional Development, Government of Western Australia, P.O. Box 20, North Beach, WA, 6920, Australia
| | - Euan S Harvey
- School of Molecular and Life Sciences, Curtin University, Kent Street, Bentley, Perth, WA, 6102, Australia
| | - Michael Bunce
- School of Molecular and Life Sciences, Curtin University, Kent Street, Bentley, Perth, WA, 6102, Australia
- Environmental Protection Authority, 215 Lambton Quay, Wellington, 6011, New Zealand
| |
Collapse
|
2
|
A ratiometric two-photon probe for Ca2+ in live tissues and its application to spinal cord injury model. Biomaterials 2017; 141:251-259. [DOI: 10.1016/j.biomaterials.2017.07.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 07/04/2017] [Accepted: 07/05/2017] [Indexed: 11/23/2022]
|
3
|
Bagherieh M, Kheirollahi A, Shahaboddin ME, Khajeh K, Golestani A. Calcium and TNFα additively affect the chondroitinase ABC I activity. Int J Biol Macromol 2017; 103:1201-1206. [DOI: 10.1016/j.ijbiomac.2017.05.177] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 05/15/2017] [Accepted: 05/30/2017] [Indexed: 12/28/2022]
|
4
|
Hill Lucas J, Emery DG, Rosenberg LJ. REVIEW ■ : Physical Injury of Neurons: Important Roles for Sodium and Chloride Ions. Neuroscientist 2016. [DOI: 10.1177/107385849700300208] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
There is growing evidence that ions other than Ca2+ play important roles in the deterioration of neuronal elements in both gray and white matter after physical injury. This review features information gathered with a tissue culture model of dendrite transection regarding the contributions of Na+ and CI- to ultrastructural damage and neuronal death. This information and the results of other in vitro investigations of physical and ischemic/excitotoxic injuries indicate that elevation of internal Na+ is an early event that may contribute significantly to neuronal injury through effects on Na+-driven transport mechanisms. Proposed deleterious consequences include cytoplasmic acidification, reduced mitochondrial energy production, and elevation of intracellular Ca2+ and extracellular excitatory amino acids to toxic levels. Prevention of Na+ entry into neurons after injury has been found to limit ultrastructural damage, prevent death, and preserve electrophysiological function. Although the role of CI- in neuronal injury is less well defined, there is also evidence that elevation of intracellular CI- contributes to structural damage, particularly to the smooth endoplasmic reticulum. In terventions that limit Na+- and CI--mediated damage to injured neurons may have utility in neurosurgery and as acute phase treatments for nervous system trauma and other pathological states. NEURO SCIENTIST 3:89-101, 1997
Collapse
Affiliation(s)
- Jen Hill Lucas
- Department of Physiology The Ohio State University Columbus,
Ohio
| | - Dennis G. Emery
- Department of Zoology and Genetics lowa State University
Ames, Iowa
| | | |
Collapse
|
5
|
Samantaray S, Das A, Matzelle DC, Yu SP, Wei L, Varma A, Ray SK, Banik NL. Administration of low dose estrogen attenuates persistent inflammation, promotes angiogenesis, and improves locomotor function following chronic spinal cord injury in rats. J Neurochem 2016; 137:604-17. [PMID: 26998684 DOI: 10.1111/jnc.13610] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 01/22/2016] [Accepted: 02/17/2016] [Indexed: 02/06/2023]
Abstract
Spinal cord injury (SCI) causes loss of neurological function and, depending upon the severity of injury, may lead to paralysis. Currently, no FDA-approved pharmacotherapy is available for SCI. High-dose methylprednisolone is widely used, but this treatment is controversial. We have previously shown that low doses of estrogen reduces inflammation, attenuates cell death, and protects axon and myelin in SCI rats, but its effectiveness in recovery of function is not known. Therefore, the goal of this study was to investigate whether low doses of estrogen in post-SCI would reduce inflammation, protect cells and axons, and improve locomotor function during the chronic phase of injury. Injury (40 g.cm force) was induced at thoracic 10 in young adult male rats. Rats were treated with 10 or 100 μg 17β-estradiol (estrogen) for 7 days following SCI and compared with vehicle-treated injury and laminectomy (sham) controls. Histology (H&E staining), immunohistofluorescence, Doppler laser technique, and Western blotting were used to monitor tissue integrity, gliosis, blood flow, angiogenesis, the expression of angiogenic factors, axonal degeneration, and locomotor function (Basso, Beattie, and Bresnahan rating) following injury. To assess the progression of recovery, rats were sacrificed at 7, 14, or 42 days post injury. A reduction in glial reactivity, attenuation of axonal and myelin damage, protection of cells, increased expression of angiogenic factors and microvessel growth, and improved locomotor function were found following estrogen treatment compared with vehicle-treated SCI rats. These results suggest that treatment with a very low dose of estrogen has significant therapeutic implications for the improvement of locomotor function in chronic SCI. Experimental studies with low dose estrogen therapy in chronic spinal cord injury (SCI) demonstrated the potential for multi-active beneficial outcomes that could ameliorate the degenerative pathways in chronic SCI as shown in (a). Furthermore, the alterations in local spinal blood flow could be significantly alleviated with low dose estrogen therapy. This therapy led to the preservation of the structural integrity of the spinal cord (b), which in turn led to the improved functional recovery as shown (c).
Collapse
Affiliation(s)
- Supriti Samantaray
- Department of Neurology and Neurosurgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Arabinda Das
- Department of Neurology and Neurosurgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Denise C Matzelle
- Department of Neurology and Neurosurgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Shan P Yu
- Department of Anesthesia, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Ling Wei
- Department of Anesthesia, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Abhay Varma
- Department of Neurology and Neurosurgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Swapan K Ray
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Naren L Banik
- Department of Neurology and Neurosurgery, Medical University of South Carolina, Charleston, South Carolina, USA.,Ralph H. Johnson Veterans Administration Medical Center, Charleston, South Carolina, USA
| |
Collapse
|
6
|
Fuhrmann A, Li J, Chien S, Engler AJ. Cation type specific cell remodeling regulates attachment strength. PLoS One 2014; 9:e102424. [PMID: 25014042 PMCID: PMC4094514 DOI: 10.1371/journal.pone.0102424] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 06/18/2014] [Indexed: 11/19/2022] Open
Abstract
Single-molecule experiments indicate that integrin affinity is cation-type-dependent, but in spread cells integrins are engaged in complex focal adhesions (FAs), which can also regulate affinity. To better understand cation-type-dependent adhesion in fully spread cells, we investigated attachment strength by application of external shear. While cell attachment strength is indeed modulated by cations, the regulation of integrin-mediated adhesion is also exceedingly complex, cell specific, and niche dependent. In the presence of magnesium only, fibroblasts and fibrosarcoma cells remodel their cytoskeleton to align in the direction of applied shear in an α5-integrin/fibronectin-dependent manner, which allows them to withstand higher shear. In the presence of calcium or on collagen in modest shear, fibroblasts undergo piecewise detachment but fibrosarcoma cells exhibit increased attachment strength. These data augment the current understanding of force-mediated detachment by suggesting a dynamic interplay in situ between cell adhesion and integrins depending on local niche cation conditions.
Collapse
Affiliation(s)
- Alexander Fuhrmann
- Department of Bioengineering, Institute of Engineering in Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Julie Li
- Department of Bioengineering, Institute of Engineering in Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Shu Chien
- Department of Bioengineering, Institute of Engineering in Medicine, University of California San Diego, La Jolla, California, United States of America
- Department of Medicine, Institute of Engineering in Medicine, University of California San Diego, La Jolla, California, United States of America
- Sanford Consortium for Regenerative Medicine, La Jolla, California, United States of America
| | - Adam J. Engler
- Department of Bioengineering, Institute of Engineering in Medicine, University of California San Diego, La Jolla, California, United States of America
- Sanford Consortium for Regenerative Medicine, La Jolla, California, United States of America
- * E-mail:
| |
Collapse
|
7
|
Hendricks BK, Shi R. Mechanisms of neuronal membrane sealing following mechanical trauma. Neurosci Bull 2014; 30:627-44. [PMID: 24993771 DOI: 10.1007/s12264-013-1446-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Accepted: 09/20/2013] [Indexed: 12/11/2022] Open
Abstract
Membrane integrity is crucial for maintaining the intricate signaling and chemically-isolated intracellular environment of neurons; disruption risks deleterious effects, such as unregulated ionic flux, neuronal apoptosis, and oxidative radical damage as observed in spinal cord injury and traumatic brain injury. This paper, in addition to a discussion of the current understanding of cellular tactics to seal membranes, describes two major factors involved in membrane repair. These are line tension, the hydrophobic attractive force between two lipid free-edges, and membrane tension, the rigidity of the lipid bilayer with respect to the tethered cortical cytoskeleton. Ca(2+), a major mechanistic trigger for repair processes, increases following flux through a membrane injury site, and activates phospholipase enzymes, calpain-mediated cortical cytoskeletal proteolysis, protein kinase cascades, and lipid bilayer microdomain modification. The membrane tension appears to be largely modulated through vesicle dynamics, cytoskeletal organization, membrane curvature, and phospholipase manipulation. Dehydration of the phospholipid gap edge and modification of membrane packaging, as in temperature variation, experimentally impact line tension. Due to the time-sensitive nature of axonal sealing, increasing the efficacy of axolemmal sealing through therapeutic modification would be of great clinical value, to deter secondary neurodegenerative effects. Better therapeutic enhancement of membrane sealing requires a complete understanding of its intricate underlying neuronal mechanism.
Collapse
Affiliation(s)
- Benjamin K Hendricks
- Department of Basic Medical Sciences, College of Veterinary Medicine, Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA
| | | |
Collapse
|
8
|
Bains W. Transglutaminse 2 and EGGL, the protein cross-link formed by transglutaminse 2, as therapeutic targets for disabilities of old age. Rejuvenation Res 2013; 16:495-517. [PMID: 23968147 PMCID: PMC3869435 DOI: 10.1089/rej.2013.1452] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Accepted: 08/22/2013] [Indexed: 12/17/2022] Open
Abstract
Aging of the extracellular matrix (ECM), the protein matrix that surrounds and penetrates the tissues and binds the body together, contributes significantly to functional aging of tissues. ECM proteins become increasingly cross-linked with age, and this cross-linking is probably important in the decline of the ECM's function. This article reviews the role of ε-(γ-glutamyl)-lysine (EGGL), a cross-link formed by transglutaminase enzymes, and particularly the widely expressed isozyme transglutaminase 2 (TG2), in the aging ECM. There is little direct data on EGGL accumulation with age, and no direct evidence of a role of EGGL in the aging of the ECM with pathology. However, several lines of circumstantial evidence suggest that EGGL accumulates with age, and its association with pathology suggests that this might reflect degradation of ECM function. TG activity increases with age in many circumstances. ECM protein turnover is such that some EGGL made by TG is likely to remain in place for years, if not decades, in healthy tissue, and both EGGL and TG levels are enhanced by age-related diseases. If further research shows EGGL does accumulate with age, removing it could be of therapeutic benefit. Also reviewed is the blockade of TG and active removal of EGGL as therapeutic strategies, with the conclusion that both have promise. EGGL removal may have benefit for acute fibrotic diseases, such as tendinopathy, and for treating generalized decline in ECM function with old age. Extracellular TG2 and EGGL are therefore therapeutic targets both for specific and more generalized diseases of aging.
Collapse
Affiliation(s)
- William Bains
- SRF Laboratory, Department of Chemical Engineering and Biotechnology, University of Cambridge , Cambridge, United Kingdom
| |
Collapse
|
9
|
Polyethylene glycol repairs membrane damage and enhances functional recovery: a tissue engineering approach to spinal cord injury. Neurosci Bull 2013; 29:460-6. [PMID: 23893430 DOI: 10.1007/s12264-013-1364-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Accepted: 06/14/2013] [Indexed: 10/26/2022] Open
Abstract
The integrity of the neuronal membrane is crucial for its function and cellular survival; thus, ineffective repair of damaged membranes may be one of the key elements underlying the neuronal degeneration and overall functional loss that occurs after spinal cord injury (SCI). it has been shown that polyethylene glycol (PEG) can reseal axonal membranes following various injuries in multiple in vitro and in vivo injury models. in addition, PEG may also directly prevent the effects of mitochondria-derived oxidative stress on intracellular components. Thus, PEG repairs mechanically injured cells by at least two distinct pathways: resealing of the disrupted plasma membrane and direct protection of mitochondria. Besides repairing primary membrane damage, PEG treatment also results in significant attenuation of oxidative stress, likely due to its capacity to reseal the membrane, thereby breaking the cycle of cellular damage and free-radical production. Based on this, in addition to the practicality of its application, we expect that PEG may be established as an effective treatment for SCI where membrane disruption and mitochondrial damage are implicated.
Collapse
|
10
|
Gurkoff G, Shahlaie K, Lyeth B, Berman R. Voltage-gated calcium channel antagonists and traumatic brain injury. Pharmaceuticals (Basel) 2013; 6:788-812. [PMID: 24276315 PMCID: PMC3816709 DOI: 10.3390/ph6070788] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Revised: 06/06/2013] [Accepted: 06/06/2013] [Indexed: 01/17/2023] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of death and disability in the United States. Despite more than 30 years of research, no pharmacological agents have been identified that improve neurological function following TBI. However, several lines of research described in this review provide support for further development of voltage gated calcium channel (VGCC) antagonists as potential therapeutic agents. Following TBI, neurons and astrocytes experience a rapid and sometimes enduring increase in intracellular calcium ([Ca2+]i). These fluxes in [Ca2+]i drive not only apoptotic and necrotic cell death, but also can lead to long-term cell dysfunction in surviving cells. In a limited number of in vitro experiments, both L-type and N-type VGCC antagonists successfully reduced calcium loads as well as neuronal and astrocytic cell death following mechanical injury. In rodent models of TBI, administration of VGCC antagonists reduced cell death and improved cognitive function. It is clear that there is a critical need to find effective therapeutics and rational drug delivery strategies for the management and treatment of TBI, and we believe that further investigation of VGCC antagonists should be pursued before ruling out the possibility of successful translation to the clinic.
Collapse
Affiliation(s)
- Gene Gurkoff
- Department of Neurological Surgery, One Shields Avenue, University of California, Davis, CA 95616, USA; E-Mails: (K.S.); (B.L.); (R.B.)
- NSF Center for Biophotonics Science and Technology, Suite 2700 Stockton Blvd, Suite 1400, Sacramento, CA, 95817, USA
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-530-754-7501; Fax: +1-530-754-5125
| | - Kiarash Shahlaie
- Department of Neurological Surgery, One Shields Avenue, University of California, Davis, CA 95616, USA; E-Mails: (K.S.); (B.L.); (R.B.)
| | - Bruce Lyeth
- Department of Neurological Surgery, One Shields Avenue, University of California, Davis, CA 95616, USA; E-Mails: (K.S.); (B.L.); (R.B.)
| | - Robert Berman
- Department of Neurological Surgery, One Shields Avenue, University of California, Davis, CA 95616, USA; E-Mails: (K.S.); (B.L.); (R.B.)
| |
Collapse
|
11
|
Majumder R, Koklic T, Rezaie AR, Lentz BR. Phosphatidylserine-induced factor Xa dimerization and binding to factor Va are competing processes in solution. Biochemistry 2013; 52:143-51. [PMID: 23214401 PMCID: PMC3544317 DOI: 10.1021/bi301239z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A soluble, short chain phosphatidylserine, 1,2-dicaproyl-sn-glycero-3-phospho-l-serine (C6PS), binds to discrete sites on FXa, FVa, and prothrombin to alter their conformations, to promote FXa dimerization (K(d) ~ 14 nM), and to enhance both the catalytic activity of FXa and the cofactor activity of FVa. In the presence of calcium, C6PS binds to two sites on FXa, one in the epidermal growth factor-like (EGF) domain and one in the catalytic domain; the latter interaction is sensitive to Na(+) binding and probably represents a protein recognition site. Here we ask whether dimerization of FXa and its binding to FVa in the presence of C6PS are competitive processes. We monitored FXa activity at 5, 20, and 50 nM FXa while titrating with FVa in the presence of 400 μM C6PS and 3 or 5 mM Ca(2+) to show that the apparent K(d) of FVa-FXa interaction increased with an increase in FXa concentration at 5 mM Ca(2+), but the K(d) was only slightly affected at 3 mM Ca(2+). A mixture of 50 nM FXa and 50 nM FVa in the presence of 400 μM C6PS yielded both Xa homodimers and Xa·Va heterodimers, but no FXa dimers bound to FVa. A mutant FXa (R165A) that has reduced prothrombinase activity showed both weakened dimerization (K(d) ~ 147 nM) and weakened FVa binding (apparent K(d) values of 58, 92, and 128 nM for 5, 20, and 50 nM R165A FXa, respectively). Native gel electrophoresis showed that the GLA-EGF(NC) fragment of FXa (lacking the catalytic domain) neither dimerized nor formed a complex with FVa in the presence of 400 μM C6PS and 5 mM Ca(2+). Our results demonstrate that the dimerization site and FVa-binding site are both located in the catalytic domain of FXa and that these sites are linked thermodynamically.
Collapse
Affiliation(s)
- Rinku Majumder
- Department of Biochemistry and Biophysics and Program in Molecular & Cellular Biophysics, CB # 7260, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599-7260
| | - Tilen Koklic
- Laboratory of Biophysics (EPR center), Jozef Stefan Institute, Jamova 39, 1000 Ljubljana, Slovenia
| | - Alireza R. Rezaie
- Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Saint Louis, MO-63104
| | - Barry R. Lentz
- Department of Biochemistry and Biophysics and Program in Molecular & Cellular Biophysics, CB # 7260, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599-7260
| |
Collapse
|
12
|
Sahota IS, Ravensbergen HRJC, McGrath MS, Claydon VE. Cerebrovascular responses to orthostatic stress after spinal cord injury. J Neurotrauma 2012; 29:2446-56. [PMID: 22720841 DOI: 10.1089/neu.2012.2379] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Orthostatic hypotension (OH) is a debilitating condition affecting individuals with spinal cord injury (SCI) that may be associated with cerebral hypoperfusion. We studied orthostatic cerebral control in individuals with SCI with different levels and severities of injury to spinal cardiovascular autonomic pathways. We measured beat-to-beat cardiovascular and cerebrovascular responses to passive orthostatic stress in 16 controls and 26 subjects with chronic SCI. Cerebrovascular control was assessed from diastolic cerebral blood flow velocity (CBFV(D)), and indices of static and dynamic cerebral autoregulation. Severity of autonomic injury was inferred from spectral analyses of systolic arterial pressure, and supine plasma noradrenaline concentrations. Symptoms of OH were evaluated using questionnaires. CBFV(D) decreased during orthostasis only in individuals with autonomically complete injuries above T6. Orthostatic CBFV(D) was significantly correlated (p<0.05) with the severity of autonomic injury. Individuals with injuries above T6 had impaired dynamic autoregulation (p<0.05) compared to controls, and this was correlated (p<0.05) with the severity of autonomic injury. Individuals with autonomically complete SCI reported increased severity of symptoms relative to controls (p<0.05). Symptom severity was correlated with the efficacy of dynamic autoregulation. During orthostatic stress, SCI individuals have impaired cerebrovascular control that is related to the level and severity of autonomic injury.
Collapse
Affiliation(s)
- Inderjeet S Sahota
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada
| | | | | | | |
Collapse
|
13
|
Imagama T, Ogino K, Takemoto K, Kato Y, Kataoka H, Suzuki H, Ran Z, Setiawan H, Fujikura Y, Taguchi T. Regulation of nitric oxide generation by up-regulated arginase I in rat spinal cord injury. J Clin Biochem Nutr 2012; 51:68-75. [PMID: 22798716 PMCID: PMC3391866 DOI: 10.3164/jcbn.d-11-00011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2011] [Accepted: 11/07/2011] [Indexed: 01/12/2023] Open
Abstract
Recently, arginase is suggested to regulate nitric oxide production by competing with nitric oxide synthase for the same substrate, L-arginine, in experimental asthma. We investigated the role of arginase and its relationship to nitric oxide production after spinal cord injury. Rats were subjected to laminectomy and complete transection of their spinal cords (injury group) or laminectomy only (sham group). In the injury group, arginase I was increased in the macrophages at the transection edge, and the peak was observed 48 h after spinal cord injury. However, nitric oxide production decreased significantly in the injury group despite increased nitric oxide synthase2 mRNA expression compared with the sham group. We also demonstrated the reduction in L-arginine concentrations, which was inversely associated with changes in arginase activity. Therefore, arginase appeared to regulate nitric oxide production by consuming L-arginine. The regulation of arginase activity and L-arginine levels may improve nitroxidative stress and reduce tissue damage in spinal cord injury.
Collapse
Affiliation(s)
- Takashi Imagama
- Department of Orthopaedic Surgery, Graduate School of Medicine, Yamaguchi University, 1-1-1 Minamikogushi, Ube, Yamaguchi 755-8505, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Chew DJ, Fawcett JW, Andrews MR. The challenges of long-distance axon regeneration in the injured CNS. PROGRESS IN BRAIN RESEARCH 2012. [PMID: 23186719 DOI: 10.1016/b978-0-444-59544-7.00013-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Injury to the central nervous system (CNS) that results in long-tract axonal damage typically leads to permanent functional deficits in areas innervated at, and below, the level of the lesion. The initial ischemia, inflammation, and neurodegeneration are followed by a progressive generation of scar tissue, dieback of transected axons, and demyelination, creating an area inhibitory to regrowth and recovery. Two ways to combat this inhibition is to therapeutically target the extrinsic and intrinsic properties of the axon-scar environment. Scar tissue within and surrounding the lesion site can be broken down using an enzyme known as chondroitinase. Negative regulators of adult neuronal growth, such as Nogo, can be neutralized with antibodies. Both therapies greatly improve functional recovery in animal models. Alternatively, modifying the intrinsic growth properties of CNS neurons through gene therapy or pharmacotherapy has also shown promising axonal regeneration after injury. Despite these promising therapies, the main challenge of long-distance axon regeneration still remains; achieving a level of functional and organized connectivity below the level of the lesion that mimics the intact CNS.
Collapse
Affiliation(s)
- Daniel J Chew
- Centre for Brain Repair, University of Cambridge, Forvie Site, Robinson Way, Cambridge, UK
| | | | | |
Collapse
|
15
|
Low dose estrogen prevents neuronal degeneration and microglial reactivity in an acute model of spinal cord injury: effect of dosing, route of administration, and therapy delay. Neurochem Res 2011; 36:1809-16. [PMID: 21611834 DOI: 10.1007/s11064-011-0498-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/05/2011] [Indexed: 10/18/2022]
Abstract
Spinal cord injury (SCI), depending on the severity of injury, leads to neurological dysfunction and paralysis. Methylprednisolone, the only currently available therapy renders limited protection in SCI. Therefore, other therapeutic agents must be tested to maximize neuroprotection and functional recovery. Previous data from our laboratory indicate that estrogen (17β-estradiol) at a high dose may attenuate multiple damaging pathways involved in SCI and improve locomotor outcome. Since use of high dose estrogen may have detrimental side effects and therefore may never be used in the clinic, the current study investigated the efficacy of this steroid hormone at very low doses in SCI. In particular, we tested the impact of dosing (1-10 μg/kg), mode of delivery (intravenous vs. osmotic pump), and delay in estrogen application (15 min-4 h post-SCI) on microgliosis and neuronal death in acute SCI in rats. Treatment with 17β-estradiol (1-10 μg/kg) significantly reduced microglial activation and also attenuated apoptosis of neurons compared to untreated SCI animals. The attenuation of cell death and inflammation by estrogen was observed regardless of mode and time of delivery following injury. These findings suggest estrogen as a potential agent for the treatment of individuals with SCI.
Collapse
|
16
|
Affiliation(s)
- John A Kessler
- Department of Neurology, Northwestern University, Chicago, Illinois 60611, USA.
| |
Collapse
|
17
|
Park J, Fan Z, Kumon RE, El-Sayed MEH, Deng CX. Modulation of intracellular Ca2+ concentration in brain microvascular endothelial cells in vitro by acoustic cavitation. ULTRASOUND IN MEDICINE & BIOLOGY 2010; 36:1176-87. [PMID: 20620704 PMCID: PMC3139909 DOI: 10.1016/j.ultrasmedbio.2010.04.006] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2009] [Revised: 03/04/2010] [Accepted: 04/14/2010] [Indexed: 05/08/2023]
Abstract
Localized delivery of therapeutic agents through the blood-brain barrier (BBB) is a clinically significant task that remains challenging. Ultrasound (US) application after intravenous administration of microbubbles has been shown to generate localized BBB opening in animal models but the detailed mechanisms are not yet fully described. The current study investigates the effects of US-stimulated microbubbles on in vitro murine brain microvascular endothelial (bEnd.3) cells by monitoring sonoporation and changes in intracellular calcium concentration ([Ca(2+)](i)) using real-time fluorescence and high-speed brightfield microscopy. Cells seeded in microchannels were exposed to a single US pulse (1.25 MHz, 10 cycles, 0.24 MPa peak negative pressure) in the presence of Definity microbubbles and extracellular calcium concentration [Ca(2+)](o) = 0.9 mM. Disruption of the cell membrane was assessed using propidium iodide (PI) and change in the [Ca(2+)](i) was measured using fura-2. Cells adjacent to a microbubble exhibited immediate [Ca(2+)](i) changes after US pulse with and without PI uptake and the [Ca(2+)](i) changes were twice as large in cells with PI uptake. Cell viability assays showed that sonoporated cells could survive with modulation of [Ca(2+)](i) and uptake of PI. Cells located near sonoporated cells were observed to exhibit changes in [Ca(2+)](i) that were delayed from the time of US application and without PI uptake. These results demonstrate that US-stimulated microbubbles not only directly cause changes in [Ca(2+)](i) in brain endothelial cells in addition to sonoporation but also generate [Ca(2+)](i) transients in cells not directly interacting with microbubbles, thereby affecting cells in larger regions beyond the cells in contact with microbubbles.
Collapse
Affiliation(s)
| | | | | | | | - Cheri X. Deng
- Address correspondence to: Cheri X. Deng, Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel Blvd, Ann Arbor, MI 48109–2099, USA. Tel: +1 734-936-2855; Fax: +1 734-936-1905.
| |
Collapse
|
18
|
Abstract
Stem cell therapy is a potential treatment for spinal cord injury (SCI), and a variety of different stem cell types have been evaluated in animal models and humans with SCI. No consensus exists regarding the type of stem cell, if any, that will prove to be effective therapeutically. Most data suggest that no single therapy will be sufficient to overcome all the biological complications caused by SCI. Rationales for therapeutic use of stem cells for SCI include replacement of damaged neurons and glial cells, secretion of trophic factors, regulation of gliosis and scar formation, prevention of cyst formation, and enhancement of axon elongation. Most therapeutic approaches that use stem cells involve implantation of these cells into the spinal cord. The attendant risks of stem cell therapy for SCI--including tumor formation, or abnormal circuit formation leading to dysfunction--must be weighed against the potential benefits of this approach. This Review will examine the biological effects of SCI, the opportunities for stem cell treatment, and the types of stem cells that might be used therapeutically. The limited information available on the possible benefits of stem cell therapy to humans will also be discussed.
Collapse
Affiliation(s)
- Vibhu Sahni
- MGH-HMS Center for Nervous System Repair, Harvard Medical School, 50 Blossom Street, Boston, MA 02114, USA
| | | |
Collapse
|
19
|
Nehrt A, Hamann K, Ouyang H, Shi R. Polyethylene Glycol Enhances Axolemmal Resealing following Transection in Cultured Cells and in Ex Vivo Spinal Cord. J Neurotrauma 2010; 27:151-61. [PMID: 19691421 DOI: 10.1089/neu.2009.0993] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Affiliation(s)
- Ashley Nehrt
- Center for Paralysis Research, Department of Basic Medical Sciences, School of Veterinary Medicine, Purdue University, West Lafayette, Indiana
| | - Kristin Hamann
- Center for Paralysis Research, Department of Basic Medical Sciences, School of Veterinary Medicine, Purdue University, West Lafayette, Indiana
| | - Hui Ouyang
- Center for Paralysis Research, Department of Basic Medical Sciences, School of Veterinary Medicine, Purdue University, West Lafayette, Indiana
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana
| | - Riyi Shi
- Center for Paralysis Research, Department of Basic Medical Sciences, School of Veterinary Medicine, Purdue University, West Lafayette, Indiana
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana
| |
Collapse
|
20
|
Al-Qudah KM, Al-Majali AM. Higher Lipid Peroxidation Indices in Horses Eliminated from Endurance Race Because of Synchronous Diaphragmatic Flutter (Thumps). J Equine Vet Sci 2008. [DOI: 10.1016/j.jevs.2008.08.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
21
|
Wang QS, Hou LY, Zhang CL, Zhao XL, Yu SF, Xie KQ. 2,5-hexanedione (HD) treatment alters calmodulin, Ca2+/calmodulin-dependent protein kinase II, and protein kinase C in rats' nerve tissues. Toxicol Appl Pharmacol 2008; 232:60-8. [DOI: 10.1016/j.taap.2008.05.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2007] [Revised: 04/30/2008] [Accepted: 05/19/2008] [Indexed: 11/29/2022]
|
22
|
Lirk P, Poroli M, Rigaud M, Fuchs A, Fillip P, Huang CY, Ljubkovic M, Sapunar D, Hogan Q. Modulators of calcium influx regulate membrane excitability in rat dorsal root ganglion neurons. Anesth Analg 2008; 107:673-85. [PMID: 18633052 DOI: 10.1213/ane.0b013e31817b7a73] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Chronic neuropathic pain resulting from neuronal damage remains difficult to treat, in part, because of incomplete understanding of underlying cellular mechanisms. We have previously shown that inward Ca2+ flux (I(Ca)) across the sensory neuron plasmalemma is decreased in a rodent model of chronic neuropathic pain, but the direct consequence of this loss of I(Ca) on function of the sensory neuron has not been defined. We therefore examined the extent to which altered membrane properties after nerve injury, especially increased excitability that may contribute to chronic pain, are attributable to diminished Ca2+ entry. METHODS Intracellular microelectrode measurements were obtained from A-type neurons of dorsal root ganglia excised from uninjured rats. Recording conditions were varied to suppress or promote I(Ca) while biophysical variables and excitability were determined. RESULTS Both lowered external bath Ca2+ concentration and blockade of I(Ca) with bath cadmium diminished the duration and area of the after-hyperpolarization (AHP), accompanied by decreased current threshold for action potential (AP) initiation and increased repetitive firing during sustained depolarization. Reciprocally, elevated bath Ca2+ increased the AHP and suppressed repetitive firing. Voltage sag during neuronal hyperpolarization, indicative of the cation-nonselective H-current, diminished with decreased bath Ca2+, cadmium application, or chelation of intracellular Ca2+. Additional recordings with selective blockers of I(Ca) subtypes showed that N-, P/Q, L-, and R-type currents each contribute to generation of the AHP and that blockade of any of these, and the T-type current, slows the AP upstroke, prolongs the AP duration, and (except for L-type current) decreases the current threshold for AP initiation. CONCLUSIONS Taken together, our findings show that suppression of I(Ca) decreases the AHP, reduces the hyperpolarization-induced voltage sag, and increases excitability in sensory neurons, replicating changes that follow peripheral nerve trauma. This suggests that the loss of I(Ca) previously demonstrated in injured sensory neurons contributes to their dysfunction and hyperexcitability, and may lead to neuropathic pain.
Collapse
Affiliation(s)
- Philipp Lirk
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Erol FS, Kaplan M, Tiftikci M, Yakar H, Ozercan I, Ilhan N, Topsakal C. Comparison of the effects of octreotide and melatonin in preventing nerve injury in rats with experimental spinal cord injury. J Clin Neurosci 2008; 15:784-90. [DOI: 10.1016/j.jocn.2007.06.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2007] [Accepted: 06/01/2007] [Indexed: 11/28/2022]
|
24
|
Nehrt A, Rodgers R, Shapiro S, Borgens R, Shi R. The critical role of voltage-dependent calcium channel in axonal repair following mechanical trauma. Neuroscience 2007; 146:1504-12. [PMID: 17448606 PMCID: PMC2701192 DOI: 10.1016/j.neuroscience.2007.02.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2006] [Revised: 02/07/2007] [Accepted: 02/09/2007] [Indexed: 11/30/2022]
Abstract
Membrane disruption following mechanical injury likely plays a critical role in the pathology of spinal cord trauma. It is known that intracellular calcium is a key factor that is essential to membrane resealing. However, the differential role of calcium influx through the injury site and through voltage dependent calcium channels (VDCC) has not been examined in detail. Using a well-established ex vivo guinea-pig spinal cord white matter preparation, we have found that axonal membrane resealing was significantly inhibited following transection or compression in the presence of cadmium, a non-specific calcium channel blocker, or nimodipine, a specific L-type calcium channel blocker. Membrane resealing was assessed by the changes of membrane potential and compound action potential (CAP), and exclusion of horseradish peroxidase 60 min following trauma. Furthermore, 1 microM BayK 8644, a VDCC agonist, significantly enhanced membrane resealing. Interestingly, this effect was completely abolished when the concentration of BayK 8644 was increased to 30 microM. These data suggest that VDCC play a critical role in membrane resealing. Further, there is likely an appropriate range of calcium influx through VDCC which ensures effective axonal membrane resealing. Since elevated intracellular calcium has also been linked to axonal deterioration, blockage of VDCC is proposed to be a clinical treatment for various injuries. The knowledge gained in this study will likely help us better understand the role of calcium in various CNS trauma, which is critical for designing new approaches or perhaps optimizing the effectiveness of existing methods in the treatment of CNS trauma.
Collapse
Affiliation(s)
- Ashley Nehrt
- Center for Paralysis Research, Department of Basic Medical Sciences, Weldon School of Biomedical Engineering, Purdue University
| | - Richard Rodgers
- Department of Neurosurgery, School of Medicine, Indiana University
| | - Scott Shapiro
- Department of Neurosurgery, School of Medicine, Indiana University
| | - Richard Borgens
- Center for Paralysis Research, Department of Basic Medical Sciences, Weldon School of Biomedical Engineering, Purdue University
| | - Riyi Shi
- Center for Paralysis Research, Department of Basic Medical Sciences, Weldon School of Biomedical Engineering, Purdue University
| |
Collapse
|
25
|
Folkerts MM, Parks EA, Dedman JR, Kaetzel MA, Lyeth BG, Berman RF. Phosphorylation of Calcium Calmodulin—Dependent Protein Kinase II following Lateral Fluid Percussion Brain Injury in Rats. J Neurotrauma 2007; 24:638-50. [PMID: 17439347 DOI: 10.1089/neu.2006.0188] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Traumatic brain injury (TBI) can dramatically increase levels of intracellular calcium ([Ca(2+)](i)). One consequence of increased [Ca(2+)](i) would be altered activity and function of calcium-regulated proteins, including calcium-calmodulin-dependent protein kinase II (CaMKII), which is autophosphorylated on Thr(286)(pCaMKII(286)) in the presence of calcium and calmodulin. Therefore, we hypothesized that TBI would result in increased levels of pCaMKII(286), and that such increases would occur early after injury in brain regions known to be damaged following lateral fluid percussion TBI (i.e., hippocampus and cortex). In order to test this hypothesis, immunostaining of CaMKII was examined in rat hippocampus and cortex after lateral fluid percussion (LFP) injury using an antibody directed against pCaMKII(286). LFP injury produced a marked increase in pCaMKII(286) immunostaining in the hippocampus and overlying cortex 30 min after TBI. The pattern of increased immunostaining was uneven, and unexpectedly absent in some hippocampal CA3 pyramidal neurons. This suggests that phosphatase activity may also increase following TBI, resulting in dephosphorylation of pCaMKII(286) in subpopulations of CA3 pyramidal neurons. Western blotting confirmed a rapid increase in levels of pCaMKII(286) at 10 and 30 min after brain injury, and that it was transient and no longer significantly elevated when examined at 3, 8, and 24 h. These results demonstrate that TBI alters the autophosphorylation state of CaMKII, an important neuronal regulator of critical cell functions, including enzyme activities, cell structure, gene expression, and neuronal plasticity, and provide a molecular mechanism that is likely to contribute to cell injury and impaired plasticity after TBI.
Collapse
Affiliation(s)
- Michael M Folkerts
- Department of Neurological Surgery, University of California Davis, Davis, California 95616, USA
| | | | | | | | | | | |
Collapse
|
26
|
Xiong Y, Rabchevsky AG, Hall ED. Role of peroxynitrite in secondary oxidative damage after spinal cord injury. J Neurochem 2007; 100:639-49. [PMID: 17181549 DOI: 10.1111/j.1471-4159.2006.04312.x] [Citation(s) in RCA: 153] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Peroxynitrite (PON, ONOO(-)), formed by nitric oxide synthase-generated nitric oxide radical ( NO) and superoxide radical (O(2) (-)), is a crucial player in post-traumatic oxidative damage. In the present study, we determined the spatial and temporal characteristics of PON-derived oxidative damage after a moderate contusion injury in rats. Our results showed that 3-nitrotyrosine (3-NT), a specific marker for PON, rapidly accumulated at early time points (1 and 3 h) and a significant increase compared with sham rats was sustained to 1 week after injury. Additionally, there was a coincident and maintained increase in the levels of protein oxidation-related protein carbonyl and lipid peroxidation-derived 4-hydroxynonenal (4-HNE). The peak increases of 3-NT and 4-HNE were observed at 24 h post-injury. In our immunohistochemical results, the co-localization of 3-NT and 4-HNE results indicates that PON is involved in lipid peroxidative as well as protein nitrative damage. One of the consequences of oxidative damage is an exacerbation of intracellular calcium overload, which activates the cysteine protease calpain leading to the degradation of several cellular targets including cytoskeletal protein (alpha-spectrin). Western blot analysis of alpha-spectrin breakdown products showed that the 145-kDa fragments of alpha-spectrin, which are specifically generated by calpain, were significantly increased as soon as 1 h following injury although the peak increase did not occur until 72 h post-injury. The later activation of calpain is most likely linked to PON-mediated secondary oxidative impairment of calcium homeostasis. Scavengers of PON, or its derived free radical species, may provide an improved antioxidant neuroprotective approach for the treatment of post-traumatic oxidative damage in the injured spinal cord.
Collapse
Affiliation(s)
- Yiqin Xiong
- Spinal Cord and Brain Injury Research Center, University of Kentucky Chandler Medical Center, Lexington, KY 40536, USA
| | | | | |
Collapse
|
27
|
Osuka K, Watanabe Y, Usuda N, Atsuzawa K, Aoshima C, Yamauchi K, Takayasu M, Yoshida J. Phosphorylation of neuronal nitric oxide synthase at Ser847 in the nucleus intermediolateralis after spinal cord injury in mice. Neuroscience 2007; 145:241-7. [PMID: 17258865 DOI: 10.1016/j.neuroscience.2006.10.034] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2006] [Revised: 10/05/2006] [Accepted: 10/26/2006] [Indexed: 11/24/2022]
Abstract
We previously demonstrated that Ca2+/calmodulin (CaM)-dependent protein kinase IIalpha (CaM-KIIalpha) can phosphorylate neuronal nitric oxide synthase (nNOS) at Ser847 and attenuate NOS activity in neuronal cells. In the present study we focused on chronological alteration in levels and cellular location of nNOS, phosphorylated (p)-Ser847-nNOS (NP847), CaM-KII and p-Thr286-CaM-KIIalpha following spinal cord injury (SCI) in mice. Western blot analysis showed nNOS to be significantly phosphorylated at Ser847 from 3 h after SCI, peaking at 24 h and gradually decreasing thereafter, and CaM-KII to be colocalized with nNOS after SCI. Immunohistochemical analysis revealed that SCI causes an increase in both NP847 and p-Thr286-CaM-KIIalpha in the nucleus intermediolateralis. These findings suggest that SCI induces p-Thr286-CaM-KIIalpha, which phosphorylates the nNOS at Ser847 in the nucleus intermediolateralis where NO is thought to play a role as a neurotransmitter in autonomic preganglionic neurons. Thus, the NP847 signaling pathway might be involved in the autonomic failure which occurs immediately after SCI.
Collapse
Affiliation(s)
- K Osuka
- Department of Neurosurgery, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa, Nagoya, 466-8550, Japan
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
Among seven members of P2X ionotropic receptors activated by extracellular ATP, the P2X(7) subtype is unique in that it can function as a cation channel, a nonselective pore, or even a signaling complex coupled with multiple downstream components. Several roles of P2X(7) receptors have been described in CNS cells in the past decade, including release of cytokines and transmitters, modulation of presynaptic transmitter release, and activation of multiple signaling pathways. The finding that P2X(7) pores may directly mediate efflux of cytosolic glutamate, GABA, and ATP in glial cells is particularly interesting, as it provides a novel mechanism of glial transmitter release that may play important roles not only in physiological intercellular communication but also in pathological neural injury.
Collapse
Affiliation(s)
- Shumin Duan
- Institute of Neuroscience and Key Laboratory of Neurobiology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Joseph T Neary
- Research Service, Miami VA Medical Center, Departments of Pathology, Biochemistry and Molecular Biology, and Neuroscience Program, University of Miami School of Medicine, Miami, Florida
| |
Collapse
|
29
|
Usul H, Cakir E, Arslan E, Peksoylu B, Alver A, Sayin OC, Topbas M, Baykal S. Effects of Clotrimazole on Experimental Spinal Cord Injury. Arch Med Res 2006; 37:571-5. [PMID: 16740425 DOI: 10.1016/j.arcmed.2005.11.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2005] [Accepted: 11/16/2006] [Indexed: 10/24/2022]
Abstract
BACKGROUND We examined a possible neuroprotective effect of clotrimazole on spinal cord clip compression injury. METHODS Rivlin and Tator's acute extradural clip compression injury (CCI) model was used for producing SCI on 24 albino Wistar rats weighing 180-250 g. All rats were anesthetized with 30 mg/kg ketamine HCl intraperitoneally and were breathing spontaneously without tracheal intubation. Total laminectomy of T8-T12 was performed on all rats under operation microscope, and CCI was performed on all rats (expect those in group 1) with a 50-g closing force aneurysm clip for 1 min. Three hours later, all of the rats were killed with sodium pentobarbital. Spinal cords were excised for a length of 2 cm; 1 cm rostrally and caudally to the injury site and deep frozen at -76 degrees C for biochemical studies. RESULTS Treatment with clotrimazole decreased MDA levels in rats with SCI with a statistically significant difference. CONCLUSIONS To our knowledge, this the first study that shows the effects of clotrimazole on spinal cord clip compression injury. Clotrimazole was found to be effective on spinal cord clip compression injury, but further investigations are mandatory.
Collapse
Affiliation(s)
- Haydar Usul
- Department of Neurosurgery, Karadeniz Technical University School of Medicine, Trabzon, Turkey
| | | | | | | | | | | | | | | |
Collapse
|
30
|
BANIK NL, SHIELDS DC, RAY S, DAVIS B, MATZELLE D, WILFORD G, HOGAN EL. Role of Calpain in Spinal Cord Injury: Effects of Calpain and Free Radical Inhibitorsa. Ann N Y Acad Sci 2006; 844:131-137. [DOI: 10.1111/j.1749-6632.1998.tb08228.x] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
31
|
Chesler M. Failure and function of intracellular pH regulation in acute hypoxic-ischemic injury of astrocytes. Glia 2005; 50:398-406. [PMID: 15846798 DOI: 10.1002/glia.20141] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Astrocytes can die rapidly following ischemic and traumatic injury to the CNS. Brain acid-base status has featured prominently in theories of acute astrocyte injury. Failure of astrocyte pH regulation can lead to cell loss under conditions of severe acidosis. By contrast, the function of astrocyte pH regulatory mechanisms appears to be necessary for acute cell death following the simulation of transient ischemia and reperfusion. Severe lactic acidosis, and the failure of astrocytes to regulate intracellular pH (pH(i)) have been emphasized in brain ischemia under hyperglycemic conditions. Direct measurements of astrocyte pH(i) after cardiac arrest demonstrated a mean pH(i) of 5.3 in hyperglycemic rats. In addition, both in vivo and in vitro studies of astrocytes have shown similar pH levels to be cytotoxic. Whereas astrocytes exposed to hypoxia alone may require 12-24 h to die, acidosis has been found to exacerbate and speed hypoxic loss of these cells. Recently, astrocyte cultures were exposed to hypoxic, acidic media in which the large ionic perturbations characteristic of brain ischemia were simulated. Upon return to normal saline ("reperfusion"), the majority of cells died. This injury was dependent on external Ca2+ and was prevented by inhibition of reversed Na(+)-Ca2+ exchange, blockade of Na(+)-H+ exchange, or by low pH of the reperfusion saline. These data suggested that cytotoxic elevation of [Ca2+]i occurred during reperfusion due to a sequence of activated Na(+)-H+ exchange, cytosolic Na+ loading, and resultant reversal of Na(+)-Ca2+ exchange. The significance of this reperfusion model to ischemic astrocyte injury in vivo is discussed.
Collapse
Affiliation(s)
- Mitchell Chesler
- Department of Neurosurgery and Department of Physiology and Neuroscience, New York University School of Medicine, New York, New York
| |
Collapse
|
32
|
Yezierski RP. Spinal Cord Injury: A Model of Central Neuropathic Pain. Neurosignals 2005; 14:182-93. [PMID: 16215301 DOI: 10.1159/000087657] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2005] [Indexed: 01/05/2023] Open
Abstract
The condition of pain after spinal cord injury (SCI) affects the life quality of nearly 70% of individuals with SCI. Clinical studies over the past decade have provided important insights into the complexities of the clinical and psychosocial characteristics of this debilitating consequence of SCI. The use of experimental models developed to study at-level or below-level pain has provided an appreciation for the mechanism(s) responsible for the onset and progression of these conditions. Important to the studies related to SCI pain has been the focus on the molecular, biochemical, anatomical, and functional consequences of SCI that have identified potential therapeutic targets for the design of novel treatment strategies.
Collapse
Affiliation(s)
- Robert P Yezierski
- Comprehensive Center for Pain Research and the McKnight Brain Institute University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
33
|
Wang X, Arcuino G, Takano T, Lin J, Peng WG, Wan P, Li P, Xu Q, Liu QS, Goldman SA, Nedergaard M. P2X7 receptor inhibition improves recovery after spinal cord injury. Nat Med 2004; 10:821-7. [PMID: 15258577 DOI: 10.1038/nm1082] [Citation(s) in RCA: 404] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2004] [Accepted: 06/25/2004] [Indexed: 01/29/2023]
Abstract
Secondary injury exacerbates the extent of spinal cord insults, yet the mechanistic basis of this phenomenon has largely been unexplored. Here we report that broad regions of the peritraumatic zone are characterized by a sustained process of pathologic, high ATP release. Spinal cord neurons expressed P2X7 purine receptors (P2X7R), and exposure to ATP led to high-frequency spiking, irreversible increases in cytosolic calcium and cell death. To assess the potential effect of P2X7R blockade in ameliorating acute spinal cord injury (SCI), we delivered P2X7R antagonists OxATP or PPADS to rats after acute impact injury. We found that both OxATP and PPADS significantly improved functional recovery and diminished cell death in the peritraumatic zone. These observations demonstrate that SCI is associated with prolonged purinergic receptor activation, which results in excitotoxicity-based neuronal degeneration. P2X7R antagonists inhibit this process, reducing both the histological extent and functional sequelae of acute SCI.
Collapse
Affiliation(s)
- Xiaohai Wang
- Department of Neurosurgery, Center for Aging and Developmental Biology, University of Rochester Medical Center, Rochester, New York 14642, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Araújo Couto L, Sampaio Narciso M, Hokoç JN, Blanco Martinez AM. Calpain inhibitor 2 prevents axonal degeneration of opossum optic nerve fibers. J Neurosci Res 2004; 77:410-9. [PMID: 15248297 DOI: 10.1002/jnr.20170] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The ultrastructural change that characterizes the onset of Wallerian degeneration is the disintegration of axoplasmic microtubules and neurofilaments, which are converted into an amorphous and granular material, followed by myelin breakdown. The mechanism underlying such processes is an increase in the amount of intracellular calcium, leading to activation of proteases called calpains. The aim of this study was to evaluate by quantitative ultrastructural analysis whether nerve fibers can be preserved by the use of an exogenous inhibitor of these proteases (calpain inhibitor-2, Mu-F-hF-FMK), after optic nerve crush. For that, the left optic nerves of opossums, Didelphis aurita, were crushed with the aid of a fine forceps, and half of them received a calpain inhibitor mixed with Elvax resin. Ninety-six hours after the lesion, the animals were reanesthetized and transcardially perfused, and the optic nerves were removed, the right ones being used as normal nerves. Afterward, the optic nerves were dissected and processed for routine transmission electron microscopy and quantitative and statistical analysis. The results of this analysis showed that the group that received the calpain inhibitor presented a reduction of astrogliosis, maintaining the optic nerve structure in an organized state; a significant decrease in the number of degenerating fibers; and a significant increase in the number of fibers with preserved cytoskeleton and preservation of axonal and myelin area and integrity, reducing the enlargement and edema of the axon. In conclusion, our findings suggest that calpain inhibitor is able to provide neuroprotection of the central nervous system fibers after a crush lesion.
Collapse
Affiliation(s)
- Luciana Araújo Couto
- Departamento de Histologia e Embriologia, Instituto de Ciências Biomédicas, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | | | | | | |
Collapse
|
35
|
Usul H, Cakir E, Cobanoglu U, Alver A, Peksoylu B, Topbas M, Baykal S. The effects of tyrphostine Ag 556 on experimental spinal cord ischemia reperfusion injury. ACTA ACUST UNITED AC 2004; 61:45-54; discussion 54. [PMID: 14706378 DOI: 10.1016/s0090-3019(03)00539-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND To investigate the effects of Tyrphostin Ag 556 on spinal cord ischemia reperfusion injury. METHODS The inhibition of tyrosine kinase may represent a novel approach in the treatment of spinal cord ischemia reperfusion injury. Recently, a family of tyrosine kinase inhibitors, the tyrphostins, has been successfully used in models of endotoxemia, peritonitis, and hypovolemic shock. MATERIALS AND METHODS Twenty-four Wistar rats were used in the study. Rats were divided into 4 groups of 6 animals. The groups were named as sham operated group, injury group, vehicle group, and treatment group. Clamping of the abdominal aorta was performed for 45 minutes with all of the groups except sham-operated group. All of the rats were sacrificed 24 hours after the operation for biochemical and ultrastructural studies. RESULTS Tyrphostin Ag 556 treatment was found effective on experimental spinal cord ischemia reperfusion injury. The Malondialdehyde (MDA) values of the treatment group were statistically significant lower then the other reperfusion injury groups. The histologic examination showed better cellular structure in the treatment group than the other reperfusion injury groups. The neurologic scores of the treatment group also improved after treatment. CONCLUSIONS Tyrphostin Ag 556 alters spinal cord ischemia reperfusion injury by inhibiting protein kinases. Further investigations will be required to determine the long-term effects of this drug.
Collapse
Affiliation(s)
- Haydar Usul
- Departments of Neurosurgery, Trabzon, Turkey
| | | | | | | | | | | | | |
Collapse
|
36
|
Ray SK, Hogan EL, Banik NL. Calpain in the pathophysiology of spinal cord injury: neuroprotection with calpain inhibitors. BRAIN RESEARCH. BRAIN RESEARCH REVIEWS 2003; 42:169-85. [PMID: 12738057 DOI: 10.1016/s0165-0173(03)00152-8] [Citation(s) in RCA: 149] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Spinal cord injury (SCI) evokes an increase in intracellular free Ca(2+) level resulting in activation of calpain, a Ca(2+)-dependent cysteine protease, which cleaves many cytoskeletal and myelin proteins. Calpain is widely expressed in the central nervous system (CNS) and regulated by calpastatin, an endogenous calpain-specific inhibitor. Calpastatin degraded by overactivation of calpain after SCI may lose its regulatory efficiency. Evidence accumulated over the years indicates that uncontrolled calpain activity mediates the degradation of many cytoskeletal and membrane proteins in the course of neuronal death and contributes to the pathophysiology of SCI. Cleavage of the key cytoskeletal and membrane proteins by calpain is an irreversible process that perturbs the integrity and stability of CNS cells leading to cell death. Calpain in conjunction with caspases, most notably caspase-3, can cause apoptosis of the CNS cells following trauma. Aberrant Ca(2+) homeostasis following SCI inevitably activates calpain, which has been shown to play a crucial role in the pathophysiology of SCI. Therefore, calpain appears to be a potential therapeutic target in SCI. Substantial research effort has been focused upon the development of highly specific inhibitors of calpain and caspase-3 for therapeutic applications. Administration of cell permeable and specific inhibitors of calpain and caspase-3 in experimental animal models of SCI has provided significant neuroprotection, raising the hope that humans suffering from SCI may be treated with these inhibitors in the near future.
Collapse
Affiliation(s)
- Swapan K Ray
- Department of Neurology, Medical University of South Carolina, 96 Jonathan Lucas Street, Suite 309, P.O. Box 250606, Charleston, SC 29425, USA
| | | | | |
Collapse
|
37
|
Chernoff EAG, Stocum DL, Nye HLD, Cameron JA. Urodele spinal cord regeneration and related processes. Dev Dyn 2003; 226:295-307. [PMID: 12557207 DOI: 10.1002/dvdy.10240] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Urodele amphibians, newts and salamanders, can regenerate lesioned spinal cord at any stage of the life cycle and are the only tetrapod vertebrates that regenerate spinal cord completely as adults. The ependymal cells play a key role in this process in both gap replacement and caudal regeneration. The ependymal response helps to produce a different response to neural injury compared with mammalian neural injury. The regenerating urodele cord produces new neurons as well as supporting axonal regrowth. It is not yet clear to what extent urodele spinal cord regeneration recapitulates embryonic anteroposterior and dorsoventral patterning gene expression to achieve functional reconstruction. The source of axial patterning signals in regeneration would be substantially different from those in developing tissue, perhaps with signals propagated from the stump tissue. Examination of the effects of fibroblast growth factor and epidermal growth factor on ependymal cells in vivo and in vitro suggest a connection with neural stem cell behavior as described in developing and mature mammalian central nervous system. This review coordinates the urodele regeneration literature with axial patterning, stem cell, and neural injury literature from other systems to describe our current understanding and assess the gaps in our knowledge about urodele spinal cord regeneration.
Collapse
Affiliation(s)
- Ellen A G Chernoff
- Indiana University-Purdue University Indianapolis, Department of Biology, and Indiana University Center for Regenerative Biology and Medicine, Indianapolis, Indiana 46202, USA.
| | | | | | | |
Collapse
|
38
|
Beattie MS, Hermann GE, Rogers RC, Bresnahan JC. Cell death in models of spinal cord injury. PROGRESS IN BRAIN RESEARCH 2002; 137:37-47. [PMID: 12440358 DOI: 10.1016/s0079-6123(02)37006-7] [Citation(s) in RCA: 325] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Current treatments for acute spinal cord injury are based on animal models of human spinal cord injury (SCI). These models have shown that the initial traumatic injury to cord tissue is followed by a long period of secondary injury that includes a number of cellular and biochemical cascades. These secondary injury processes are potential targets for therapies. Continued refinement of rat and mouse models of SCI, along with more detailed analyses of the biology of the lesion in these models, points to both necrotic and apoptotic mechanisms of cell death after SCI. In this chapter, we review recent evidence for long-term apoptotic death of oligodendrocytes in long tracts undergoing Wallerian degeneration following SCI. This process appears to be related closely to activation of microglial cells. It is has been thought that microglial cells might be the source of cytotoxic cytokines, such as tumor necrosis factor-alpha (TNF-alpha), that kill oligodendrocytes. However, more recent evidence in vivo suggests that TNF-alpha by itself may not induce necrosis or apoptosis in oligodendrocytes. We review data that suggests other possible pathways for apoptosis, such as the neurotrophin receptor p75 which is expressed in both neurons and oligodendrocytes after SCI in rats and mice. In addition, it appears that microglial activation and TNF-alpha may be important in acute SCI. Ninety minutes after a moderate contusion lesion, microglia are activated and surround dying neurons. In an 'atraumatic' model of SCI, we have now shown that TNF-alpha appears to greatly potentiate cell death mediated by glutamate receptors. These studies emphasize that multiple mechanisms and interactions contribute to secondary injury after SCI. Continued study of both contusion models and other new approaches to studying these mechanisms will be needed to maximize strategies for acute and chronic therapies, and for neural repair.
Collapse
Affiliation(s)
- Michael S Beattie
- Department of Neuroscience, The Ohio State University Medical Center, 333 W. 10th Avenue, Columbus, OH 43210, USA.
| | | | | | | |
Collapse
|
39
|
Nakahara S, Yone K, Setoguchi T, Yamaura I, Arishima Y, Yoshino S, Komiya S. Changes in nitric oxide and expression of nitric oxide synthase in spinal cord after acute traumatic injury in rats. J Neurotrauma 2002; 19:1467-74. [PMID: 12490011 DOI: 10.1089/089771502320914697] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The aim of this study was to observe the time course of NO production and NOS expression in the spinal cord following acute traumatic injury. Rat spinal cord was injured by extradural static weight-compression, which resulted in an incomplete transverse spinal cord lesion with paralysis of the lower extremities. Using this model, measurement of NO by microdialysis and Griess reaction and histological and immunohistochemical examinations using polyclonal antibodies to nNOS and iNOS were performed from immediately to 14 days after injury. In injured cord, the amount of NO markedly increased immediately after injury and gradually decreased between 1 and 12 h after injury. A second wave of increase in NO level was observed at 24 h and 3 days after injury. Histologically, hematomas and necrotic changes were observed after injury and demyelination of nerve fibers increased with time in the compressed segment. Immunohistochemically, the number of cells with expression of nNOS was increased immediately to 12 h after injury. Expression of iNOS was observed from 12 h to 3 days after injury. These findings suggested that the initial maximal increase of NO production might be caused mainly by nNOS and that the second wave of increase in NO might be due mainly to iNOS.
Collapse
Affiliation(s)
- Shinji Nakahara
- Department of Orthopaedic Surgery, Faculty of Medicine, Kagoshima University, Kagoshima, Japan
| | | | | | | | | | | | | |
Collapse
|
40
|
Rosenberg LJ, Wrathall JR. Time course studies on the effectiveness of tetrodotoxin in reducing consequences of spinal cord contusion. J Neurosci Res 2001; 66:191-202. [PMID: 11592114 DOI: 10.1002/jnr.1211] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Focal injection of the sodium channel blocker tetrodotoxin (TTX) into the injury site at either 5 or 15 min after a standardized thoracic contusion spinal cord injury (SCI) reduces white matter pathology and loss of axons in the first 24 hr after injury. Focal injection of TTX at 15 min after SCI also reduces chronic white matter loss and hindlimb functional deficits. We have now tested the hypothesis that the reduction in chronic deficits with TTX treatment is associated with long-term preservation of axons after SCI and compared both acute (24 hr) and chronic (6 weeks) effects of TTX administered at 15 min prior to and 5 min or 4 hr after SCI. Our results indicate a significant reduction of acute white matter pathology in rats treated with TTX at 15 min before and 5 min after injury but no effect when treatment was delayed until 4 hr after contusion. Compared with injury controls, groups treated with TTX at 5 min and 4 hr after injury did not show a significant deficit reduction, nor was there a significant sparing of white matter at 6 weeks compared with injury controls. In contrast, the group treated with TTX at 15 min before SCI demonstrated significantly reduced hindlimb functional deficits beginning at 1 week after injury and throughout the 6 weeks of the study. This was associated with a significantly higher axon density in the ventromedial white matter at 6 weeks. The results demonstrate that blockade of sodium channels preserves axons from loss after SCI and points to the importance of time of administration of such drugs for therapeutic effectiveness.
Collapse
Affiliation(s)
- L J Rosenberg
- Department of Neuroscience, Georgetown University, 3970 Reservoir Road NW, Washington, DC 20007, USA
| | | |
Collapse
|
41
|
Tuna M, Polat S, Erman T, Ildan F, Göçer AI, Tuna N, Tamer L, Kaya M, Cetinalp E. Effect of anti-rat interleukin-6 antibody after spinal cord injury in the rat: inducible nitric oxide synthase expression, sodium- and potassium-activated, magnesium-dependent adenosine-5'-triphosphatase and superoxide dismutase activation, and ultrastructural changes. J Neurosurg 2001; 95:64-73. [PMID: 11453434 DOI: 10.3171/spi.2001.95.1.0064] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT The inflammatory cells that accumulate at the damaged site after spinal cord injury (SCI) may secrete interleukin-6 (IL-6), a mediator known to induce the expression of inducible nitric oxide synthase (iNOS). Any increased production of NO by iNOS activity would aggravate the primary neurological damage in SCI. If this mechanism does occur, the direct or indirect effects of IL-6 antagonists on iNOS activity should modulate this secondary injury. In this study, the authors produced spinal cord damage in rats and applied anti-rat IL-6 antibody to neutralize IL-6 bioactivity and to reduce iNOS. They determined the spinal cord tissue activities of Na+-K+/Mg++ adenosine-5'-triphosphatase (ATPase) and superoxide dismutase, evaluated iNOS immunoreactivity, and examined ultrastructural findings to assess the results of this treatment. METHODS Seventy rats were randomly allocated to four groups. Group I (10 rats) were killed to provide normal spinal cord tissue for testing. In Group II 20 rats underwent six-level laminectomy for the effects of total laminectomy alone to be determined. In Group III 20 rats underwent six-level T2-7 laminectomy and SCI was produced by extradural compression of the exposed cord. The same procedures were performed in the 20 Group IV rats, but these rats also received one (2 microg) intraperitoneal injection of anti-rat IL-6 antibody immediately after the injury and a second dose 24 hours posttrauma. Half of the rats from each of Groups II through IV were killed at 2 hours and the other half at 48 hours posttrauma. The exposed cord segments were immediately removed and processed for analysis. CONCLUSIONS The results showed that neutralizing IL-6 bioactivity with anti-rat IL-6 antibody significantly attenuates iNOS activity and reduces secondary structural changes in damaged rat spinal cord tissue.
Collapse
Affiliation(s)
- M Tuna
- Department of Neurosurgery, Cukurova University, School of Medicine, Adana, Turkey.
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Ray SK, Matzelle DD, Wilford GG, Hogan EL, Banik NL. Cell death in spinal cord injury (SCI) requires de novo protein synthesis. Calpain inhibitor E-64-d provides neuroprotection in SCI lesion and penumbra. Ann N Y Acad Sci 2001; 939:436-49. [PMID: 11462799 DOI: 10.1111/j.1749-6632.2001.tb03655.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Degradation of cytoskeletal proteins by calpain, a Ca(2+)-dependent cysteine protease, may promote neuronal apoptosis in the lesion and surrounding areas following spinal cord injury (SCI). Clinically relevant moderate (40 g-cm force) SCI in rats was induced at T12 by a standardized weight-drop method. Internucleosomal DNA fragmentation or apoptosis in the lesion was inhibited by 24-h treatment of SCI rats with cycloheximide (1 mg/kg), indicating a requirement for de novo protein synthesis in this process. To prove an involvement of calpain activity in mediation of apoptosis in SCI, we treated SCI rats with a cell-permeable calpain inhibitor E-64-d (1 mg/kg). Following 24-h treatment, a 5-cm-long spinal cord section centered at the lesion was collected, and divided equally into five segments (1 cm each) to determine calpain activity, as shown by degradation of the 68-kD neurofilament protein (NFP), and apoptosis as indicated by internucleosomal DNA fragmentation. Neurodegeneration propagated from the site of injury to neighboring rostral and caudal regions. Both calpain activity and apoptosis were readily detectable in the lesion, and moderately so in neighboring areas of untreated SCI rats, whereas these were almost undetectable in E-64-d-treated SCI rats, and absent in sham animals. Results indicate that apoptosis in the SCI lesion and penumbra is prominently associated with calpain activity and is inhibited by the calpain inhibitor E-64-d providing neuroprotective benefit.
Collapse
Affiliation(s)
- S K Ray
- Department of Neurology, Medical University of South Carolina, Charleston, South Carolina 29425, USA
| | | | | | | | | |
Collapse
|
43
|
Bondarenko A, Chesler M. Rapid astrocyte death induced by transient hypoxia, acidosis, and extracellular ion shifts. Glia 2001; 34:134-42. [PMID: 11307162 DOI: 10.1002/glia.1048] [Citation(s) in RCA: 87] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Death of astrocytes requires hours to days in injury models that use hypoxia, acidosis, or calcium paradox protocols. These methods do not incorporate the shifts in extracellular K(+), Na(+), Cl(-), and Ca(2+) that accompany acute brain insults. We studied astrocyte survival after exposure to hypoxic, acidic, ion-shifted Ringer (HAIR), with respective [Ca(2+)], [K(+)], [Na(+)], [Cl(-)], and [HCO(-)(3)] of 0.13, 65, 51, 75, and 13 mM (15% CO(2)/85% N(2), pH 6.6). Intracellular pH (pH(i)) was monitored with the fluorescent dye BCECF. Cell death was indicated by a steep fall in the pH-insensitive, 440-nm-induced fluorescence (F440) and was confirmed by propidium iodide staining. After 15-40-min HAIR exposure, reperfusion with standard Ringer caused death of most cultured (and acutely dissociated) astrocytes within 20 min. Cell death was not prevented if low Ca(2+) was maintained during reperfusion. Survival fell with increased HAIR duration, elevated temperature, or absence of external glucose. Comparable durations of hypoxia, acidosis, or ion shifts alone did not lead to acute cell death, while modest loss was noted when acidosis was paired with either hypoxia or ion shifts. Severe cell loss required the triad of hypoxia, acidosis, and ion shifts. Intracellular pH was significantly higher in HAIR media, compared with solutions of low pH alone or with low pH plus hypoxia. These results indicate that astrocytes can be killed rapidly by changes in the extracellular microenvironment that occur in settings of traumatic and ischemic brain injury.
Collapse
Affiliation(s)
- A Bondarenko
- Department of Neurosurgery, New York University School of Medicine, New York, New York 10016, USA
| | | |
Collapse
|
44
|
Rosenberg LJ, Emery DG, Lucas JH. Effects of sodium and chloride on neuronal survival after neurite transection. J Neuropathol Exp Neurol 2001; 60:33-48. [PMID: 11202174 DOI: 10.1093/jnen/60.1.33] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
An in vitro investigation was undertaken to study the roles of Na+ and Cl- in mammalian spinal cord (SC) neuron deterioration and death after injury involving physical disruption of the plasma membrane. Individual SC neurons in monolayer cultures were subjected to UV laser microbeam transection of a primary dendrite. Neurons lesioned in modified ionic environments (MIEs) where 50%-75% of the NaCl was replaced with sucrose had higher survival (65%-75%) than neurons lesioned in medium with normal (125 mM) NaCl (28%; p < 0.001). Subsequent experiments found a comparable increase in lesioned neuron survival in MIEs in which only Na+ was replaced with specific ionic substitutes; however, replacement of Cl- was not protective. Electron microscope examinations of neurons fixed <16 min after lesioning showed a dramatic decrease in vesiculation of the smooth endoplasmic reticulum and Golgi apparatus in the low NaCl or low Na+ MIEs. It is hypothesized that Na+ entry after membrane disruption may stimulate elevation of [Ca+2]i leading to ultrastructural disruption and death of injured neurons. The results of these studies suggest that a low NaCl MIE may be useful as an irrigant to limit damage spread and cell death within CNS tissues during surgery or after trauma.
Collapse
Affiliation(s)
- L J Rosenberg
- Department of Neuroscience, Georgetown University, Washington, DC, USA
| | | | | |
Collapse
|
45
|
Shi R, Asano T, Vining NC, Blight AR. Control of membrane sealing in injured mammalian spinal cord axons. J Neurophysiol 2000; 84:1763-9. [PMID: 11024068 DOI: 10.1152/jn.2000.84.4.1763] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The process of sealing of damaged axons was examined in isolated strips of white matter from guinea pig spinal cord by recording the "compound membrane potential," using a sucrose-gap technique, and by examining uptake of horseradish peroxidase (HRP). Following axonal transection, exponential recovery of membrane potential occurred with a time constant of 20 +/- 5 min, at 37 degrees C, and extracellular calcium activity ([Ca(2+)](o)) of 2 mM. Most axons excluded HRP by 30 min following transection. The rate of sealing was reduced by lowering calcium and was effectively blocked at [Ca(2+)](o) </= 0.5 mM, under which condition most axons continued to take up HRP for more than 1 h. Sealing at higher [Ca(2+)](o) was blocked by calpain inhibitors (calpeptin and calpain inhibitor-1) indicating a requirement for type II (mM) calpain in the sealing process. Following compression injury, the amplitude of the maximal compound action potential conducted through the injury site was reduced. The extent of amplitude reduction was increased when the tract was superfused with calcium-free Krebs' solution (Ca(2+) replaced by Mg(2+)). These results suggest that the fall in [Ca(2+)](o) seen following injury in vivo is sufficient to prevent membrane sealing and may paradoxically contribute to axonal dieback, retrograde cell death, and "secondary" axonal disruption.
Collapse
Affiliation(s)
- R Shi
- Division of Neurosurgery, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA.
| | | | | | | |
Collapse
|
46
|
Shields DC, Schaecher KE, Hogan EL, Banik NL. Calpain activity and expression increased in activated glial and inflammatory cells in penumbra of spinal cord injury lesion. J Neurosci Res 2000; 61:146-50. [PMID: 10878587 DOI: 10.1002/1097-4547(20000715)61:2<146::aid-jnr5>3.0.co;2-c] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Following traumatic injury of the spinal cord, cells adjacent to the lesion are subject to ischemic cell death as a result of vascular disruption and secondary inflammatory responses. Proteases such as calcium-activated neutral proteinase (calpain) have been implicated in axon and myelin destruction following injury since they degrade structural proteins in the axon-myelin unit. To examine the role of calpain in cell death following spinal cord injury (SCI), calpain activity and translational expression were evaluated using Western blotting techniques. Calpain activity (as measured by specific substrate degradation) was significantly increased in and around the lesion site as early as 4 hr following injury with continued elevation at 48 hr compared to sham controls. Likewise, calpain expression was significantly increased in both the lesion site and penumbra at 4 and 48 hr after injury. Using double immunofluorescent labeling for calpain and cell-specific markers, this increase in calpain expression was found to be due in part to activated glial/inflammatory cells such as astrocytes, microglia, and infiltrating macrophages in these areas. Thus, since calpain degrades many myelin and axonal structural proteins, the increased activity and expression of this enzyme may be responsible for destruction of myelinated axons adjacent to the lesion site following SCI.
Collapse
Affiliation(s)
- D C Shields
- Department of Neurology, Medical University of South Carolina, 171 Ashley Avenue, Charleston, SC 29425, USA
| | | | | | | |
Collapse
|
47
|
Ray SK, Matzelle DC, Wilford GG, Hogan EL, Banik NL. E-64-d prevents both calpain upregulation and apoptosis in the lesion and penumbra following spinal cord injury in rats. Brain Res 2000; 867:80-9. [PMID: 10837800 DOI: 10.1016/s0006-8993(00)02260-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Calpain, a Ca(2+)-dependent cysteine protease, has been implicated in cytoskeletal protein degradation and neurodegeneration in the lesion and adjacent areas following spinal cord injury (SCI). To attenuate apoptosis or programmed cell death (PCD) in SCI, we treated injured rats with E-64-d, a cell permeable and selective inhibitor of calpain. SCI was induced on T12 by the weight-drop (40 g-cm force) method. Within 15 min, E-64-d (1 mg/kg) in 1.5% DMSO was administered i.v. to the SCI rats. Following 24 h treatment, a 5-cm long spinal cord section with the lesion in the center was collected. The spinal cord section was divided equally into five 1-cm segments (S1: distant rostral, S2: near rostral, S3: lesion or injury, S4: near caudal and S5: distant caudal) for analysis. Determination of mRNA levels by reverse transcriptase-polymerase chain reaction (RT-PCR) indicated that ratios of bax/bcl-2 and calpain/calpastatin were increased in spinal cord segments from injured rats compared to controls. Degradation of the 68-kD neurofilament protein and internucleosomal DNA fragmentation were also increased. All of these changes were maximally increased in the lesion and gradually decreased in the adjacent areas of SCI rats, while largely undetectable in E-64-d treated rats and absent in sham controls. The results indicate that apoptosis in rat SCI appears to be associated with calpain activity which can be attenuated by the calpain inhibitor E-64-d.
Collapse
Affiliation(s)
- S K Ray
- Department of Neurology, Medical University of South Carolina, 96 Jonathan Lucas Street, Suite 309, Charleston, SC 29425, USA
| | | | | | | | | |
Collapse
|
48
|
Paemeleire K, Leybaert L. Ionic changes accompanying astrocytic intercellular calcium waves triggered by mechanical cell damaging stimulation. Brain Res 2000; 857:235-45. [PMID: 10700572 DOI: 10.1016/s0006-8993(99)02436-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Mechanically poking or damaging a single cell within a confluent astrocyte culture produces the so-called intercellular calcium (Ca(2+)) waves, that is, cell-to-cell propagating changes of intracellular free Ca(2+). We were interested whether intercellular Ca(2+) waves are also associated with changes in other intra- or extracellular ions. To that purpose, we investigated spatiotemporal changes of intracellular Ca(2+) (Ca(i)2+), sodium (Na(i)+) and protons (H(i)+) in primary cultures of rat cortical astrocytes using microfluorescence imaging with fura-2, SBFI and BCECF, respectively; changes of extracellular potassium (K(e)+) were monitored with K(+)-sensitive microelectrodes. Mechanical damage to a single cell by stimulation with a piezo-electrically driven micropipette initiated intercellular Ca(2+) waves that propagated to about 160 microm away from the stimulation point. Na(i)(+) increases could be detected in cells located 2-3 cell diameters from the stimulated cell, acidification was observed 1-2 cell diameters away and Ke(+) increases were measured up to 75 microm away. Kinetic analysis suggests that the Na(i)(+) and H(i)(+) changes occur after, and thus secondary to the Ca(i)(2+) changes. In contrast, K(e)(+) changes occurred very fast, even before the Ca(i)(2+) changes, but their propagation speed was too fast to implicate them as a trigger of Ca(i)(2+) changes. As Na(i)(+) is an important regulator of glycolysis in astrocytes, we hypothesize that astrocytic Na(i)(+) changes in cells located remotely from a damaged cell might be a signal that activates glycolysis thereby producing more lactate that is transferred to the neurons and increases their energy potential to survive the inflicted damage.
Collapse
Affiliation(s)
- K Paemeleire
- Department of Physiology and Pathophysiology, University of Ghent, De Pintelaan 185 (Blok B), B-9000, Ghent, Belgium
| | | |
Collapse
|
49
|
LoPachin RM, Gaughan CL, Lehning EJ, Kaneko Y, Kelly TM, Blight A. Experimental spinal cord injury: spatiotemporal characterization of elemental concentrations and water contents in axons and neuroglia. J Neurophysiol 1999; 82:2143-53. [PMID: 10561394 DOI: 10.1152/jn.1999.82.5.2143] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
To examine the role of axonal ion deregulation in acute spinal cord injury (SCI), white matter strips from guinea pig spinal cord were incubated in vitro and were subjected to graded focal compression injury. At several postinjury times, spinal segments were removed from incubation and rapidly frozen. X-ray microanalysis was used to measure percent water and dry weight elemental concentrations (mmol/kg) of Na, P, Cl, K, Ca, and Mg in selected morphological compartments of myelinated axons and neuroglia from spinal cord cryosections. As an index of axon function, compound action potentials (CAP) were measured before compression and at several times thereafter. Axons and mitochondria in epicenter of severely compressed spinal segments exhibited early (5 min) increases in mean Na and decreases in K and Mg concentrations. These elemental changes were correlated to a significant reduction in CAP amplitude. At later postcompression times (15 and 60 min), elemental changes progressed and were accompanied by alterations in compartmental water content and increases in mean Ca. Swollen axons were evident at all postinjury times and were characterized by marked element and water deregulation. Neuroglia and myelin in severely injured epicenter also exhibited significant disruptions. In shoulder areas (adjacent to epicenter) of severely injured spinal strips, axons and mitochondria exhibited modest increases in mean Na in conjunction with decreases in K, Mg, and water content. Following moderate compression injury to spinal strips, epicenter axons exhibited early (10 min postinjury) element and water deregulation that eventually recovered to near control values (60 min postinjury). Na(+) channel blockade by tetrodotoxin (TTX, 1 microM) perfusion initiated 5 min after severe crush diminished both K loss and the accumulation of Na, Cl, and Ca in epicenter axons and neuroglia, whereas in shoulder regions TTX perfusion completely prevented subcellular elemental deregulation. TTX perfusion also reduced Na entry in swollen axons but did not affect K loss or Ca gain. Thus graded compression injury of spinal cord produced subcellular elemental deregulation in axons and neuroglia that correlated with the onset of impaired electrophysiological function and neuropathological alterations. This suggests that the mechanism of acute SCI-induced structural and functional deficits are mediated by disruption of subcellular ion distribution. The ability of TTX to reduce elemental deregulation in compression-injured axons and neuroglia implicates a significant pathophysiological role for Na(+) influx in SCI and suggests Na(+) channel blockade as a pharmacotherapeutic strategy.
Collapse
Affiliation(s)
- R M LoPachin
- Department of Anesthesiology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York 10467, USA
| | | | | | | | | | | |
Collapse
|
50
|
Samii A, Badie H, Fu K, Luther RR, Hovda DA. Effects of an N-type calcium channel antagonist (SNX 111; Ziconotide) on calcium-45 accumulation following fluid-percussion injury. J Neurotrauma 1999; 16:879-92. [PMID: 10547097 DOI: 10.1089/neu.1999.16.879] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Accumulation of calcium following experimental traumatic brain injury (TBI) has been demonstrated to be a prominent pathophysiological component that can compromise mitochondrial functioning and threaten cell survival. The omega-conopeptide SNX-111, also known as Ziconotide, is a potent antagonist of the voltage-gated N-type calcium channel and has demonstrated significant neuroprotective effects against ischemia-induced neuronal injury. To determine whether this compound would be effective in reducing calcium accumulation associated with TBI, SNX-111 was administered intravenously to rats 1 hour following a moderate (2.2 to 2.75 atm) lateral fluid-percussion injury (or sham) at doses of 1 (n = 30), 3 (n = 31), or 5 (n = 30) mg/kg; another group received 0.9% saline solution (n = 35). Brains were processed for calcium 45 (45Ca) autoradiography at 6, 12, 24, 48, and 96 hours following insult. Optical density measurements of 20 cortical and subcortical regions were analyzed. Injured animals administered saline solution exhibited a significant increase in 45Ca uptake within 12 regions ipsilateral to the site of injury. The most prominent increases were evident throughout the ipsilateral cerebral cortex. SNX-111 reduced the injury-induced calcium accumulation within the ipsilateral cortex in a dose-response fashion when measured at 6, 12, and 48 hours after insult. These drug-induced reductions in calcium accumulation were as high as 75% in the ipsilateral cerebral cortex, and up to 50% in other ipsilateral regions (including thalamus and hippocampus). Consequently, the results suggest that posttraumatic blocking of the voltage-gated N-type calcium channel after injury reduces prolonged, trauma-induced calcium accumulation.
Collapse
Affiliation(s)
- A Samii
- Brain Research Institute, UCLA School of Medicine, Los Angeles, California 90095-7039, USA
| | | | | | | | | |
Collapse
|