151
|
Park HJ, Kim JY. Incidence of Neutropenia With Valproate and Quetiapine Combination Treatment in Subjects With Acquired Brain Injuries. Arch Phys Med Rehabil 2015; 97:183-8. [PMID: 26427579 DOI: 10.1016/j.apmr.2015.09.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 09/01/2015] [Accepted: 09/10/2015] [Indexed: 01/18/2023]
Abstract
OBJECTIVE To investigate whether the incidence of neutropenia was higher in subjects who received a combination treatment with valproate and quetiapine than in those who were administered monotherapy. DESIGN Retrospective cohort study. SETTING Rehabilitation department of a university hospital. PARTICIPANTS Patients with acquired brain injuries who had taken valproate for seizures or quetiapine for delirium for >7 days (N=101). Data were extracted from electronic medical records of the hospital. INTERVENTIONS Not applicable. MAIN OUTCOME MEASURES Incidence of neutropenia (absolute neutrophil count<2000 cells/μL) was elicited from the weekly complete blood cell records for 71.07±43.71 days of observation. The odds ratio for neutropenia development was calculated and adjusted for variables that showed significant differences between patients with or without neutropenia. RESULTS The incidence of neutropenia was significantly higher in the group receiving the combination treatment than in those receiving the monotherapy (32.26% vs 12.90%, adjusted P=.036), despite a lack of any differences in the daily doses of the medications. Coadministration of quetiapine and valproate was the predictor of neutropenia development when age, body weight, and underlying diseases were adjusted in the logistic regression model (odds ratio=3.749; 95% confidence interval, 1.161-12.099; P=.027). CONCLUSIONS Administration of quetiapine together with valproate in patients with acquired brain injury could increase the incidence of medication-induced neutropenia.
Collapse
Affiliation(s)
- Hee Jung Park
- Department of Rehabilitation Medicine, Dankook University College of Medicine, Cheonan, South Korea
| | - Jung Yoon Kim
- Ewha Brain Institute, Ewha Womans University, Seoul, South Korea.
| |
Collapse
|
152
|
Abstract
Priming is a type of implicit learning wherein a stimulus prompts a change in behavior. Priming has been long studied in the field of psychology. More recently, rehabilitation researchers have studied motor priming as a possible way to facilitate motor learning. For example, priming of the motor cortex is associated with changes in neuroplasticity that are associated with improvements in motor performance. Of the numerous motor priming paradigms under investigation, only a few are practical for the current clinical environment, and the optimal priming modalities for specific clinical presentations are not known. Accordingly, developing an understanding of the various types of motor priming paradigms and their underlying neural mechanisms is an important step for therapists in neurorehabilitation. Most importantly, an understanding of the methods and their underlying mechanisms is essential for optimizing rehabilitation outcomes. The future of neurorehabilitation is likely to include these priming methods, which are delivered prior to or in conjunction with primary neurorehabilitation therapies. In this Special Interest article, we discuss those priming paradigms that are supported by the greatest amount of evidence, including (i) stimulation-based priming, (ii) motor imagery and action observation, (iii) sensory priming, (iv) movement-based priming, and (v) pharmacological priming.Video Abstract available. (see Supplemental Digital Content 1, http://links.lww.com/JNPT/A86) for more insights from the authors.
Collapse
|
153
|
Xu Y, Zhang G, Kang Z, Xu Y, Jiang W, Zhang S. Cornin increases angiogenesis and improves functional recovery after stroke via the Ang1/Tie2 axis and the Wnt/β-catenin pathway. Arch Pharm Res 2015; 39:133-42. [DOI: 10.1007/s12272-015-0652-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 08/11/2015] [Indexed: 11/28/2022]
|
154
|
Cramer SC. Drugs to Enhance Motor Recovery After Stroke. Stroke 2015; 46:2998-3005. [PMID: 26265126 DOI: 10.1161/strokeaha.115.007433] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 07/15/2015] [Indexed: 12/13/2022]
Affiliation(s)
- Steven C Cramer
- From the Deparments of Neurology, Anatomy & Neurobiology, and Physical Medicine & Rehabilitation, University of California, Irvine, CA.
| |
Collapse
|
155
|
Horn SD, Corrigan JD, Beaulieu CL, Bogner J, Barrett RS, Giuffrida CG, Ryser DK, Cooper K, Carroll DM, Deutscher D. Traumatic Brain Injury Patient, Injury, Therapy, and Ancillary Treatments Associated With Outcomes at Discharge and 9 Months Postdischarge. Arch Phys Med Rehabil 2015; 96:S304-29. [PMID: 26212406 PMCID: PMC4517296 DOI: 10.1016/j.apmr.2014.11.030] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2014] [Revised: 10/28/2014] [Accepted: 11/20/2014] [Indexed: 11/19/2022]
Abstract
OBJECTIVE To examine associations of patient and injury characteristics, inpatient rehabilitation therapy activities, and neurotropic medications with outcomes at discharge and 9 months postdischarge for patients with traumatic brain injury (TBI). DESIGN Prospective, longitudinal observational study. SETTING Inpatient rehabilitation centers. PARTICIPANTS Consecutive patients (N=2130) enrolled between 2008 and 2011, admitted for inpatient rehabilitation after an index TBI injury. INTERVENTIONS Not applicable. MAIN OUTCOME MEASURES Rehabilitation length of stay, discharge to home, and FIM at discharge and 9 months postdischarge. RESULTS The admission FIM cognitive score was used to create 5 relatively homogeneous subgroups for subsequent analysis of treatment outcomes. Within each subgroup, significant associations were found between outcomes and patient and injury characteristics, time spent in therapy activities, and medications used. Patient and injury characteristics explained on average 35.7% of the variation in discharge outcomes and 22.3% in 9-month outcomes. Adding time spent and level of effort in therapy activities and percentage of stay using specific medications explained approximately 20% more variation for discharge outcomes and 12.9% for 9-month outcomes. After patient, injury, and treatment characteristics were used to predict outcomes, center differences added only approximately 1.9% additional variance explained. CONCLUSIONS At discharge, greater effort during therapy sessions, time spent in more complex therapy activities, and use of specific medications were associated with better outcomes for patients in all admission FIM cognitive subgroups. At 9 months postdischarge, similar but less pervasive associations were observed for therapy activities, but not classes of medications. Further research is warranted to examine more specific combinations of therapy activities and medications that are associated with better outcomes.
Collapse
Affiliation(s)
- Susan D Horn
- Institute for Clinical Outcomes Research, International Severity Information Systems, Salt Lake City, UT.
| | - John D Corrigan
- Department of Physical Medicine and Rehabilitation, Ohio State University, Columbus, OH
| | | | - Jennifer Bogner
- Department of Physical Medicine and Rehabilitation, Ohio State University, Columbus, OH
| | - Ryan S Barrett
- Institute for Clinical Outcomes Research, International Severity Information Systems, Salt Lake City, UT
| | | | - David K Ryser
- Neuro Specialty Rehabilitation Unit, Intermountain Medical Center, Salt Lake City, UT
| | - Kelli Cooper
- Neuro Specialty Rehabilitation Unit, Intermountain Medical Center, Salt Lake City, UT
| | - Deborah M Carroll
- Neuro Specialty Rehabilitation Unit, Intermountain Medical Center, Salt Lake City, UT
| | | |
Collapse
|
156
|
Behrouz R, Godoy DA, Azarpazhooh MR, Di Napoli M. Altered mental status in the neurocritical care unit. J Crit Care 2015; 30:1272-7. [PMID: 26315655 DOI: 10.1016/j.jcrc.2015.07.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Revised: 07/21/2015] [Accepted: 07/22/2015] [Indexed: 11/24/2022]
Abstract
Altered mental status is a common pathological entity in critically ill patients and particularly in those with preexisting cerebral injury. In the neurological critical care unit, the prevalence of altered mental status is especially high because of the inherent nervous system disease of these patients. Altered mental status can be crudely divided into encephalopathy and delirium. Although often used interchangeably, the 2 pathological entities have subtle differences in etiology and presentation. This is a review of delirium and encephalopathy in the neurological critical care unit.
Collapse
Affiliation(s)
- Réza Behrouz
- Department of Neurology, School of Medicine, University of Texas Health Science Center, San Antonio, TX, USA.
| | - Daniel A Godoy
- The Neurointensive Care Unit, Sanatorio Pasteur; and the Intensive Care Unit, Hospital Interzonal de Agudos ''San Juan Bautista," Catamarca, Argentina
| | - Mahmoud R Azarpazhooh
- Department of Clinical Neurological Sciences, Schulich School of Medicine, Western University, London, ON, Canada
| | - Mario Di Napoli
- Neurological Service, San Camillo de' Lellis General Hospital, Rieti, Italy; Neurological Section, SMDN Centre for Cardiovascular Medicine and Cerebrovascular Disease Prevention, Sulmona, L'Aquila, Italy
| |
Collapse
|
157
|
Haulcomb MM, Mesnard-Hoaglin NA, Batka RJ, Meadows RM, Miller WM, Mcmillan KP, Brown TJ, Sanders VM, Jones KJ. Identification of B6SJL mSOD1(G93A) mouse subgroups with different disease progression rates. J Comp Neurol 2015; 523:2752-68. [PMID: 26010802 DOI: 10.1002/cne.23814] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 05/14/2015] [Accepted: 05/15/2015] [Indexed: 12/16/2022]
Abstract
Disease progression rates among patients with amyotrophic lateral sclerosis (ALS) vary greatly. Although the majority of affected individuals survive 3-5 years following diagnosis, some subgroups experience a more rapidly progressing form, surviving less than 1 year, and other subgroups experience slowly progressing forms, surviving nearly 50 years. Genetic heterogeneity and environmental factors pose significant barriers in investigating patient progression rates. Similar to the case for humans, variation in survival within the mSOD1 mouse has been well documented, but different progression rates have not been investigated. The present study identifies two subgroups of B6SJL mSOD1(G93A) mice with different disease progression rates, a fast progression group (FPG) and slow progression group, as evidenced by differences in the rate of motor function decline. In addition, increased disease-associated gene expression within the FPG facial motor nucleus confirmed the presence of a more severe phenotype. We hypothesize that a more severe disease phenotype could be the result of 1) an earlier onset of axonal disconnection with a consistent degeneration rate or 2) a more severe or accelerated degenerative process. We performed a facial nerve transection axotomy in both mSOD1 subgroups prior to disease onset as a method to standardize the axonal disconnection. Instead of leading to comparable gene expression in both subgroups, this standardization did not eliminate the severe phenotype in the FPG facial nucleus, suggesting that the FPG phenotype is the result of a more severe or accelerated degenerative process. We theorize that these mSOD1 subgroups are representative of the rapid and slow disease phenotypes often experienced in ALS.
Collapse
Affiliation(s)
- Melissa M Haulcomb
- Neuroscience Program, Loyola University Medical Center, Maywood, Illinois, 60153.,Research and Development Service, Hines Veterans Administration Hospital, Hines, Illinois, 60141.,Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana, 46202.,Research and Development Service, Roudebush Veterans Administration Medical Center, Indianapolis, Indiana, 46202
| | - Nichole A Mesnard-Hoaglin
- Neuroscience Program, Loyola University Medical Center, Maywood, Illinois, 60153.,Research and Development Service, Hines Veterans Administration Hospital, Hines, Illinois, 60141
| | - Richard J Batka
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana, 46202.,Research and Development Service, Roudebush Veterans Administration Medical Center, Indianapolis, Indiana, 46202
| | - Rena M Meadows
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana, 46202.,Research and Development Service, Roudebush Veterans Administration Medical Center, Indianapolis, Indiana, 46202.,Program in Medical Neurosciences, Indiana University School of Medicine, Indianapolis, Indiana, 46202
| | - Whitney M Miller
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana, 46202.,Research and Development Service, Roudebush Veterans Administration Medical Center, Indianapolis, Indiana, 46202
| | - Kathryn P Mcmillan
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana, 46202.,Research and Development Service, Roudebush Veterans Administration Medical Center, Indianapolis, Indiana, 46202
| | - Todd J Brown
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana, 46202.,Research and Development Service, Roudebush Veterans Administration Medical Center, Indianapolis, Indiana, 46202
| | - Virginia M Sanders
- Department of Molecular Virology, Immunology, and Medical Genetics, The Ohio State University, Columbus, Ohio, 43210
| | - Kathryn J Jones
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana, 46202.,Research and Development Service, Roudebush Veterans Administration Medical Center, Indianapolis, Indiana, 46202
| |
Collapse
|
158
|
Wei W, Wang H, Wu Y, Ding K, Li T, Cong Z, Xu J, Zhou M, Huang L, Ding H, Wu H. Alpha lipoic acid inhibits neural apoptosis via a mitochondrial pathway in rats following traumatic brain injury. Neurochem Int 2015; 87:85-91. [PMID: 26055972 DOI: 10.1016/j.neuint.2015.06.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 05/28/2015] [Accepted: 06/03/2015] [Indexed: 11/18/2022]
Abstract
Alpha lipoic acid (ALA) is a powerful antioxidant that has proven protective effects against brain damage following a traumatic brain injury (TBI) in rats. However, the molecular mechanisms underlying these effects are not well understood. This study investigated the effect of ALA on neural apoptosis and the potential mechanism of these effects in the weight-drop model of TBI in male Sprague-Dawley rats that were treated with ALA (20 or 100 mg/kg) or vehicle via intragastric administration 30 min after TBI. Brain samples were collected 48 h later for analysis. ALA treatment resulted in a downregulation of caspase-3 expression, reduced the number of positive cells in the terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling assay and improved neuronal survival. Furthermore, the level of malondialdehyde and glutathione peroxidase activity were restored, while Bcl-2-associated X protein translocation to mitochondria and cytochrome c release into the cytosol were reduced by ALA treatment. These results demonstrate that ALA improves neurological outcome in rats by protecting neural cell against apoptosis via a mechanism that involves the mitochondria following TBI.
Collapse
Affiliation(s)
- Wuting Wei
- Department of Neurosurgery, School of Medicine, Southern Medical University (Guangzhou), Jinling Hospital, 305 East Zhongshan Road, Nanjing, Jiangsu Province 210002, China
| | - Handong Wang
- Department of Neurosurgery, School of Medicine, Southern Medical University (Guangzhou), Jinling Hospital, 305 East Zhongshan Road, Nanjing, Jiangsu Province 210002, China.
| | - Yong Wu
- Department of Neurosurgery, School of Medicine, Nanjing University, Jinling Hospital, 305 East Zhongshan Road, Nanjing, Jiangsu Province 210002, China
| | - Ke Ding
- Department of Neurosurgery, School of Medicine, Nanjing University, Jinling Hospital, 305 East Zhongshan Road, Nanjing, Jiangsu Province 210002, China
| | - Tao Li
- Department of Neurosurgery, School of Medicine, Nanjing University, Jinling Hospital, 305 East Zhongshan Road, Nanjing, Jiangsu Province 210002, China
| | - Zixiang Cong
- Department of Neurosurgery, School of Medicine, Nanjing University, Jinling Hospital, 305 East Zhongshan Road, Nanjing, Jiangsu Province 210002, China
| | - Jianguo Xu
- Department of Neurosurgery, School of Medicine, Nanjing University, Jinling Hospital, 305 East Zhongshan Road, Nanjing, Jiangsu Province 210002, China
| | - Mengliang Zhou
- Department of Neurosurgery, School of Medicine, Nanjing University, Jinling Hospital, 305 East Zhongshan Road, Nanjing, Jiangsu Province 210002, China
| | - Litian Huang
- Department of Neurosurgery, School of Medicine, Southern Medical University (Guangzhou), Jinling Hospital, 305 East Zhongshan Road, Nanjing, Jiangsu Province 210002, China
| | - Hui Ding
- Department of Neurosurgery, School of Medicine, Southern Medical University (Guangzhou), Jinling Hospital, 305 East Zhongshan Road, Nanjing, Jiangsu Province 210002, China
| | - Heming Wu
- Department of Neurosurgery, School of Medicine, Nanjing University, Jinling Hospital, 305 East Zhongshan Road, Nanjing, Jiangsu Province 210002, China
| |
Collapse
|
159
|
Sakic B, Cooper MPA, Taylor SE, Stojanovic M, Zagorac B, Kapadia M. Behavioral Phenotyping of Murine Disease Models with the Integrated Behavioral Station (INBEST). J Vis Exp 2015. [PMID: 25938737 PMCID: PMC4650674 DOI: 10.3791/51524] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Due to rapid advances in genetic engineering, small rodents have become the preferred subjects in many disciplines of biomedical research. In studies of chronic CNS disorders, there is an increasing demand for murine models with high validity at the behavioral level. However, multiple pathogenic mechanisms and complex functional deficits often impose challenges to reliably measure and interpret behavior of chronically sick mice. Therefore, the assessment of peripheral pathology and a behavioral profile at several time points using a battery of tests are required. Video-tracking, behavioral spectroscopy, and remote acquisition of physiological measures are emerging technologies that allow for comprehensive, accurate, and unbiased behavioral analysis in a home-base-like setting. This report describes a refined phenotyping protocol, which includes a custom-made monitoring apparatus (Integrated Behavioral Station, INBEST) that focuses on prolonged measurements of basic functional outputs, such as spontaneous activity, food/water intake and motivated behavior in a relatively stress-free environment. Technical and conceptual improvements in INBEST design may further promote reproducibility and standardization of behavioral studies.
Collapse
Affiliation(s)
- Boris Sakic
- Department of Psychiatry and Behavioral Neurosciences, McMaster University;
| | | | - Sarah E Taylor
- Department of Psychology, Neuroscience & Behaviour, McMaster University
| | - Milica Stojanovic
- Department of Psychology, Neuroscience & Behaviour, McMaster University
| | - Bosa Zagorac
- Department of Psychology, Neuroscience & Behaviour, McMaster University
| | | |
Collapse
|
160
|
Glucose administration after traumatic brain injury exerts some benefits and no adverse effects on behavioral and histological outcomes. Brain Res 2015; 1614:94-104. [PMID: 25911580 DOI: 10.1016/j.brainres.2015.04.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 04/10/2015] [Accepted: 04/11/2015] [Indexed: 11/22/2022]
Abstract
The impact of hyperglycemia after traumatic brain injury (TBI), and even the administration of glucose-containing solutions to head injured patients, remains controversial. In the current study adult male Sprague-Dawley rats were tested on behavioral tasks and then underwent surgery to induce sham injury or unilateral controlled cortical impact (CCI) injury followed by injections (i.p.) with either a 50% glucose solution (Glc; 2g/kg) or an equivalent volume of either 0.9% or 8% saline (Sal) at 0, 1, 3 and 6h post-injury. The type of saline treatment did not significantly affect any outcome measures, so these data were combined. Rats with CCI had significant deficits in beam-walking traversal time and rating scores (p's < 0.001 versus sham) that recovered over test sessions from 1 to 13 days post-injury (p's < 0.001), but these beam-walking deficits were not affected by Glc versus Sal treatments. Persistent post-CCI deficits in forelimb contraflexion scores and forelimb tactile placing ability were also not differentially affected by Glc or Sal treatments. However, deficits in latency to retract the right hind limb after limb extension were significantly attenuated in the CCI-Glc group (p < 0.05 versus CCI-Sal). Both CCI groups were significantly impaired in a plus maze test of spatial working memory on days 4, 9 and 14 post-surgery (p < 0.001 versus sham), and there was no effect of Glc versus Sal on this cognitive outcome measure. At 15 days post-surgery the loss of cortical tissue volume (p < 0.001 versus sham) was significantly less in the CCI-Glc group (30.0%; p < 0.05) compared to the CCI-Sal group (35.7%). Counts of surviving hippocampal hilar neurons revealed a significant (~40%) loss ipsilateral to CCI (p < 0.001 versus sham), but neuronal loss in the hippocampus was not different in the CCI-Sal and CCI-Glc groups. Taken together, these results indicate that an early elevation of blood glucose may improve some neurological outcomes and, importantly, the induction of hyperglycemia after isolated TBI did not adversely affect any sensorimotor, cognitive or histological outcomes.
Collapse
|
161
|
Phelps TI, Bondi CO, Ahmed RH, Olugbade YT, Kline AE. Divergent long-term consequences of chronic treatment with haloperidol, risperidone, and bromocriptine on traumatic brain injury-induced cognitive deficits. J Neurotrauma 2015; 32:590-7. [PMID: 25275833 PMCID: PMC4394178 DOI: 10.1089/neu.2014.3711] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
Antipsychotic drugs (APDs) are provided in the clinic to manage traumatic brain injury (TBI)-induced agitation and aggression. Experimental TBI studies consistently show that daily administration of the APDs, haloperidol (HAL) and risperidone (RISP), hinder recovery. However, it is unknown how long the adverse effects remain after cessation of treatment. To elucidate this clinically relevant issue, anesthetized male rats were randomly assigned to four TBI (controlled cortical impact) and four sham groups administered HAL (0.5 mg/kg), RISP (0.45 mg/kg), bromocriptine (BRO; 5.0 mg/kg, included as a control for D2 receptor action), or vehicle (VEH; 1 mL/kg) 24 h after surgery and once-daily for 19 days. Motor and cognitive recovery was assessed on days 1-5 and 14-19, respectively, and again at 1 and 3 months after drug withdrawal. No overall group differences were observed for motor function among the TBI groups, although the HAL group showed a greater beam-walk deficit on day 5 versus the VEH and BRO groups. Cognitive recovery was significantly impaired in the HAL and RISP groups during the treatment phase versus VEH and BRO. Further, BRO was superior to VEH (p=0.0042). At 1 month, both groups that received APDs continued to exhibit significant cognitive impairment versus VEH and BRO; at 3 months, only the HAL group was impaired. Moreover, the HAL, RISP, and VEH groups continued to be cognitively deficient versus BRO, which also reduced cortical damage. These data replicate previous reports that HAL and RISP impede cognitive recovery after TBI and expand the literature by revealing that the deleterious effects persist for 3 months after drug discontinuation. BRO conferred cognitive benefits when administered concomitantly with behavioral testing, thus replicating previous findings, and also after cessation demonstrating enduring efficacy.
Collapse
Affiliation(s)
- Thomas I. Phelps
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Corina O. Bondi
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
- Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Rashid H. Ahmed
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Yewande T. Olugbade
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Anthony E. Kline
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
- Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Psychology, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
162
|
Mering S, Jolkkonen J. Proper housing conditions in experimental stroke studies-special emphasis on environmental enrichment. Front Neurosci 2015; 9:106. [PMID: 25870536 PMCID: PMC4378295 DOI: 10.3389/fnins.2015.00106] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 03/12/2015] [Indexed: 12/20/2022] Open
Abstract
Environmental enrichment provides laboratory animals with novelty and extra space, allowing different forms of multisensory stimulation ranging from social grouping to enhanced motor activity. At the extreme end of the spectrum, one can have a super-enriched environment. Environmental enrichment is believed to result in improved cognitive and sensorimotor functions both in naïve rodents and in animals with brain lesions such as those occurring after a stroke. Robust behavioral effects in animals which have suffered a stroke are probably related not only to neuronal plasticity in the perilesional cortex but also in remote brain areas. There is emerging evidence to suggest that testing restorative therapies in an enriched environment can maximize treatment effects, e.g., the perilesional milieu seems to be more receptive to concomitant pharmacotherapy and/or cell therapy. This review provides an updated overview on the effect of an enriched environment in stroke animals from the practical points to be considered when planning experiments to the mechanisms explaining why combined therapies can contribute to behavioral improvement in a synergistic manner.
Collapse
Affiliation(s)
- Satu Mering
- Lab Animal Centre, University of Eastern Finland Kuopio, Finland
| | - Jukka Jolkkonen
- Institute of Clinical Medicine - Neurology, University of Eastern Finland Kuopio, Finland
| |
Collapse
|
163
|
Yang LY, Chu YH, Tweedie D, Yu QS, Pick CG, Hoffer BJ, Greig NH, Wang JY. Post-trauma administration of the pifithrin-α oxygen analog improves histological and functional outcomes after experimental traumatic brain injury. Exp Neurol 2015; 269:56-66. [PMID: 25819102 DOI: 10.1016/j.expneurol.2015.03.015] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 03/13/2015] [Accepted: 03/17/2015] [Indexed: 01/10/2023]
Abstract
Traumatic brain injury (TBI) is a major cause of death and disability worldwide. Programmed death of neuronal cells plays a crucial role in acute and chronic neurodegeneration following TBI. The tumor suppressor protein p53, a transcription factor, has been recognized as an important regulator of apoptotic neuronal death. The p53 inactivator pifithrin-α (PFT-α) has been shown to be neuroprotective against stroke. A previous cellular study indicated that PFT-α oxygen analog (PFT-α (O)) is more stable and active than PFT-α. We aimed to investigate whether inhibition of p53 using PFT-α or PFT-α (O) would be a potential neuroprotective strategy for TBI. To evaluate whether these drugs protect against excitotoxicity in vitro, primary rat cortical cultures were challenged with glutamate (50mM) in the presence or absence of various concentrations of the p53 inhibitors PFT-α or PFT-α (O). Cell viability was estimated by LDH assay. In vivo, adult Sprague Dawley rats were subjected to controlled cortical impact (CCI, with 4m/s velocity, 2mm deformation). Five hours after injury, PFT-α or PFT-α (O) (2mg/kg, i.v.) was administered to animals. Sensory and motor functions were evaluated by behavioral tests at 24h after TBI. The p53-positive neurons were identified by double staining with cell-specific markers. Levels of mRNA encoding for p53-regulated genes (BAX, PUMA, Bcl-2 and p21) were measured by reverse transcription followed by real time-PCR from TBI animals without or with PFT-α/PFT-α (O) treatment. We found that PFT-α(O) (10 μM) enhanced neuronal survival against glutamate-induced cytotoxicity in vitro more effectively than PFT-α (10 μM). In vivo PFT-α (O) treatment enhanced functional recovery and decreased contusion volume at 24h post-injury. Neuroprotection by PFT-α (O) treatment also reduced p53-positive neurons in the cortical contusion region. In addition, p53-regulated PUMA mRNA levels at 8h were significantly reduced by PFT-α (O) administration after TBI. PFT-α (O) treatment also decreased phospho-p53 positive neurons in the cortical contusion region. Our data suggest that PFT-α (O) provided a significant reduction of cortical cell death and protected neurons from glutamate-induced excitotoxicity in vitro, as well as improved neurological functional outcome and reduced brain injury in vivo via anti-apoptotic mechanisms. The inhibition of p53-induced apoptosis by PFT-α (O) provides a useful tool to evaluate reversible apoptotic mechanisms and may develop into a novel therapeutic strategy for TBI.
Collapse
Affiliation(s)
- L-Y Yang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Y-H Chu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - D Tweedie
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, USA
| | - Q-S Yu
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, USA
| | - C G Pick
- Department of Anatomy and Anthropology, Sackler School of Medicine and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - B J Hoffer
- Department of Neurosurgery, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - N H Greig
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, USA
| | - J-Y Wang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
164
|
Bach-y-Rita P. Theoretical and Practical Considerations in the Restoration of Function After Stroke. Top Stroke Rehabil 2015; 8:1-15. [PMID: 14523734 DOI: 10.1310/8t1t-etxu-8pdf-9x7f] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Recovery of function after stroke appears to include elements of both restoration and compensation. The brain is highly plastic, which allows reorganization after damage. Stroke produces permanent damage to the brain, so recovery must be based on activity in surviving cells that are either adjacent, contralateral, or in a different region. Furthermore, representation of a particular function in the brain is usually not limited to a single brain region. Multiple representation provides the opportunity for brain reorganization; functions are assumed by surviving brain structures. Compensation can be the initial response in the recovery phase and may persist through later phases because of the new habits formed (as the restraint therapy studies of Taub and others would suggest), because the damage to the brain is extensive and hinders restoration, because of secondary pathology, such as tendon shortening and muscle wasting, that does not allow brain reorganization to be translated into functional recovery, or because of inadequate (especially late) rehabilitation.
Collapse
Affiliation(s)
- P Bach-y-Rita
- Department of Rehabilitation Medicine, University of Wisconsin-Madison, USA
| |
Collapse
|
165
|
Shutter LA, Ween JE. Mental Status and Behavioral Changes in the Early Postacute Phase of Care. Top Stroke Rehabil 2015; 9:39-47. [PMID: 14523716 DOI: 10.1310/2mm7-lafx-a0dp-3lvl] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Neurocognitive sequelae of stroke create a significant impact on the patient and frequently disrupt recovery from the physical manifestations. This article strives to review types of neurocognitive changes after stroke and discuss the underlying pathophysiology. Management options are presented for the diverse changes that may be encountered.
Collapse
Affiliation(s)
- Lori A Shutter
- NeuroTrauma Unit, Loma Linda University and Medical Center and Casa Colina Centers for Rehabilitation, Loma Linda, California, USA
| | | |
Collapse
|
166
|
Teasell R, Bitensky J, Foley N, Bayona NA. Training and Stimulation in Post Stroke Recovery Brain Reorganization. Top Stroke Rehabil 2015; 12:37-45. [PMID: 16110426 DOI: 10.1310/e893-m0pr-njej-1gxm] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
In both animal and clinical studies, training or rehabilitation increases cortical representation with subsequent functional recovery, whereas a lack of rehabilitation or training decreases cortical representation and delays recovery. Animals exposed to enriched environments post stroke have improved functional outcomes compared with animals exposed to nonenriched environments. In humans, stroke units may be the closest approximation there is to an enriched environment. However, studies indicate that patients spend the majority of time being inactive and alone while on a stroke unit. Given the animal evidence (which emphasizes increased stimulation and increased activity), there is clearly an opportunity for improving the stroke rehabilitation experience to maximize post stroke recovery.
Collapse
Affiliation(s)
- Robert Teasell
- Department of Physical Medicine and Rehabilitation, St. Joseph's Health Care and the University of Western Ontario, London, Ontario, Canada
| | | | | | | |
Collapse
|
167
|
Teasell R, Bitensky J, Salter K, Bayona NA. The Role of Timing and Intensity of Rehabilitation Therapies. Top Stroke Rehabil 2015; 12:46-57. [PMID: 16110427 DOI: 10.1310/etdp-6dr4-d617-vmvf] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
In both animal and clinical studies, motor rehabilitation and training increase cortical representation and improve recovery, whereas lack of training decreases cortical representation for particular motor functions. In animals, delays in providing rehabilitation reduce the impact of therapy with a worsening in motor outcomes and a corresponding reduction in cortical reorganization. In clinical studies, there is an association between earlier admission to rehabilitation and better outcomes that correlates with animal work both in terms of functional gains from chronic stroke deficits and cortical reorganization. There is a likely relationship between therapy intensity and improvements in functional outcomes. Clinically, greater intensity of stroke rehabilitation has been associated with improved outcomes.
Collapse
Affiliation(s)
- Robert Teasell
- Department of Physical Medicine and Rehabilitation, St. Joseph's Health Care and the University of Western Ontario, London, Ontario, Canada
| | | | | | | |
Collapse
|
168
|
Abstract
Impaired motor function after stroke is a major cause of disability in young stroke survivors. The plasticity of the adult human brain provides opportunities to enhance traditional rehabilitation programs for these individuals. Younger stroke patients appear to have a greater ability to recover from stroke and are likely to benefit substantially from treatments that facilitate plasticity-mediated recovery. The use of new exercise treatments, such as constraint-induced movement therapy, robot-aided rehabilitation, and partial body weight supported treadmill training are being studied intensively and are likely to ultimately be incorporated into standard poststroke rehabilitation. Medications to enhance recovery, growth factors, and stem cells will also be components of rehabilitation for the young stroke survivor in the foreseeable future.
Collapse
Affiliation(s)
- Joel Stein
- Spaulding Rehabilitation Hospital, Boston, Massachusetts, USA
| |
Collapse
|
169
|
Abstract
Neuroimaging techniques provide information on the neural substrates underlying functional recovery after stroke, the number one cause of long-term disability. Despite the methodological difficulties, they promise to offer insight into the mechanisms by which therapeutic interventions can modulate human cortical plasticity. This information should lead to the development of new, targeted interventions to maximize recovery.
Collapse
Affiliation(s)
- Timea Hodics
- Department of Neurology, Georgetown University Hospital, Washington, DC, USA
| | | |
Collapse
|
170
|
Kochanek PM, Jackson TC, Ferguson NM, Carlson SW, Simon DW, Brockman EC, Ji J, Bayir H, Poloyac SM, Wagner AK, Kline AE, Empey PE, Clark RS, Jackson EK, Dixon CE. Emerging therapies in traumatic brain injury. Semin Neurol 2015; 35:83-100. [PMID: 25714870 PMCID: PMC4356170 DOI: 10.1055/s-0035-1544237] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Despite decades of basic and clinical research, treatments to improve outcomes after traumatic brain injury (TBI) are limited. However, based on the recent recognition of the prevalence of mild TBI, and its potential link to neurodegenerative disease, many new and exciting secondary injury mechanisms have been identified and several new therapies are being evaluated targeting both classic and novel paradigms. This includes a robust increase in both preclinical and clinical investigations. Using a mechanism-based approach the authors define the targets and emerging therapies for TBI. They address putative new therapies for TBI across both the spectrum of injury severity and the continuum of care, from the field to rehabilitation. They discussTBI therapy using 11 categories, namely, (1) excitotoxicity and neuronal death, (2) brain edema, (3) mitochondria and oxidative stress, (4) axonal injury, (5) inflammation, (6) ischemia and cerebral blood flow dysregulation, (7) cognitive enhancement, (8) augmentation of endogenous neuroprotection, (9) cellular therapies, (10) combination therapy, and (11) TBI resuscitation. The current golden age of TBI research represents a special opportunity for the development of breakthroughs in the field.
Collapse
Affiliation(s)
- Patrick M. Kochanek
- Safar Center for Resuscitation Research, University of Pittburgh School of Medicine, Pittsburgh, Pennsylvania
- Departments of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Travis C. Jackson
- Safar Center for Resuscitation Research, University of Pittburgh School of Medicine, Pittsburgh, Pennsylvania
- Departments of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Nikki Miller Ferguson
- Safar Center for Resuscitation Research, University of Pittburgh School of Medicine, Pittsburgh, Pennsylvania
- Departments of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Shaun W. Carlson
- Safar Center for Resuscitation Research, University of Pittburgh School of Medicine, Pittsburgh, Pennsylvania
- Departmentol Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Dennis W. Simon
- Safar Center for Resuscitation Research, University of Pittburgh School of Medicine, Pittsburgh, Pennsylvania
- Departments of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Erik C. Brockman
- Safar Center for Resuscitation Research, University of Pittburgh School of Medicine, Pittsburgh, Pennsylvania
- Departments of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Jing Ji
- Safar Center for Resuscitation Research, University of Pittburgh School of Medicine, Pittsburgh, Pennsylvania
- Departments of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Hülya Bayir
- Safar Center for Resuscitation Research, University of Pittburgh School of Medicine, Pittsburgh, Pennsylvania
- Departments of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Environmental and Occupational Health, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Samuel M. Poloyac
- Safar Center for Resuscitation Research, University of Pittburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Environmental and Occupational Health, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Amy K. Wagner
- Safar Center for Resuscitation Research, University of Pittburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Anthony E. Kline
- Safar Center for Resuscitation Research, University of Pittburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Philip E. Empey
- Safar Center for Resuscitation Research, University of Pittburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Environmental and Occupational Health, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Robert S.B. Clark
- Safar Center for Resuscitation Research, University of Pittburgh School of Medicine, Pittsburgh, Pennsylvania
- Departments of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Edwin K. Jackson
- Safar Center for Resuscitation Research, University of Pittburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - C. Edward Dixon
- Safar Center for Resuscitation Research, University of Pittburgh School of Medicine, Pittsburgh, Pennsylvania
- Departmentol Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
171
|
Iaccarino MA, Bhatnagar S, Zafonte R. Rehabilitation after traumatic brain injury. HANDBOOK OF CLINICAL NEUROLOGY 2015; 127:411-22. [PMID: 25702231 DOI: 10.1016/b978-0-444-52892-6.00026-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Traumatic brain injury (TBI) is a growing problem in the US, with significant morbidity and economic implications. This diagnosis spans a wide breath of injuries from concussion to severe TBI. Thus, rehabilitation is equally diverse in its treatment strategies targeting those symptoms that are functionally limiting with the ultimate goal of independence and community reintegration. In severe TBI, rehabilitation can be lifelong. Acute care rehabilitation focuses on emergence from coma and prognostication of recovery. Therapeutic modalities and exercise, along with pharmacologic intervention, can target long-term motor and cognitive sequelae. Complications of severe TBI that are functionally limiting and impede therapy include heterotopic ossification, agitation, dysautonomia, and spasticity. In mild TBI, most patients recover quickly but education on repeat exposure is imperative, with the implications of consecutive injuries being potentially devastating. Furthermore, rehabilitation targets lingering symptoms including sleep disturbance, visuospatial deficits, headaches, and cognitive dysfunction. As research on the entire TBI population improves, commonalities in the disease process may emerge, helping rationalize therapeutic interventions and providing more robust targets for treatment.
Collapse
Affiliation(s)
- Mary Alexis Iaccarino
- Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital and Harvard Medical School, Boston, MA, USA
| | - Saurabha Bhatnagar
- Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital, Veterans Administration, Boston and Harvard Medical School, Boston, MA, USA
| | - Ross Zafonte
- Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital, Massachusetts General Hospital, Brigham and Woman's Hopsital, and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
172
|
Forsyth R, Basu A. The promotion of recovery through rehabilitation after acquired brain injury in children. Dev Med Child Neurol 2015; 57:16-22. [PMID: 25200439 DOI: 10.1111/dmcn.12575] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/15/2014] [Indexed: 12/12/2022]
Abstract
A degree of motor recovery is typically seen after acquired brain injury in children. The extent to which rehabilitation efforts can claim credit for this is disputed. Strong correlations between late impairment outcomes and early severity and impairment indices are seen both in adults and children. These correlations have been interpreted by some as evidence that recovery is largely intrinsic and that any additional rehabilitation effects are small. Such views are belied by published animal studies demonstrating the possibility of large rehabilitation effects. Animal models suggest that to achieve similar rehabilitation treatment effect sizes in clinical practice, rehabilitation 'doses' should be greater, rehabilitation efforts should start sooner, and premature accommodation of impairment should be avoided.
Collapse
Affiliation(s)
- Rob Forsyth
- Institute of Neuroscience, Sir James Spence Institute, Royal Victoria Infirmary, Newcastle University, Newcastle upon Tyne, Newcastle, UK
| | | |
Collapse
|
173
|
Zhu Y, Fotinos A, Mao LL, Atassi N, Zhou EW, Ahmad S, Guan Y, Berry JD, Cudkowicz ME, Wang X. Neuroprotective agents target molecular mechanisms of disease in ALS. Drug Discov Today 2015; 20:65-75. [DOI: 10.1016/j.drudis.2014.08.016] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Revised: 08/02/2014] [Accepted: 08/31/2014] [Indexed: 12/14/2022]
|
174
|
Coucha M, Li W, Hafez S, Abdelsaid M, Johnson MH, Fagan SC, Ergul A. SOD1 overexpression prevents acute hyperglycemia-induced cerebral myogenic dysfunction: relevance to contralateral hemisphere and stroke outcomes. Am J Physiol Heart Circ Physiol 2014; 308:H456-66. [PMID: 25552308 DOI: 10.1152/ajpheart.00321.2014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Admission hyperglycemia (HG) amplifies vascular injury and neurological deficits in acute ischemic stroke, but the mechanisms remain controversial. We recently reported that ischemia-reperfusion (I/R) injury impairs the myogenic response in both hemispheres via increased nitration. However, whether HG amplifies contralateral myogenic dysfunction and whether loss of tone in the contralateral hemisphere contributes to stroke outcomes remain to be determined. Our hypothesis was that contralateral myogenic dysfunction worsens stroke outcomes after acute hyperglycemic stroke in an oxidative stress-dependent manner. Male wild-type or SOD1 transgenic rats were injected with saline or 40% glucose solution 10 min before surgery and then subjected to 30 min of ischemia/45 min or 24 h of reperfusion. In another set of animals (n = 5), SOD1 was overexpressed only in the contralateral hemisphere by stereotaxic adenovirus injection 2-3 wk before I/R. Myogenic tone and neurovascular outcomes were determined. HG exacerbated myogenic dysfunction in contralateral side only, which was associated with infarct size expansion, increased edema, and more pronounced neurological deficit. Global and selective SOD1 overexpression restored myogenic reactivity in ipsilateral and contralateral sides, respectively, and enhanced neurovascular outcomes. In conclusion, our results show that SOD1 overexpression nullified the detrimental effects of HG on myogenic tone and stroke outcomes and that the contralateral hemisphere may be a novel target for the management of acute hyperglycemic stroke.
Collapse
Affiliation(s)
- Maha Coucha
- Department of Physiology, Georgia Regents University, Augusta, Georgia
| | - Weiguo Li
- Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia; Department of Physiology, Georgia Regents University, Augusta, Georgia
| | - Sherif Hafez
- Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia; Program in Clinical and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, Georgia
| | - Mohammed Abdelsaid
- Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia; Department of Physiology, Georgia Regents University, Augusta, Georgia
| | - Maribeth H Johnson
- Department of Biostatistics, Georgia Regents University, Augusta, Georgia
| | - Susan C Fagan
- Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia; Department of Neurology, Georgia Regents University, Augusta, Georgia; and Program in Clinical and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, Georgia
| | - Adviye Ergul
- Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia; Department of Physiology, Georgia Regents University, Augusta, Georgia; Program in Clinical and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, Georgia
| |
Collapse
|
175
|
Perez-Polo J, Rea H, Johnson K, Parsley M, Unabia G, Xu GY, Prough D, DeWitt D, Spratt H, Hulsebosch C. A rodent model of mild traumatic brain blast injury. J Neurosci Res 2014; 93:549-61. [DOI: 10.1002/jnr.23513] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2014] [Revised: 10/02/2014] [Accepted: 10/05/2014] [Indexed: 01/08/2023]
Affiliation(s)
| | - H.C. Rea
- University of Texas Medical Branch; Galveston Texas
| | - K.M. Johnson
- University of Texas Medical Branch; Galveston Texas
| | - M.A. Parsley
- University of Texas Medical Branch; Galveston Texas
| | - G.C. Unabia
- University of Texas Medical Branch; Galveston Texas
| | - G.-Y. Xu
- University of Texas Medical Branch; Galveston Texas
| | - D. Prough
- University of Texas Medical Branch; Galveston Texas
| | - D.S. DeWitt
- University of Texas Medical Branch; Galveston Texas
| | - H. Spratt
- University of Texas Medical Branch; Galveston Texas
| | | |
Collapse
|
176
|
Effectiveness of Amantadine Hydrochloride in the Reduction of Chronic Traumatic Brain Injury Irritability and Aggression. J Head Trauma Rehabil 2014; 29:391-9. [DOI: 10.1097/01.htr.0000438116.56228.de] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
177
|
Jeltsch-David H, Muller S. Neuropsychiatric systemic lupus erythematosus and cognitive dysfunction: the MRL-lpr mouse strain as a model. Autoimmun Rev 2014; 13:963-73. [PMID: 25183233 DOI: 10.1016/j.autrev.2014.08.015] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 07/10/2014] [Indexed: 12/19/2022]
Abstract
Mouse models of autoimmunity, such as (NZB×NZW)F1, MRL/MpJ-Fas(lpr) (MRL-lpr) and BXSB mice, spontaneously develop systemic lupus erythematosus (SLE)-like syndromes with heterogeneity and complexity that characterize human SLE. Despite their inherent limitations, such models have highly contributed to our current understanding of the pathogenesis of SLE as they provide powerful tools to approach the human disease at the genetic, cellular, molecular and environmental levels. They also allow novel treatment strategies to be evaluated in a complex integrated system, a favorable context knowing that very few murine models that adequately mimic human autoimmune diseases exist. As we move forward with more efficient medications to treat lupus patients, certain forms of the disease that requires to be better understood at the mechanistic level emerge. This is the case of neuropsychiatric (NP) events that affect 50-60% at SLE onset or within the first year after SLE diagnosis. Intense research performed at deciphering NP features in lupus mouse models has been undertaken. It is central to develop the first lead molecules aimed at specifically treating NPSLE. Here we discuss how mouse models, and most particularly MRL-lpr female mice, can be used for studying the pathogenesis of NPSLE in an animal setting, what are the NP symptoms that develop, and how they compare with human SLE, and, with a critical view, what are the neurobehavioral tests that are pertinent for evaluating the degree of altered functions and the progresses resulting from potentially active therapeutics.
Collapse
Affiliation(s)
- Hélène Jeltsch-David
- CNRS, Immunopathologie et chimie thérapeutique/Laboratory of excellence Medalis, Institut de Biologie Moléculaire et Cellulaire, 67000 Strasbourg, France.
| | - Sylviane Muller
- CNRS, Immunopathologie et chimie thérapeutique/Laboratory of excellence Medalis, Institut de Biologie Moléculaire et Cellulaire, 67000 Strasbourg, France.
| |
Collapse
|
178
|
Bhakta BB, Hartley S, Holloway I, Couzens JA, Ford GA, Meads D, Sackley CM, Walker MF, Ruddock SP, Farrin AJ. The DARS (Dopamine Augmented Rehabilitation in Stroke) trial: protocol for a randomised controlled trial of Co-careldopa treatment in addition to routine NHS occupational and physical therapy after stroke. Trials 2014; 15:316. [PMID: 25106447 PMCID: PMC4138395 DOI: 10.1186/1745-6215-15-316] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 07/21/2014] [Indexed: 11/20/2022] Open
Abstract
Background Stroke has a huge impact, leaving more than a third of affected people with lasting disability and rehabilitation remains a cornerstone treatment in the National Health Service (NHS). Recovery of mobility and arm function post-stroke occurs through re-learning to use the affected body parts and/or learning to compensate with the lesser affected side. Promising evidence suggests that the addition of Co-careldopa to physical therapy and occupational therapy may improve the recovery of arm and leg movement and lead to improved function. Methods/design Dopamine Augmented Rehabilitation in Stroke (DARS) is a multi-centre double-blind, randomised, placebo, controlled clinical trial of Co-careldopa in addition to routine NHS occupational therapy and physical therapy as part of early stroke rehabilitation. Participants will be randomised on a 1:1 basis to either Co-careldopa or placebo. The primary objective of the trial is to determine whether the addition of six weeks of Co-careldopa treatment to rehabilitation therapy can improve the proportion of patients who can walk independently eight weeks post-randomisation. Discussion The DARS trial will provide evidence as to whether Co-careldopa, in addition to routine NHS occupational and physical therapy, leads to a greater recovery of motor function, a reduction in carer dependency and advance rehabilitation treatments for people with stroke. Trial registration ISRCTN99643613 assigned on 4 December 2009.
Collapse
|
179
|
Yonkenafil: a novel phosphodiesterase type 5 inhibitor induces neuronal network potentiation by a cGMP-dependent Nogo-R axis in acute experimental stroke. Exp Neurol 2014; 261:267-77. [PMID: 25064698 DOI: 10.1016/j.expneurol.2014.07.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 06/14/2014] [Accepted: 07/04/2014] [Indexed: 11/21/2022]
Abstract
Yonkenafil is a novel phosphodiesterase type 5 (PDE5) inhibitor. Here we evaluated the effect of yonkenafil on ischemic injury and its possible mechanism of action. Male Sprague-Dawley rats underwent middle cerebral artery occlusion, followed by intraperitoneal or intravenous treatment with yonkenafil starting 2h later. Behavioral tests were carried out on day 1 or day 7 after reperfusion. Nissl staining, Fluoro-Jade B staining and electron microscopy studies were carried out 24h post-stroke, together with an analysis of infarct volume and severity of edema. Levels of cGMP-dependent Nogo-66 receptor (Nogo-R) pathway components, hsp70, apaf-1, caspase-3, caspase-9, synaptophysin, PSD-95/neuronal nitric oxide synthases (nNOS), brain-derived neurotrophic factor (BDNF)/tropomyosin-related kinase B (TrkB) and nerve growth factor (NGF)/tropomyosin-related kinase A (TrkA) were also measured after 24h. Yonkenafil markedly inhibited infarction and edema, even when administration was delayed until 4h after stroke onset. This protection was associated with an improvement in neurological function and was sustained for 7d. Yonkenafil enlarged the range of penumbra, reduced ischemic cell apoptosis and the loss of neurons, and modulated the expression of proteins in the Nogo-R pathway. Moreover, yonkenafil protected the structure of synapses and increased the expression of synaptophysin, BDNF/TrkB and NGF/TrkA. In conclusion, yonkenafil protects neuronal networks from injury after stroke.
Collapse
|
180
|
Ponsford J, Janzen S, McIntyre A, Bayley M, Velikonja D, Tate R. INCOG Recommendations for Management of Cognition Following Traumatic Brain Injury, Part I. J Head Trauma Rehabil 2014; 29:307-20. [DOI: 10.1097/htr.0000000000000074] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
181
|
Tosun C, Koltz MT, Kurland DB, Ijaz H, Gurakar M, Schwartzbauer G, Coksaygan T, Ivanova S, Gerzanich V, Simard JM. The protective effect of glibenclamide in a model of hemorrhagic encephalopathy of prematurity. Brain Sci 2014; 3:215-38. [PMID: 23667741 PMCID: PMC3647482 DOI: 10.3390/brainsci3010215] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
We studied a model of hemorrhagic encephalopathy of prematurity (EP) that closely recapitulates findings in humans with hemorrhagic EP. This model involves tandem insults of 20 min intrauterine ischemia (IUI) plus an episode of elevated venous pressure induced by intraperitoneal glycerol on post-natal day (P) 0. We examined Sur1 expression, which is upregulated after focal ischemia but has not been studied after brief global ischemia including IUI. We found that 20 min IUI resulted in robust upregulation of Sur1 in periventricular microvessels and tissues. We studied tandem insult pups from untreated or vehicle-treated dams (TI-CTR), and tandem insult pups from dams administered a low-dose, non-hypoglycemogenic infusion of the Sur1 blocker, glibenclamide, for 1 week after IUI (TI-GLIB). Compared to pups from the TI-CTR group, pups from the TI-GLIB group had significantly fewer and less severe hemorrhages on P1, performed significantly better on the beam walk and accelerating Rotarod on P35 and in tests of thigmotaxis and rapid learning on P35–49, and had significantly greater body and brain weights at P52. We conclude that low-dose glibenclamide administered to the mother at the end of pregnancy protects pups subjected to IUI from post-natal events of elevated venous pressure and its consequences.
Collapse
Affiliation(s)
- Cigdem Tosun
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; E-Mails: (C.T.); (M.T.K.); (D.B.K.); (H.I.); (M.G.); (G.S.); (S.I.); (V.G.)
| | - Michael T. Koltz
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; E-Mails: (C.T.); (M.T.K.); (D.B.K.); (H.I.); (M.G.); (G.S.); (S.I.); (V.G.)
| | - David B. Kurland
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; E-Mails: (C.T.); (M.T.K.); (D.B.K.); (H.I.); (M.G.); (G.S.); (S.I.); (V.G.)
| | - Hina Ijaz
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; E-Mails: (C.T.); (M.T.K.); (D.B.K.); (H.I.); (M.G.); (G.S.); (S.I.); (V.G.)
| | - Melda Gurakar
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; E-Mails: (C.T.); (M.T.K.); (D.B.K.); (H.I.); (M.G.); (G.S.); (S.I.); (V.G.)
| | - Gary Schwartzbauer
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; E-Mails: (C.T.); (M.T.K.); (D.B.K.); (H.I.); (M.G.); (G.S.); (S.I.); (V.G.)
| | - Turhan Coksaygan
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; E-Mail:
| | - Svetlana Ivanova
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; E-Mails: (C.T.); (M.T.K.); (D.B.K.); (H.I.); (M.G.); (G.S.); (S.I.); (V.G.)
| | - Volodymyr Gerzanich
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; E-Mails: (C.T.); (M.T.K.); (D.B.K.); (H.I.); (M.G.); (G.S.); (S.I.); (V.G.)
| | - J. Marc Simard
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; E-Mails: (C.T.); (M.T.K.); (D.B.K.); (H.I.); (M.G.); (G.S.); (S.I.); (V.G.)
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; E-Mail:
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-410-328-0850; Fax: +1-410-328-0124
| |
Collapse
|
182
|
Jia F, Yin YH, Gao GY, Wang Y, Cen L, Jiang JY. MMP-9 inhibitor SB-3CT attenuates behavioral impairments and hippocampal loss after traumatic brain injury in rat. J Neurotrauma 2014; 31:1225-34. [PMID: 24661104 DOI: 10.1089/neu.2013.3230] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The aim of this study was to evaluate the potential efficacy of SB-3CT, a matrix metallopeptidase 9 inhibitor, on behavioral and histological outcomes after traumatic brain injury (TBI) in rats. Adult male Sprague-Dawley rats were randomly divided into three groups (n=15/group): TBI with SB-3CT treatment, TBI with saline, and sham injury. The TBI model was induced by a fluid percussion TBI device. SB-3CT (50 mg/kg in 10% dimethyl sulfoxide) was administered intraperitoneally at 30 min, 6 h, and 12 h after the TBI. Motor function (beam-balance/beam-walk tests) and spatial learning/memory (Morris water maze) were assessed on post-operative Days 1-5 and 11-15, respectively. Fluoro-Jade staining, immunofluorescence, and cresyl violet-staining were carried out for histopathological evaluation at 24 h, 72 h, and 15 days after TBI, respectively. It was shown that TBI can result in significant behavioral deficit induced by acute neurodegeneration, increased expression of cleaved caspase-3, and long-term neuronal loss. SB-3CT intervention via the current regime provides robust behavioral protection and hippocampal neurons preservation from the deleterious effects of TBI. Hence, the efficacy of SB-3CT on TBI prognosis could be ascertained. It is believed that the current study adds to the growing literature in identifying SB-3CT as a potential therapy for human brain injury.
Collapse
Affiliation(s)
- Feng Jia
- 1 Department of Neurosurgery, Shanghai JiaoTong University , Shanghai, China
| | | | | | | | | | | |
Collapse
|
183
|
Pearson-Fuhrhop KM, Cramer SC. Pharmacogenetics of neural injury recovery. Pharmacogenomics 2014; 14:1635-43. [PMID: 24088134 DOI: 10.2217/pgs.13.152] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Relatively few pharmacological agents are part of routine care for neural injury, although several are used or under consideration in acute stroke, chronic stroke, traumatic brain injury and secondary stroke prevention. Tissue plasminogen activator is approved for the treatment of acute ischemic stroke, and genetic variants may impact the efficacy and safety of this drug. In the chronic phase of stroke, several drugs such as L-dopa, fluoxetine and donepezil are under investigation for enhancing rehabilitation therapy, with varying levels of evidence. One potential reason for the mixed efficacy displayed by these drugs may be the influence of genetic factors that were not considered in prior studies. An understanding of the genetics impacting the efficacy of dopaminergic, serotonergic and cholinergic drugs may allow clinicians to target these potential therapies to those patients most likely to benefit. In the setting of stroke prevention, which is directly linked to neural injury recovery, the most highly studied pharmacogenomic interactions pertain to clopidogrel and warfarin. Incorporating pharmacogenomics into neural injury recovery has the potential to maximize the benefit of several current and potential pharmacological therapies and to refine the choice of pharmacological agent that may be used to enhance benefits from rehabilitation therapy.
Collapse
Affiliation(s)
- Kristin M Pearson-Fuhrhop
- Department of Anatomy & Neurobiology, University of California, Irvine, 200 S Manchester Avenue, Suite 206, Orange, CA 92868, USA
| | | |
Collapse
|
184
|
Docosahexaenoic acid reduces ER stress and abnormal protein accumulation and improves neuronal function following traumatic brain injury. J Neurosci 2014; 34:3743-55. [PMID: 24599472 DOI: 10.1523/jneurosci.2872-13.2014] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
In this study, we investigated the development of endoplasmic reticulum (ER) stress after traumatic brain injury (TBI) and the efficacy of post-TBI administration of docosahexaenoic acid (DHA) in reducing ER stress. TBI was induced by cortical contusion injury in Sprague-Dawley rats. Either DHA (16 mg/kg in DMSO) or vehicle DMSO (1 ml/kg) was administered intraperitoneally at 5 min after TBI, followed by a daily dose for 3-21 d. TBI triggered sustained expression of the ER stress marker proteins including phosphorylated eukaryotic initiation factor-2α, activating transcription factor 4, inositol requiring kinase 1, and C/EBP homologous protein in the ipsilateral cortex at 3-21 d after TBI. The prolonged ER stress was accompanied with an accumulation of abnormal ubiquitin aggregates and increased expression of amyloid precursor protein (APP) and phosphorylated tau (p-Tau) in the frontal cortex after TBI. The ER stress marker proteins were colocalized with APP accumulation in the soma. Interestingly, administration of DHA attenuated all ER stress marker proteins and reduced the accumulation of both ubiquitinated proteins and APP/p-Tau proteins. In addition, the DHA-treated animals exhibited early recovery of their sensorimotor function after TBI. In summary, our study demonstrated that TBI induces a prolonged ER stress, which is positively correlated with abnormal APP accumulation. The sustained ER stress may play a role in chronic neuronal damage after TBI. Our findings illustrate that post-TBI administration of DHA has therapeutic potentials in reducing ER stress, abnormal protein accumulation, and neurological deficits.
Collapse
|
185
|
Valdez G, Heyer MP, Feng G, Sanes JR. The role of muscle microRNAs in repairing the neuromuscular junction. PLoS One 2014; 9:e93140. [PMID: 24664281 PMCID: PMC3963997 DOI: 10.1371/journal.pone.0093140] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2013] [Accepted: 03/01/2014] [Indexed: 01/11/2023] Open
Abstract
microRNAs have been implicated in mediating key aspects of skeletal muscle development and responses to diseases and injury. Recently, we demonstrated that a synaptically enriched microRNA, miR-206, functions to promote maintenance and repair of the neuromuscular junction (NMJ); in mutant mice lacking miR-206, reinnervation is impaired following nerve injury and loss of NMJs is accelerated in a mouse model of amyotrophic lateral sclerosis (ALS). Here, we asked whether other microRNAs play similar roles. One attractive candidate is miR-133b because it is in the same transcript that encodes miR-206. Like miR-206, miR-133b is concentrated near NMJs and induced after denervation. In miR-133b null mice, however, NMJ development is unaltered, reinnervation proceeds normally following nerve injury, and disease progression is unaffected in the SOD1(G93A) mouse model of ALS. To determine if miR-206 compensates for the loss of miR-133b, we generated mice lacking both microRNAs. The phenotype of these double mutants resembled that of miR-206 single mutants. Finally, we used conditional mutants of Dicer, an enzyme required for the maturation of most microRNAs, to generate mice in which microRNAs were depleted from skeletal muscle fibers postnatally, thus circumventing a requirement for microRNAs in embryonic muscle development. Reinnervation of muscle fibers following injury was impaired in these mice, but the defect was similar in magnitude to that observed in miR-206 mutants. Together, these results suggest that miR-206 is the major microRNA that regulates repair of the NMJ following nerve injury.
Collapse
Affiliation(s)
- Gregorio Valdez
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, Massachusetts, United States of America
- Virginia Tech Carilion Research Institute, Virginia Tech, Roanoke, Virginia, United States of America
- * E-mail:
| | - Mary P. Heyer
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Molecular Therapeutics, The Scripps Research Institute Florida, Jupiter, Florida, United States of America
| | - Guoping Feng
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina, United States of America
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Joshua R. Sanes
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, Massachusetts, United States of America
| |
Collapse
|
186
|
Vu Q, Xie K, Eckert M, Zhao W, Cramer SC. Meta-analysis of preclinical studies of mesenchymal stromal cells for ischemic stroke. Neurology 2014; 82:1277-86. [PMID: 24610327 DOI: 10.1212/wnl.0000000000000278] [Citation(s) in RCA: 151] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
OBJECTIVES To evaluate the quality of preclinical evidence for mesenchymal stromal cell (MSC) treatment of ischemic stroke, determine effect size of MSC therapy, and identify clinical measures that correlate with differences in MSC effects. METHODS A literature search identified studies of MSCs in animal models of cerebral ischemia. For each, a Quality Score was derived, and effect size of MSCs was determined for the most common behavioral and histologic endpoints. RESULTS Of 46 studies, 44 reported that MSCs significantly improved outcome. The median Quality Score was 5.5 (of 10). The median effect size was 1.78 for modified Neurological Severity Score, 1.73 for the adhesive removal test, 1.02 for the rotarod test, and 0.93 for infarct volume reduction. Quality Score correlated significantly and positively with effect size for the modified Neurological Severity Score. Effect sizes varied significantly with clinical measures such as administration route (intracerebral > intra-arterial > IV, although effect size for IV was nonetheless very large at 1.55) and species receiving MSCs (primate > rat > mouse). Because many MSC mechanisms are restorative, analyses were repeated examining only the 36 preclinical studies administering MSCs ≥ 24 hours poststroke; results were overall very similar. CONCLUSIONS In preclinical studies, MSCs have consistently improved multiple outcome measures, with very large effect sizes. Results were robust across species studied, administration route, species of MSC origin, timing, degree of immunogenicity, and dose, and in the presence of comorbidities. In contrast to meta-analyses of preclinical data for other stroke therapies, higher-quality MSC preclinical studies were associated with larger behavioral gains. These findings support the utility of further studies to translate MSCs in the treatment of ischemic stroke in humans.
Collapse
Affiliation(s)
- Quynh Vu
- From the Departments of Neurology and Anatomy & Neurobiology (Q.V., K.X., S.C.C.), Pharmaceutical Sciences, Biomedical Engineering, and Chao Family Comprehensive Cancer Center (M.E., W.Z.), and Sue and Bill Gross Stem Cell Research Center (Q.V., K.X., M.E., W.Z., S.C.C.), University of California, Irvine
| | | | | | | | | |
Collapse
|
187
|
Cramer SC, Hill MD. Human choriogonadotropin and epoetin alfa in acute ischemic stroke patients (REGENESIS-LED trial). Int J Stroke 2014; 9:321-7. [PMID: 24588854 DOI: 10.1111/ijs.12260] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2013] [Accepted: 10/14/2013] [Indexed: 11/28/2022]
Abstract
INTRODUCTION Preclinical studies suggest that growth factors in the early days after stroke improve final outcome. A prior study found three doses of human choriogonadotropin alfa followed by three doses of erythropoietin to be safe after stroke in humans. A proof of concept trial (REGENESIS) was initiated but placed on regulatory hold during review of an erythropoietin neuroprotective trial. Due to financial constraints, the trial was largely moved to India, using lower erythropoietin doses, as the REGENESIS-LED trial. METHODS Entry criteria included National Institutes of Health Stroke Scale 8-20, supratentorial ischemic stroke, and 24-48 h poststroke at start of therapy. Patients were randomized to three QOD doses of subcutaneous human choriogonadotropin alfa followed by three QD doses of intravenous erythropoietin (three escalating dose cohorts, 4000-20,000 IU/dose) vs. placebo. Primary outcomes were safety and neurological recovery. RESULTS The study was halted early by the sponsor after 96 enrollees. There was no significant difference across treatment groups in the proportion of patients experiencing death, serious adverse events, or any adverse event. There was no significant difference in National Institutes of Health Stroke Scale score change from baseline to Day 90 between placebo and active treatment, whether active cohorts were analyzed together or separately, and no exploratory secondary measure of neurological recovery showed a significant difference between groups. DISCUSSION Administration of human choriogonadotropin alfa followed by erythropoietin is safe after a new ischemic stroke. At the doses studied, placebo and active groups did not differ significantly in neurological recovery. Study limitations, such as the use of multiple assessors, differences in rehabilitation care, and being underpowered to show efficacy, are discussed.
Collapse
Affiliation(s)
- Steven C Cramer
- Departments of Neurology and Anatomy & Neurobiology, University of California, Irvine, CA, USA
| | | | | |
Collapse
|
188
|
Abstract
In the study, we used functional magnetic resonance imaging associated with behavioral assessment to observe the effects of venlafaxine on the modulation of human motor cortex activation and to provide preliminary data for further assessing its influence on motor functional reorganization after stroke injury. In a randomized, double-blind, crossover study, 8 right-handed subjects received 75 mg of either venlafaxine or a placebo daily over a period of 7 days separated by 3 washout days. The volunteers were asked to execute motor tasks, which included the dynamometer and finger-tapping test. In addition, laboratory tests and functional magnetic resonance imaging examination, before the start of the experiment and after administration of placebo and venlafaxine, were performed. It was shown that the finger-tapping rate of each hand in the venlafaxine stage was significantly improved compared with that observed in the placebo stage (n = 8, F left hand = 57.69, F right hand = 184.48, P < 0.001). The changes in the recorded grip strengths of both hands were not significant between the stages (n = 8, F = 2.63, P > 0.05). In the venlafaxine stage, the activations of the contralateral primary sensorimotor cortex, contralateral premotor cortex, and contralateral supplementary motor area were enhanced significantly, whereas the activation of the bilateral parietal cortices was decreased when compared with the placebo stage. Meanwhile, the enhancement of contralateral primary sensorimotor cortex activation had a positive correlation with the improvement of the finger-tapping rate. It was concluded that venlafaxine could modulate the cortical excitability and improve finger dexterity and reaction speed, which greatly related to the increase of contralateral motor cortical excitability.
Collapse
|
189
|
Abstract
Much progress has been made in understanding how behavioral experience and neural activity can modify the structure and function of neural circuits during development and in the adult brain. Studies of physiological and molecular mechanisms underlying activity-dependent plasticity in animal models have suggested potential therapeutic approaches for a wide range of brain disorders in humans. Physiological and electrical stimulations as well as plasticity-modifying molecular agents may facilitate functional recovery by selectively enhancing existing neural circuits or promoting the formation of new functional circuits. Here, we review the advances in basic studies of neural plasticity mechanisms in developing and adult nervous systems and current clinical treatments that harness neural plasticity, and we offer perspectives on future development of plasticity-based therapy.
Collapse
Affiliation(s)
- Karunesh Ganguly
- Department of Neurology & Rehabilitation, San Francisco VA Medical Center, 4150 Clement Street, San Francisco, CA 94121, USA; Department of Neurology, University of California, San Francisco, 400 Parnassus Avenue, San Francisco, CA 94122, USA.
| | | |
Collapse
|
190
|
Rau TF, Kothiwal AS, Rova AR, Brooks DM, Rhoderick JF, Poulsen AJ, Hutchinson J, Poulsen DJ. Administration of low dose methamphetamine 12 h after a severe traumatic brain injury prevents neurological dysfunction and cognitive impairment in rats. Exp Neurol 2013; 253:31-40. [PMID: 24333768 DOI: 10.1016/j.expneurol.2013.12.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Revised: 11/20/2013] [Accepted: 12/02/2013] [Indexed: 10/25/2022]
Abstract
We recently published data that showed low dose of methamphetamine is neuroprotective when delivered 3 h after a severe traumatic brain injury (TBI). In the current study, we further characterized the neuroprotective potential of methamphetamine by determining the lowest effective dose, maximum therapeutic window, pharmacokinetic profile and gene expression changes associated with treatment. Graded doses of methamphetamine were administered to rats beginning 8 h after severe TBI. We assessed neuroprotection based on neurological severity scores, foot fault assessments, cognitive performance in the Morris water maze, and histopathology. We defined 0.250 mg/kg/h as the lowest effective dose and treatment at 12 h as the therapeutic window following severe TBI. We examined gene expression changes following TBI and methamphetamine treatment to further define the potential molecular mechanisms of neuroprotection and determined that methamphetamine significantly reduced the expression of key pro-inflammatory signals. Pharmacokinetic analysis revealed that a 24-hour intravenous infusion of methamphetamine at a dose of 0.500 mg/kg/h produced a plasma Cmax value of 25.9 ng/ml and a total exposure of 544 ng/ml over a 32 hour time frame. This represents almost half the 24-hour total exposure predicted for a daily oral dose of 25mg in a 70 kg adult human. Thus, we have demonstrated that methamphetamine is neuroprotective when delivered up to 12 h after injury at doses that are compatible with current FDA approved levels.
Collapse
Affiliation(s)
- Thomas F Rau
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT 59812, USA
| | - Aakriti S Kothiwal
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT 59812, USA
| | - Annela R Rova
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT 59812, USA
| | - Diane M Brooks
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT 59812, USA
| | - Joseph F Rhoderick
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT 59812, USA
| | - Austin J Poulsen
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT 59812, USA
| | - Jim Hutchinson
- Montana Department of Justice Forensic Science Division, 2679 Palmer Street, Missoula, MT 59808, USA
| | - David J Poulsen
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT 59812, USA.
| |
Collapse
|
191
|
Deleterious Cognitive and Motoric Effects of Haloperidol in an Adolescent With Cerebral Palsy: A Case Report. PM R 2013; 5:1077-80. [DOI: 10.1016/j.pmrj.2013.07.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Revised: 07/15/2013] [Accepted: 07/16/2013] [Indexed: 01/11/2023]
|
192
|
Motor skills training promotes motor functional recovery and induces synaptogenesis in the motor cortex and striatum after intracerebral hemorrhage in rats. Behav Brain Res 2013; 260:34-43. [PMID: 24304717 DOI: 10.1016/j.bbr.2013.11.034] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Revised: 11/18/2013] [Accepted: 11/22/2013] [Indexed: 02/04/2023]
Abstract
We investigated the effects of motor skills training on several types of motor function and synaptic plasticity following intracerebral hemorrhage (ICH) in rats. Male Wistar rats were injected with collagenase into the left striatum to induce ICH, and they were randomly assigned to the ICH or sham groups. Each group was divided into the motor skills training (acrobatic training) and control (no exercise) groups. The acrobatic group performed acrobatic training from 4 to 28 days after surgery. Motor functions were assessed by motor deficit score, the horizontal ladder test and the wide or narrow beam walking test at several time points after ICH. The number of ΔFosB-positive cells was counted using immunohistochemistry to examine neuronal activation, and the PSD95 protein levels were analyzed by Western blotting to examine synaptic plasticity in the bilateral sensorimotor cortices and striata at 14 and 29 days after ICH. Motor skills training following ICH significantly improved gross motor function in the early phase after ICH and skilled motor coordinated function in the late phase. The number of ΔFosB-positive cells in the contralateral sensorimotor cortex in the acrobatic group significantly increased compared to the control group. PSD95 protein expression in the motor cortex significantly increased in the late phase, and in the striatum, the protein level significantly increased in the early phase by motor skills training after ICH compared to no training after ICH. We demonstrated that motor skills training improved motor function after ICH in rats and enhanced the neural activity and synaptic plasticity in the striatum and sensorimotor cortex.
Collapse
|
193
|
Comparative analysis of the neurovascular injury and functional outcomes in experimental stroke models in diabetic Goto-Kakizaki rats. Brain Res 2013; 1541:106-14. [PMID: 24144674 DOI: 10.1016/j.brainres.2013.10.021] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Revised: 10/09/2013] [Accepted: 10/11/2013] [Indexed: 12/25/2022]
Abstract
Diabetes worsens functional outcome and is associated with greater hemorrhagic transformation (HT) after ischemic stroke. We have shown that diabetic Goto-Kakizaki (GK) rats develop greater HT and neurological deficit despite smaller infarcts after transient middle cerebral artery occlusion (MCAO) with the suture model. However, the impact of (1) the duration of ischemia/reperfusion (I/R); (2) the method of ischemia; and (3) acute glycemic control on neurovascular injury and functional outcome in diabetic stroke remained unanswered. Wistar and GK rats were subjected to variable MCAO by suture or embolus occlusion. A group of GK rats were treated with insulin or metformin before stroke with suture occlusion. In all groups, infarct size, edema, HT occurrence and severity, and functional outcome were measured. Infarct size at 24h was smaller in GK rats with both suture and embolic MCAO, but expanded with longer reperfusion period. Edema and HT were increased in GK rats after 90min and 3h occlusion with the suture model, but not in the embolic MCAO. Neurological deficit was greater in diabetic rats. These findings suggest that diabetes accelerates the development of HT and amplifies vascular damage in the suture model where blood flow is rapidly reestablished. Acute metformin treatment worsened the infarct size, HT, and behavior outcome, whereas insulin treatment showed a protective effect. These results suggest that the impact of ischemia/reperfusion on neurovascular injury and functional outcome especially in disease models needs to be fully characterized using different models of stroke to model the human condition.
Collapse
|
194
|
Ikeda S, Ohwatashi A, Harada K, Kamikawa Y, Yoshida A. Expected for acquisition movement exercise is more effective for functional recovery than simple exercise in a rat model of hemiplegia. SPRINGERPLUS 2013; 2:517. [PMID: 24255829 PMCID: PMC3824718 DOI: 10.1186/2193-1801-2-517] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Accepted: 10/02/2013] [Indexed: 01/19/2023]
Abstract
Background and purpose The use of novel rehabilitative approaches for effecting functional recovery following stroke is controversial. Effects of different but effective rehabilitative interventions in the hemiplegic patient are not clear. We studied the effects of different rehabilitative approaches on functional recovery in the rat photochecmical cerebral infarction model. Methods Twenty-four male Wistar rats aged 8 weeks were used. The cranial bone was exposed under deep anesthesia. Rose bengal (20 mg/kg) was injected intravenously, and the sensorimotor area of the cerebral cortex was irradiated transcranially for 20 min with a light beam of 533-nm wavelength. Animals were divided into 3 groups. In the simple-exercise group, treadmill exercise was performed for 20 min every day. In the expected for acquisition movement-training group, beam-walking exercise was done for 20 min daily. The control group was left to recover without additional intervention. Hindlimb function was evaluated with the beam-walking test. Results Following cerebral infarction, dysfunction of the contralateral extremities was observed. Functional recovery was observed earlier in the expected for acquisition training group than in the other groups. Although rats in the treadmill group recovered more quickly than controls, the beam-walking group had the shortest overall recovery time. Conclusions Exercise facilitated functional recovery in the rat hemiplegic model, and expected for acquisition exercise was more effective than simple exercise. These findings are considered to have important implications for the future development of clinical rehabilitation programs.
Collapse
Affiliation(s)
- Satoshi Ikeda
- Department of Rehabilitation and Physical Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima-shi, Kagoshima, 890-8544 Japan
| | | | | | | | | |
Collapse
|
195
|
The locus coeruleus and cerebral metabolism: Recovery of function after cortical injury. ACTA ACUST UNITED AC 2013. [DOI: 10.3758/bf03326520] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
196
|
Eckert MA, Vu Q, Xie K, Yu J, Liao W, Cramer SC, Zhao W. Evidence for high translational potential of mesenchymal stromal cell therapy to improve recovery from ischemic stroke. J Cereb Blood Flow Metab 2013; 33:1322-34. [PMID: 23756689 PMCID: PMC3764389 DOI: 10.1038/jcbfm.2013.91] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Revised: 05/06/2013] [Accepted: 05/08/2013] [Indexed: 12/27/2022]
Abstract
Although ischemic stroke is a major cause of morbidity and mortality, current therapies benefit only a small proportion of patients. Transplantation of mesenchymal stromal cells (MSC, also known as mesenchymal stem cells or multipotent stromal cells) has attracted attention as a regenerative therapy for numerous diseases, including stroke. Mesenchymal stromal cells may aid in reducing the long-term impact of stroke via multiple mechanisms that include induction of angiogenesis, promotion of neurogenesis, prevention of apoptosis, and immunomodulation. In this review, we discuss the clinical rationale of MSC for stroke therapy in the context of their emerging utility in other diseases, and their recent clinical approval for treatment of graft-versus-host disease. An analysis of preclinical studies examining the effects of MSC therapy after ischemic stroke indicates near-universal agreement that MSC have significant favorable effect on stroke recovery, across a range of doses and treatment time windows. These results are interpreted in the context of completed and ongoing human clinical trials, which provide support for MSC as a safe and potentially efficacious therapy for stroke recovery in humans. Finally, we consider principles of brain repair and manufacturing considerations that will be useful for effective translation of MSC from the bench to the bedside for stroke recovery.
Collapse
Affiliation(s)
- Mark A Eckert
- Departments of Pharmaceutical Sciences and Biomedical Engineering, Sue and Bill Gross Stem Cell Research Center, Chao Family Comprehensive Cancer Center, University of California, Irvine, California, USA
| | - Quynh Vu
- Department of Neurology, Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, California, USA
| | - Kate Xie
- Department of Neurology, Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, California, USA
| | - Jingxia Yu
- Departments of Pharmaceutical Sciences and Biomedical Engineering, Sue and Bill Gross Stem Cell Research Center, Chao Family Comprehensive Cancer Center, University of California, Irvine, California, USA
| | - Wenbin Liao
- Department of Pathology, State University of New York at Stony Brook, Stony Brook, New York, USA
| | - Steven C Cramer
- Departments of Neurology and Anatomy and Neurobiology, Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, California, USA
| | - Weian Zhao
- Departments of Pharmaceutical Sciences and Biomedical Engineering, Sue and Bill Gross Stem Cell Research Center, Chao Family Comprehensive Cancer Center, University of California, Irvine, California, USA
| |
Collapse
|
197
|
Michalopoulou PG, Lewis SW, Wykes T, Jaeger J, Kapur S. Treating impaired cognition in schizophrenia: the case for combining cognitive-enhancing drugs with cognitive remediation. Eur Neuropsychopharmacol 2013; 23:790-8. [PMID: 23619163 DOI: 10.1016/j.euroneuro.2013.03.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Revised: 02/26/2013] [Accepted: 03/08/2013] [Indexed: 12/17/2022]
Abstract
Cognitive impairment is a well-documented feature of schizophrenia and represents a major impediment to the functional recovery of patients. The therapeutic strategies to improve cognition in schizophrenia have either used medications (collectively referred to as 'cognitive-enhancing drugs' in this article) or non-pharmacological training approaches ('cognitive remediation'). Cognitive-enhancing drugs have not as yet been successful and cognitive remediation has shown modest success. Therefore, we may need to explore new therapeutic paradigms to improve cognition in schizophrenia. The optimal approach may require a combination of cognitive-enhancing drugs with cognitive remediation. We review the available data from animal and human studies that provide the conceptual basis, proof-of-concept and illustrations of success of such combination strategies in experimental and clinical paradigms in other conditions. We address the major design issues relevant to the choice of the cognitive-enhancing drugs and cognitive remediation, as well as the timing and the duration of the intervention as will be relevant for schizophrenia. Finally, we address the practical realities of the development and testing of such combined approaches in the real-world clinical situation and conclude that while scientifically attractive, there are several practical difficulties to be overcome for this approach to be clinically feasible.
Collapse
Affiliation(s)
- Panayiota G Michalopoulou
- Section on Schizophrenia, Imaging and Therapeutics, Department of Psychosis Studies, Institute of Psychiatry, King's College London, UK.
| | | | | | | | | |
Collapse
|
198
|
Ren L, Zhang WA, Fang NY, Wang JX. The influence of electro-acupuncture on neural plasticity in acute cerebral infarction. Neurol Res 2013; 30:985-9. [DOI: 10.1179/174313208x325182] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
199
|
Efficacy of Amantadine Treatment on Symptoms and Neurocognitive Performance Among Adolescents Following Sports-Related Concussion. J Head Trauma Rehabil 2013; 28:260-5. [DOI: 10.1097/htr.0b013e318257fbc6] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
200
|
Effects of edaravone, a free radical scavenger, on photochemically induced cerebral infarction in a rat hemiplegic model. ScientificWorldJournal 2013; 2013:175280. [PMID: 23853531 PMCID: PMC3703327 DOI: 10.1155/2013/175280] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 06/06/2013] [Indexed: 11/17/2022] Open
Abstract
Edaravone is a free radical scavenger that protects the adjacent cortex during cerebral infarction. We created a hemiparetic model of cerebral thrombosis from a photochemically induced infarction with the photosensitive dye, rose bengal, in rats. We examined the effects of edaravone on recovery in the model. A total of 36 adult Wistar rats were used. The right sensorimotor area was irradiated with green light with a wavelength of 533 nm (10 mm diameter), and the rose bengal was injected intravenously to create an infarction. The edaravone group was injected intraperitoneally with edaravone (3 mg/kg), and the control group was injected with saline. The recovery process of the hemiplegia was evaluated with the 7-step scale of Fenny. The infarcted areas were measured after fixation. The recovery of the paralysis in the edaravone-treated group was significantly earlier than that in the untreated group. Seven days later, both groups were mostly recovered and had scores of 7, and the infarction region was significantly smaller in the edaravone-treated group. Edaravone reduced the infarction area and promoted the functional recovery of hemiparesis from cerebral thrombosis in a rat model. These findings suggest that edaravone treatment would be effective in clinical patients recovering from cerebral infarction.
Collapse
|