101
|
Lea-Carnall CA, Trujillo-Barreto NJ, Montemurro MA, El-Deredy W, Parkes LM. Evidence for frequency-dependent cortical plasticity in the human brain. Proc Natl Acad Sci U S A 2017; 114:8871-8876. [PMID: 28765375 PMCID: PMC5565407 DOI: 10.1073/pnas.1620988114] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Frequency-dependent plasticity (FDP) describes adaptation at the synapse in response to stimulation at different frequencies. Its consequence on the structure and function of cortical networks is unknown. We tested whether cortical "resonance," favorable stimulation frequencies at which the sensory cortices respond maximally, influenced the impact of FDP on perception, functional topography, and connectivity of the primary somatosensory cortex using psychophysics and functional imaging (fMRI). We costimulated two digits on the hand synchronously at, above, or below the resonance frequency of the somatosensory cortex, and tested subjects' accuracy and speed on tactile localization before and after costimulation. More errors and slower response times followed costimulation at above- or below-resonance, respectively. Response times were faster after at-resonance costimulation. In the fMRI, the cortical representations of the two digits costimulated above-resonance shifted closer, potentially accounting for the poorer performance. Costimulation at-resonance did not shift the digit regions, but increased the functional coupling between them, potentially accounting for the improved response time. To relate these results to synaptic plasticity, we simulated a network of oscillators incorporating Hebbian learning. Two neighboring patches embedded in a cortical sheet, mimicking the two digit regions, were costimulated at different frequencies. Network activation outside the stimulated patches was greatest at above-resonance frequencies, reproducing the spread of digit representations seen with fMRI. Connection strengths within the patches increased following at-resonance costimulation, reproducing the increased fMRI connectivity. We show that FDP extends to the cortical level and is influenced by cortical resonance.
Collapse
Affiliation(s)
- Caroline A Lea-Carnall
- Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, United Kingdom;
| | - Nelson J Trujillo-Barreto
- Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, United Kingdom
| | - Marcelo A Montemurro
- Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, United Kingdom
| | - Wael El-Deredy
- Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, United Kingdom
- School of Biomedical Engineering, University of Valparaiso, Valparaiso 2366103, Chile
| | - Laura M Parkes
- Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, United Kingdom
| |
Collapse
|
102
|
Payne SL, Anandakumaran PN, Varga BV, Morshead CM, Nagy A, Shoichet MS. In Vitro Maturation of Human iPSC-Derived Neuroepithelial Cells Influences Transplant Survival in the Stroke-Injured Rat Brain. Tissue Eng Part A 2017; 24:351-360. [PMID: 28594288 DOI: 10.1089/ten.tea.2016.0515] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Stem cell transplantation is a promising strategy for brain tissue regeneration; yet, despite some success, cell survival following transplantation remains low. In this study, we demonstrate that cell viability is enhanced by control over maturation of neuronal precursor cells, which are delivered in an injectable blend of hyaluronan and methylcellulose. We selected three subpopulations of human neuronal precursor cells derived from a cortically specified neuroepithelial stem cell (cNESC) population based on differences in expression of multipotent and neuron-specific proteins: early-, mid-, and late-differentiated neurons. These cells were transplanted into an endothelin-1 stroke-injured rat brain and their survival and fate were investigated 1 week later. Significantly, more cells were found in the brain after transplanting early- or mid- differentiated cNESCs compared to the late-differentiated population. The mid-differentiated population also had significantly more β-III tubulin-positive cells than either the early- or late-differentiated populations. These results suggest that maturity has a significant impact on cell survival following transplantation and cells with an intermediate maturity differentiate to neurons.
Collapse
Affiliation(s)
- Samantha L Payne
- 1 Department of Chemical Engineering and Applied Chemistry, University of Toronto , Toronto, Canada .,2 Institute of Biomaterials and Biomedical Engineering, University of Toronto , Toronto, Canada
| | - Priya N Anandakumaran
- 2 Institute of Biomaterials and Biomedical Engineering, University of Toronto , Toronto, Canada
| | - Balazs V Varga
- 3 Lunenfeld-Tanenbaum Research Institute , Mount Sinai Hospital, Toronto, Canada
| | - Cindi M Morshead
- 4 Institute of Medical Science, University of Toronto , Toronto Canada
| | - Andras Nagy
- 3 Lunenfeld-Tanenbaum Research Institute , Mount Sinai Hospital, Toronto, Canada
| | - Molly S Shoichet
- 1 Department of Chemical Engineering and Applied Chemistry, University of Toronto , Toronto, Canada .,2 Institute of Biomaterials and Biomedical Engineering, University of Toronto , Toronto, Canada .,5 Department of Chemistry, University of Toronto , Toronto, Canada
| |
Collapse
|
103
|
Thomaty S, Pezard L, Xerri C, Brezun JM. Acute granulocyte macrophage-colony stimulating factor treatment modulates neuroinflammatory processes and promotes tactile recovery after spinal cord injury. Neuroscience 2017; 349:144-164. [DOI: 10.1016/j.neuroscience.2017.02.035] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 02/17/2017] [Accepted: 02/17/2017] [Indexed: 11/25/2022]
|
104
|
Abstract
Stroke is the leading cause of complex adult disability in the world. Recovery from stroke is often incomplete, which leaves many people dependent on others for their care. The improvement of long-term outcomes should, therefore, be a clinical and research priority. As a result of advances in our understanding of the biological mechanisms involved in recovery and repair after stroke, therapeutic opportunities to promote recovery through manipulation of poststroke plasticity have never been greater. This work has almost exclusively been carried out in preclinical animal models of stroke with little translation into human studies. The challenge ahead is to develop a mechanistic understanding of recovery from stroke in humans. Advances in neuroimaging techniques now enable us to reconcile behavioural accounts of recovery with molecular and cellular changes. Consequently, clinical trials can be designed in a stratified manner that takes into account when an intervention should be delivered and who is most likely to benefit. This approach is expected to lead to a substantial change in how restorative therapeutic strategies are delivered in patients after stroke.
Collapse
|
105
|
Quattromani MJ, Pruvost M, Guerreiro C, Backlund F, Englund E, Aspberg A, Jaworski T, Hakon J, Ruscher K, Kaczmarek L, Vivien D, Wieloch T. Extracellular Matrix Modulation Is Driven by Experience-Dependent Plasticity During Stroke Recovery. Mol Neurobiol 2017; 55:2196-2213. [PMID: 28290150 PMCID: PMC5840227 DOI: 10.1007/s12035-017-0461-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Accepted: 02/16/2017] [Indexed: 11/24/2022]
Abstract
Following stroke, complete cellular death in the ischemic brain area may ensue, with remaining brain areas undergoing tissue remodelling to various degrees. Experience-dependent brain plasticity exerted through an enriched environment (EE) promotes remodelling after central nervous system injury, such as stroke. Post-stroke tissue reorganization is modulated by growth inhibitory molecules differentially expressed within the ischemic hemisphere, like chondroitin sulfate proteoglycans found in perineuronal nets (PNNs). PNNs in the neocortex predominantly enwrap parvalbumin-containing GABAergic (PV/GABA) neurons, important in sensori-information processing. Here, we investigate how extracellular matrix (ECM) proteases and their inhibitors may participate in the regulation of PNN integrity during stroke recovery. Rats were subjected to photothrombotic stroke in the motor cortex, and functional deficits were assessed at 7 days of recovery. Sham and stroked rats were housed in either standard or EE conditions for 5 days, and infarct volumes were calculated. PNNs were visualized by immunohistochemistry and counted in the somatosensory cortex of both hemispheres. mRNA expression levels of ECM proteases and protease inhibitors were assessed by RT-qPCR and their activity analyzed by gel zymography. PNNs and protease activity were also studied in brains from stroke patients where similar results were observed. EE starting 2 days after stroke and continuing for 5 days stimulated behavioral recovery of limb-placement ability without affecting infarct size. EE promoted a decrease of PNNs around PV/GABA neurons and a concomitant modulation of the proteolytic activity and mRNA expression of ECM proteases and protease inhibitors in the somatosensory cortex. This study provides molecular targets for novel therapies that could support rehabilitation of stroke patients.
Collapse
Affiliation(s)
- Miriana Jlenia Quattromani
- Laboratory for Experimental Brain Research, Division of Neurosurgery, Department of Clinical Sciences, Lund University, BMC A13, 22184, Lund, Sweden.
| | - Mathilde Pruvost
- INSERM UMR-S U919, Serine Proteases and Pathophysiology of the Neurovascular Unit, Université Caen Basse Normandie, GIP Cyceron, F-14074, Caen, France
| | - Carla Guerreiro
- Laboratory for Experimental Brain Research, Division of Neurosurgery, Department of Clinical Sciences, Lund University, BMC A13, 22184, Lund, Sweden
| | - Fredrik Backlund
- Laboratory for Experimental Brain Research, Division of Neurosurgery, Department of Clinical Sciences, Lund University, BMC A13, 22184, Lund, Sweden
| | - Elisabet Englund
- Division of Oncology and Pathology, Lund University Hospital, 22185, Lund, Sweden
| | - Anders Aspberg
- Rheumatology and Molecular Skeletal Biology, Department of Clinical Sciences, Lund University, BMC C12, 22184, Lund, Sweden
| | - Tomasz Jaworski
- Laboratory of Neurobiology, Nencki Institute of Experimental Biology, 02-093, Warsaw, Poland
| | - Jakob Hakon
- Laboratory for Experimental Brain Research, Division of Neurosurgery, Department of Clinical Sciences, Lund University, BMC A13, 22184, Lund, Sweden
| | - Karsten Ruscher
- Laboratory for Experimental Brain Research, Division of Neurosurgery, Department of Clinical Sciences, Lund University, BMC A13, 22184, Lund, Sweden
| | - Leszek Kaczmarek
- Laboratory of Neurobiology, Nencki Institute of Experimental Biology, 02-093, Warsaw, Poland
| | - Denis Vivien
- INSERM UMR-S U919, Serine Proteases and Pathophysiology of the Neurovascular Unit, Université Caen Basse Normandie, GIP Cyceron, F-14074, Caen, France.,Department of Clinical Research, Caen University Hospital, CHU Caen, 14000, Caen, France
| | - Tadeusz Wieloch
- Laboratory for Experimental Brain Research, Division of Neurosurgery, Department of Clinical Sciences, Lund University, BMC A13, 22184, Lund, Sweden
| |
Collapse
|
106
|
Mouthon AL, Meyer-Heim A, Kurth S, Ringli M, Pugin F, van Hedel HJA, Huber R. High-Density Electroencephalographic Recordings During Sleep in Children and Adolescents With Acquired Brain Injury. Neurorehabil Neural Repair 2017; 31:462-474. [PMID: 28162033 DOI: 10.1177/1545968316688794] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Acquired brain injuries (ABI) such as traumatic brain injury (TBI) or stroke can result in motor, language, or cognitive impairments. Although a considerable number of studies have investigated functional recovery, underlying brain reorganization remains poorly understood. Accumulating evidence indicates that plastic processes in the brain are linked to changes in electroencephalographic (EEG) slow wave activity (SWA) during deep sleep (EEG spectral power 1-4.5 Hz). OBJECTIVE We investigated sleep SWA in children and adolescents with ABI. METHODS We used high-density EEG (128 electrodes) to record sleep in 22 young patients with ABI (age range = 4-16 years). We compared patients to 52 previously measured typically developing children and adolescents (age range = 4-16 years). RESULTS The pattern of alterations in SWA differed between particular patient groups. In patients with bilateral stroke, SWA was globally reduced across the entire scalp. Patients with unilateral stroke showed a local reduction in SWA over lesion areas and an increase over perilesional and contralateral brain areas. In patients with severe TBI, we found a reduction in SWA over the midline and an increase over lateral brain areas. We found no consistent pattern in patients with mild to moderate TBI. CONCLUSIONS Sleep SWA seems to be a sensitive measure to assess individual alterations in neural activity after ABI. Deviations from age norms might indirectly indicate plastic processes that have occurred since injury. Improving our understanding of neural activity after ABI could optimize clinical prognosis and guide the development of novel therapeutic interventions.
Collapse
Affiliation(s)
- Anne-Laure Mouthon
- 1 Child Development Center and Pediatric Sleep Disorders Center, University Children's Hospital Zurich, Switzerland.,2 Rehabilitation Center Affoltern am Albis, University Children's Hospital Zurich, Switzerland.,3 Children's Research Center, University Children's Hospital Zurich, Switzerland
| | - Andreas Meyer-Heim
- 2 Rehabilitation Center Affoltern am Albis, University Children's Hospital Zurich, Switzerland.,3 Children's Research Center, University Children's Hospital Zurich, Switzerland
| | - Salome Kurth
- 1 Child Development Center and Pediatric Sleep Disorders Center, University Children's Hospital Zurich, Switzerland.,3 Children's Research Center, University Children's Hospital Zurich, Switzerland.,5 Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Maya Ringli
- 1 Child Development Center and Pediatric Sleep Disorders Center, University Children's Hospital Zurich, Switzerland.,3 Children's Research Center, University Children's Hospital Zurich, Switzerland
| | - Fiona Pugin
- 1 Child Development Center and Pediatric Sleep Disorders Center, University Children's Hospital Zurich, Switzerland.,3 Children's Research Center, University Children's Hospital Zurich, Switzerland
| | - Hubertus J A van Hedel
- 2 Rehabilitation Center Affoltern am Albis, University Children's Hospital Zurich, Switzerland.,3 Children's Research Center, University Children's Hospital Zurich, Switzerland
| | - Reto Huber
- 1 Child Development Center and Pediatric Sleep Disorders Center, University Children's Hospital Zurich, Switzerland.,3 Children's Research Center, University Children's Hospital Zurich, Switzerland.,4 Department of Child and Adolescent Psychiatry and Psychotherapy, Psychiatric Hospital, University of Zurich, Switzerland
| |
Collapse
|
107
|
Sandvig I, Gadjanski I, Vlaski-Lafarge M, Buzanska L, Loncaric D, Sarnowska A, Rodriguez L, Sandvig A, Ivanovic Z. Strategies to Enhance Implantation and Survival of Stem Cells After Their Injection in Ischemic Neural Tissue. Stem Cells Dev 2017; 26:554-565. [PMID: 28103744 DOI: 10.1089/scd.2016.0268] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
High post-transplantation cell mortality is the main limitation of various approaches that are aimed at improving regeneration of injured neural tissue by an injection of neural stem cells (NSCs) and mesenchymal stromal cells (MStroCs) in and/or around the lesion. Therefore, it is of paramount importance to identify efficient ways to increase cell transplant viability. We have previously proposed the "evolutionary stem cell paradigm," which explains the association between stem cell anaerobic/microaerophilic metabolic set-up and stem cell self-renewal and inhibition of differentiation. Applying these principles, we have identified the main critical point in the collection and preparation of these cells for experimental therapy: exposure of the cells to atmospheric O2, that is, to oxygen concentrations that are several times higher than the physiologically relevant ones. In this way, the primitive anaerobic cells become either inactivated or adapted, through commitment and differentiation, to highly aerobic conditions (20%-21% O2 in atmospheric air). This inadvertently compromises the cells' survival once they are transplanted into normal tissue, especially in the hypoxic/anoxic/ischemic environment, which is typical of central nervous system (CNS) lesions. In addition to the findings suggesting that stem cells can shift to glycolysis and can proliferate in anoxia, recent studies also propose that stem cells may be able to proliferate in completely anaerobic or ischemic conditions by relying on anaerobic mitochondrial respiration. In this systematic review, we propose strategies to enhance the survival of NSCs and MStroCs that are implanted in hypoxic/ischemic neural tissue by harnessing their anaerobic nature and maintaining as well as enhancing their anaerobic properties via appropriate ex vivo conditioning.
Collapse
Affiliation(s)
- Ioanna Sandvig
- 1 Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Ivana Gadjanski
- 2 Innovation Center, Faculty of Mechanical Engineering, University of Belgrade , Belgrade, Serbia .,3 Belgrade Metropolitan University , Belgrade, Serbia
| | - Marija Vlaski-Lafarge
- 4 French Blood Institute (EFS) , Aquitaine-Limousin Branch, Bordeaux, France .,5 U1035 INSERM/Bordeaux University , Bordeaux Cedex, France
| | - Leonora Buzanska
- 6 Stem Cell Bioengineering Unit, Mossakowski Medical Research Centre Polish Academy Sciences, Warsaw, Poland
| | - Darija Loncaric
- 4 French Blood Institute (EFS) , Aquitaine-Limousin Branch, Bordeaux, France .,5 U1035 INSERM/Bordeaux University , Bordeaux Cedex, France
| | - Ana Sarnowska
- 6 Stem Cell Bioengineering Unit, Mossakowski Medical Research Centre Polish Academy Sciences, Warsaw, Poland
| | - Laura Rodriguez
- 4 French Blood Institute (EFS) , Aquitaine-Limousin Branch, Bordeaux, France .,5 U1035 INSERM/Bordeaux University , Bordeaux Cedex, France
| | - Axel Sandvig
- 1 Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway .,7 Division of Pharmacology and Clinical Neurosciences, Department of Neurosurgery and Clinical Neurophysiology, Umeå University Hospital , Umeå, Sweden
| | - Zoran Ivanovic
- 4 French Blood Institute (EFS) , Aquitaine-Limousin Branch, Bordeaux, France .,5 U1035 INSERM/Bordeaux University , Bordeaux Cedex, France
| |
Collapse
|
108
|
Kokaia Z, Tornero D, Lindvall O. Transplantation of reprogrammed neurons for improved recovery after stroke. PROGRESS IN BRAIN RESEARCH 2017; 231:245-263. [PMID: 28554399 DOI: 10.1016/bs.pbr.2016.11.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Somatic cells such as fibroblasts, reprogrammed to induced pluripotent stem cells, can be used to generate neural stem/progenitor cells or neuroblasts for transplantation. In this review, we summarize recent studies demonstrating that when grafted intracerebrally in animal models of stroke, reprogrammed neurons improve function, probably by several different mechanisms, e.g., trophic actions, modulation of inflammation, promotion of angiogenesis, cellular and synaptic plasticity, and neuroprotection. In our own work, we have shown that human skin-derived reprogrammed neurons, fated to cortical progeny, integrate in stroke-injured neuronal network and form functional afferent synapses with host neurons, responding to peripheral sensory stimulation. However, whether neuronal replacement plays a role for the improvement of sensory, motor, and cognitive deficits after transplantation of reprogrammed neurons is still unclear. We conclude that further preclinical studies are needed to understand the therapeutic potential of grafted reprogrammed neurons and to define a road map for their clinical translation in stroke.
Collapse
Affiliation(s)
- Zaal Kokaia
- Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, Lund, Sweden.
| | - Daniel Tornero
- Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, Lund, Sweden
| | - Olle Lindvall
- Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, Lund, Sweden
| |
Collapse
|
109
|
Duss SB, Seiler A, Schmidt MH, Pace M, Adamantidis A, Müri RM, Bassetti CL. The role of sleep in recovery following ischemic stroke: A review of human and animal data. Neurobiol Sleep Circadian Rhythms 2017; 2:94-105. [PMID: 31236498 PMCID: PMC6575180 DOI: 10.1016/j.nbscr.2016.11.003] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Revised: 11/04/2016] [Accepted: 11/07/2016] [Indexed: 01/02/2023] Open
Abstract
Despite advancements in understanding the pathophysiology of stroke and the state of the art in acute management of afflicted patients as well as in subsequent neurorehabilitation training, stroke remains the most common neurological cause of long-term disability in adulthood. To enhance stroke patients' independence and well-being it is necessary, therefore, to consider and develop new therapeutic strategies and approaches. We postulate that sleep might play a pivotal role in neurorehabilitation following stroke. Over the last two decades compelling evidence for a major function of sleep in neuroplasticity and neural network reorganization underlying learning and memory has evolved. Training and learning of new motor skills and knowledge can modulate the characteristics of subsequent sleep, which additionally can improve memory performance. While healthy sleep appears to support neuroplasticity resulting in improved learning and memory, disturbed sleep following stroke in animals and humans can impair stroke outcome. In addition, sleep disorders such as sleep disordered breathing, insomnia, and restless legs syndrome are frequent in stroke patients and associated with worse recovery outcomes. Studies investigating the evolution of post-stroke sleep changes suggest that these changes might also reflect neural network reorganization underlying functional recovery. Experimental and clinical studies provide evidence that pharmacological sleep promotion in rodents and treatment of sleep disorders in humans improves functional outcome following stroke. Taken together, there is accumulating evidence that sleep represents a "plasticity state" in the process of recovery following ischemic stroke. However, to test the key role of sleep and sleep disorders for stroke recovery and to better understand the underlying molecular mechanisms, experimental research and large-scale prospective studies in humans are necessary. The effects of hospital conditions, such as adjusting light conditions according to the patients' sleep-wake rhythms, or sleep promoting drugs and non-invasive brain stimulation to promote neuronal plasticity and recovery following stroke requires further investigation.
Collapse
Affiliation(s)
- Simone B. Duss
- Sleep-Wake-Epilepsy-Center, Department of Neurology, Bern University Hospital, Bern, Switzerland
| | - Andrea Seiler
- Sleep-Wake-Epilepsy-Center, Department of Neurology, Bern University Hospital, Bern, Switzerland
| | - Markus H. Schmidt
- Sleep-Wake-Epilepsy-Center, Department of Neurology, Bern University Hospital, Bern, Switzerland
- Center for Experimental Neurology (ZEN), Department of Neurology, Bern University Hospital, Bern, Switzerland
| | - Marta Pace
- Center for Experimental Neurology (ZEN), Department of Neurology, Bern University Hospital, Bern, Switzerland
| | - Antoine Adamantidis
- Center for Experimental Neurology (ZEN), Department of Neurology, Bern University Hospital, Bern, Switzerland
| | - René M. Müri
- Division of Cognitive and Restorative Neurology, Department of Neurology, Bern University Hospital, Bern, Switzerland
| | - Claudio L. Bassetti
- Sleep-Wake-Epilepsy-Center, Department of Neurology, Bern University Hospital, Bern, Switzerland
- Center for Experimental Neurology (ZEN), Department of Neurology, Bern University Hospital, Bern, Switzerland
- Division of Cognitive and Restorative Neurology, Department of Neurology, Bern University Hospital, Bern, Switzerland
| |
Collapse
|
110
|
Nie J, Yang X. Modulation of Synaptic Plasticity by Exercise Training as a Basis for Ischemic Stroke Rehabilitation. Cell Mol Neurobiol 2017; 37:5-16. [PMID: 26910247 PMCID: PMC11482112 DOI: 10.1007/s10571-016-0348-1] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 02/11/2016] [Indexed: 12/23/2022]
Abstract
In recent years, rehabilitation of ischemic stroke draws more and more attention in the world, and has been linked to changes of synaptic plasticity. Exercise training improves motor function of ischemia as well as cognition which is associated with formation of learning and memory. The molecular basis of learning and memory might be synaptic plasticity. Research has therefore been conducted in an attempt to relate effects of exercise training to neuroprotection and neurogenesis adjacent to the ischemic injury brain. The present paper reviews the current literature addressing this question and discusses the possible mechanisms involved in modulation of synaptic plasticity by exercise training. This review shows the pathological process of synaptic dysfunction in ischemic roughly and then discusses the effects of exercise training on scaffold proteins and regulatory protein expression. The expression of scaffold proteins generally increased after training, but the effects on regulatory proteins were mixed. Moreover, the compositions of postsynaptic receptors were changed and the strength of synaptic transmission was enhanced after training. Finally, the recovery of cognition is critically associated with synaptic remodeling in an injured brain, and the remodeling occurs through a number of local regulations including mRNA translation, remodeling of cytoskeleton, and receptor trafficking into and out of the synapse. We do provide a comprehensive knowledge of synaptic plasticity enhancement obtained by exercise training in this review.
Collapse
Affiliation(s)
- Jingjing Nie
- Department of Neurology, Xiang Ya Hospital, Central South University, Xiang Ya Road 87, Changsha, 410008, Hunan, China
| | - Xiaosu Yang
- Department of Neurology, Xiang Ya Hospital, Central South University, Xiang Ya Road 87, Changsha, 410008, Hunan, China.
| |
Collapse
|
111
|
Jiang L, Hu Y, He X, Lv Q, Wang TH, Xia QJ. Breviscapine reduces neuronal injury caused by traumatic brain injury insult: partly associated with suppression of interleukin-6 expression. Neural Regen Res 2017; 12:90-95. [PMID: 28250753 PMCID: PMC5319248 DOI: 10.4103/1673-5374.198990] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Breviscapine, extracted from the herb Erigeron breviscapus, is widely used for the treatment of cardiovascular diseases, cerebral infarct, and stroke, but its mechanism of action remains unclear. This study established a rat model of traumatic brain injury induced by controlled cortical impact, and injected 75 μg breviscapine via the right lateral ventricle. We found that breviscapine significantly improved neurobehavioral dysfunction at 6 and 9 days after injection. Meanwhile, interleukin-6 expression was markedly down-regulated following breviscapine treatment. Our results suggest that breviscapine is effective in promoting neurological behavior after traumatic brain injury and the underlying molecular mechanism may be associated with the suppression of interleukin-6.
Collapse
Affiliation(s)
- Ling Jiang
- Institute of Neurological Disease, Department of Anesthesiology and Translation Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yue Hu
- Institute of Neurological Disease, Department of Anesthesiology and Translation Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Xiang He
- Institute of Neurological Disease, Department of Anesthesiology and Translation Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Qiang Lv
- Institute of Neurological Disease, Department of Anesthesiology and Translation Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Ting-Hua Wang
- Institute of Neurological Disease, Department of Anesthesiology and Translation Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Qing-Jie Xia
- Institute of Neurological Disease, Department of Anesthesiology and Translation Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| |
Collapse
|
112
|
Stokowska A, Atkins AL, Morán J, Pekny T, Bulmer L, Pascoe MC, Barnum SR, Wetsel RA, Nilsson JA, Dragunow M, Pekna M. Complement peptide C3a stimulates neural plasticity after experimental brain ischaemia. Brain 2016; 140:353-369. [PMID: 27956400 DOI: 10.1093/brain/aww314] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 10/19/2016] [Accepted: 10/20/2016] [Indexed: 11/12/2022] Open
Abstract
Ischaemic stroke induces endogenous repair processes that include proliferation and differentiation of neural stem cells and extensive rewiring of the remaining neural connections, yet about 50% of stroke survivors live with severe long-term disability. There is an unmet need for drug therapies to improve recovery by promoting brain plasticity in the subacute to chronic phase after ischaemic stroke. We previously showed that complement-derived peptide C3a regulates neural progenitor cell migration and differentiation in vitro and that C3a receptor signalling stimulates neurogenesis in unchallenged adult mice. To determine the role of C3a-C3a receptor signalling in ischaemia-induced neural plasticity, we subjected C3a receptor-deficient mice, GFAP-C3a transgenic mice expressing biologically active C3a in the central nervous system, and their respective wild-type controls to photothrombotic stroke. We found that C3a overexpression increased, whereas C3a receptor deficiency decreased post-stroke expression of GAP43 (P < 0.01), a marker of axonal sprouting and plasticity, in the peri-infarct cortex. To verify the translational potential of these findings, we used a pharmacological approach. Daily intranasal treatment of wild-type mice with C3a beginning 7 days after stroke induction robustly increased synaptic density (P < 0.01) and expression of GAP43 in peri-infarct cortex (P < 0.05). Importantly, the C3a treatment led to faster and more complete recovery of forepaw motor function (P < 0.05). We conclude that C3a-C3a receptor signalling stimulates post-ischaemic neural plasticity and intranasal treatment with C3a receptor agonists is an attractive approach to improve functional recovery after ischaemic brain injury.
Collapse
Affiliation(s)
- Anna Stokowska
- Center for Brain Repair and Rehabilitation, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Alison L Atkins
- Center for Brain Repair and Rehabilitation, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Javier Morán
- Center for Brain Repair and Rehabilitation, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Tulen Pekny
- Center for Brain Repair and Rehabilitation, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Linda Bulmer
- Center for Brain Repair and Rehabilitation, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Michaela C Pascoe
- Center for Brain Repair and Rehabilitation, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Scott R Barnum
- Department of Microbiology, University of Alabama, Birmingham, Alabama, USA
| | - Rick A Wetsel
- Research Center for Immunology and Autoimmune Diseases, Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas-Houston, Houston, Texas, USA
| | - Jonas A Nilsson
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Mike Dragunow
- Department of Pharmacology and Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Marcela Pekna
- Center for Brain Repair and Rehabilitation, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden .,Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia.,Hunter Medical Research Institute, University of Newcastle, New South Wales, Australia
| |
Collapse
|
113
|
Shi J, Dong B, Mao Y, Guan W, Cao J, Zhu R, Wang S. Review: Traumatic brain injury and hyperglycemia, a potentially modifiable risk factor. Oncotarget 2016; 7:71052-71061. [PMID: 27626493 PMCID: PMC5342608 DOI: 10.18632/oncotarget.11958] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 09/02/2016] [Indexed: 12/13/2022] Open
Abstract
Hyperglycemia after severe traumatic brain injury (TBI) occurs frequently and is associated with poor clinical outcome and increased mortality. In this review, we highlight the mechanisms that lead to hyperglycemia and discuss how they may contribute to poor outcomes in patients with severe TBI. Moreover, we systematically review the proper management of hyperglycemia after TBI, covering topics such as nutritional support, glucose control, moderated hypothermia, naloxone, and mannitol treatment. However, to date, an optimal and safe glycemic target range has not been determined, and may not be safe to implement among TBI patients. Therefore, there is a mandate to explore a reasonable glycemic target range that can facilitate recovery after severe TBI.
Collapse
Affiliation(s)
- Jia Shi
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Bo Dong
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Yumin Mao
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Wei Guan
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Jiachao Cao
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Rongxing Zhu
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Suinuan Wang
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou, China
| |
Collapse
|
114
|
Klarić TS, Jaehne EJ, Koblar SA, Baune BT, Lewis MD. Alterations in anxiety and social behaviour in Npas4 deficient mice following photochemically-induced focal cortical stroke. Behav Brain Res 2016; 316:29-37. [PMID: 27574128 DOI: 10.1016/j.bbr.2016.08.050] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 08/21/2016] [Accepted: 08/25/2016] [Indexed: 01/04/2023]
Abstract
In addition to causing widespread cell death and loss of brain function, cerebral ischaemia also induces extensive neuroplasticity. In humans, stroke is often accompanied by severe cognitive and psychiatric changes that are thought to arise as a consequence of this infarct-induced remodelling. A candidate for producing these post-stroke neuropsychiatric changes is Npas4, an activity-dependent transcription factor involved in synaptic plasticity whose expression is aberrantly up-regulated following ischaemic injury. In this study we investigated the role of Npas4 in modulating these stroke-induced neuropsychiatric responses by comparing the performance of wildtype and Npas4-/- mice in various cognitive and behavioural tasks in a photochemical model of focal cortical stroke. We show that this stroke model results in impaired spatial recognition memory and a reduction in despair-like behaviour that affect both genotypes to a similar degree. Moreover, mice lacking Npas4 also show differences in some aspects of post-stroke sociability and anxiety. Specifically, we show that while stroke had no effect on anxiety levels in wildtype mice, Npas4-/- mice became significantly more anxious following stroke. In addition, Npas4-/- mice retained a greater level of sociability in the acute post-stroke period in comparison to their wildtype littermates. Thus, our findings suggest that Npas4 may be involved in post-stroke psychiatric changes related to anxiety and sociability.
Collapse
Affiliation(s)
- T S Klarić
- School of Medicine, University of Adelaide, North Terrace, Adelaide, SA, 5005, Australia
| | - E J Jaehne
- Discipline of Psychiatry, University of Adelaide, North Terrace, Adelaide, SA, 5005, Australia
| | - S A Koblar
- School of Medicine, University of Adelaide, North Terrace, Adelaide, SA, 5005, Australia
| | - B T Baune
- Discipline of Psychiatry, University of Adelaide, North Terrace, Adelaide, SA, 5005, Australia
| | - M D Lewis
- South Australian Health & Medical Research Institute, North Terrace, Adelaide, SA, 5000, Australia.
| |
Collapse
|
115
|
Dong T, Zhi L, Bhayana B, Wu MX. Cortisol-induced immune suppression by a blockade of lymphocyte egress in traumatic brain injury. J Neuroinflammation 2016; 13:197. [PMID: 27561600 PMCID: PMC5000452 DOI: 10.1186/s12974-016-0663-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 07/18/2016] [Indexed: 12/16/2022] Open
Abstract
Background Acute traumatic brain injury (TBI) represents one of major causes of mortality and disability in the USA. Neuroinflammation has been regarded both beneficial and detrimental, probably in a time-dependent fashion. Methods To address a role for neuroinflammation in brain injury, C57BL/6 mice were subjected to a closed head mild TBI (mTBI) by a standard controlled cortical impact, along with or without treatment of sphingosine 1-phosphate (S1P) or rolipram, after which the brain tissue of the impact site was evaluated for cell morphology via histology, inflammation by qRT-PCR and T cell staining, and cell death with Caspase-3 and TUNEL staining. Circulating lymphocytes were quantified by flow cytometry, and plasma hydrocortisone was analyzed by LC-MS/MS. To investigate the mechanism whereby cortisol lowered the number of peripheral T cells, T cell egress was tracked in lymph nodes by intravital confocal microscopy after hydrocortisone administration. Results We detected a decreased number of circulating lymphocytes, in particular, T cells soon after mTBI, which was inversely correlated with a transient and robust increase of plasma cortisol. The transient lymphocytopenia might be caused by cortisol in part via a blockade of lymphocyte egress as demonstrated by the ability of cortisol to inhibit T cell egress from the secondary lymphoid tissues. Moreover, exogenous hydrocortisone severely suppressed periphery lymphocytes in uninjured mice, whereas administering an egress-promoting agent S1P normalized circulating T cells in mTBI mice and increased T cells in the injured brain. Likewise, rolipram, a cAMP phosphodiesterase inhibitor, was also able to elevate cAMP levels in T cells in the presence of hydrocortisone in vitro and abrogate the action of cortisol in mTBI mice. The investigation demonstrated that the number of circulating T cells in the early phase of TBI was positively correlated with T cell infiltration and inflammatory responses as well as cell death at the cerebral cortex and hippocampus beneath the impact site. Conclusions Decreases in intracellular cAMP might be part of the mechanism behind cortisol-mediated blockade of T cell egress. The study argues strongly for a protective role of cortisol-induced immune suppression in the early stage of TBI.
Collapse
Affiliation(s)
- Tingting Dong
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, 50 Blossom Street, Boston, MA, 02114, USA
| | - Liang Zhi
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, 50 Blossom Street, Boston, MA, 02114, USA
| | - Brijesh Bhayana
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, 50 Blossom Street, Boston, MA, 02114, USA
| | - Mei X Wu
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, 50 Blossom Street, Boston, MA, 02114, USA.
| |
Collapse
|
116
|
Fink SB. A Deeper Look at the "Neural Correlate of Consciousness". Front Psychol 2016; 7:1044. [PMID: 27507950 PMCID: PMC4960249 DOI: 10.3389/fpsyg.2016.01044] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 06/27/2016] [Indexed: 11/22/2022] Open
Abstract
A main goal of the neuroscience of consciousness is: find the neural correlate to conscious experiences (NCC). When have we achieved this goal? The answer depends on our operationalization of “NCC.” Chalmers (2000) shaped the widely accepted operationalization according to which an NCC is a neural system with a state which is minimally sufficient (but not necessary) for an experience. A deeper look at this operationalization reveals why it might be unsatisfactory: (i) it is not an operationalization of a correlate for occurring experiences, but of the capacity to experience; (ii) it is unhelpful for certain cases which are used to motivate a search for neural correlates of consciousness; (iii) it does not mirror the usage of “NCC” by scientists who seek for unique correlates; (iv) it hardly allows for a form of comparative testing of hypotheses, namely experimenta crucis. Because of these problems (i–iv), we ought to amend or improve on Chalmers's operationalization. Here, I present an alternative which avoids these problems. This “NCC2.0” also retains some benefits of Chalmers's operationalization, namely being compatible with contributions from extended, embedded, enacted, or embodied accounts (4E-accounts) and allowing for the possibility of non-biological or artificial experiencers.
Collapse
Affiliation(s)
- Sascha Benjamin Fink
- Institute 3, Philosophy-Neuroscience-Cognition Program, Otto-von-Guericke-Universität Magdeburg Magdeburg, Germany
| |
Collapse
|
117
|
Sherwood MS, Weisend MP, Kane JH, Parker JG. Combining Real-Time fMRI Neurofeedback Training of the DLPFC with N-Back Practice Results in Neuroplastic Effects Confined to the Neurofeedback Target Region. Front Behav Neurosci 2016; 10:138. [PMID: 27445733 PMCID: PMC4923251 DOI: 10.3389/fnbeh.2016.00138] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 06/15/2016] [Indexed: 12/16/2022] Open
Abstract
In traditional fMRI, individuals respond to exogenous stimuli and are naïve to the effects of the stimuli on their neural activity patterns. Changes arising in the fMRI signal are analyzed post-hoc to elucidate the spatial and temporal activation of brain regions associated with the tasks performed. The advent of real-time fMRI has enabled a new method to systematically alter brain activity across space and time using neurofeedback training (NFT), providing a new tool to study internally-driven processes such as neuroplasticity. In this work, we combined n-back practice with fMRI-NFT of the left dorsolateral prefrontal cortex (DLPFC) to better understand the relationship between open- and closed-loop neuromodulation. FMRI data were acquired during both traditional n-back and NFT across five imaging sessions. Region-of-interest (ROI) and voxel-wise 2 × 2 within subjects ANOVAs were carried out to determine the effects of, and interaction between, training session and neuromodulation type. A main effect of training session was identified for only a single, highly focused cluster that shared spatial properties with the fMRI-NFT target region (left DLPFC). This finding indicates that combined open- and closed-loop neuroplastic enhancement techniques result in focal changes that are confined to the target area of NFT, and do not affect up- or down-stream network components that are normally engaged during working memory. Additionally, we identified a main effect of neuromodulation type for 15 clusters with significantly different activation between open- and closed-loop neuromodulation during training, 12 of which demonstrated higher activity during the open-loop neuromodulation. Our results, taken together with previous reports, indicate that fMRI-NFT combined with n-back practice leads to a highly focal volume exhibiting neuroplasticity without additional network effects.
Collapse
Affiliation(s)
- Matthew S Sherwood
- Wright State Research Institute, Wright State UniversityDayton, OH, USA; Department of Biomedical, Industrial, and Human Factors Engineering, Wright State UniversityDayton, OH, USA
| | | | - Jessica H Kane
- Wright State Research Institute, Wright State UniversityDayton, OH, USA; Department of Neuroscience, Cell Biology, and Physiology, Wright State UniversityDayton, OH, USA
| | - Jason G Parker
- Department of Biomedical, Industrial, and Human Factors Engineering, Wright State UniversityDayton, OH, USA; Department of Neurology, Boonshoft School of Medicine, Wright State UniversityDayton, OH, USA
| |
Collapse
|
118
|
Cortical Reorganization following Injury Early in Life. Neural Plast 2016; 2016:8615872. [PMID: 27298741 PMCID: PMC4889847 DOI: 10.1155/2016/8615872] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2015] [Accepted: 04/17/2016] [Indexed: 11/29/2022] Open
Abstract
The brain has a remarkable capacity for reorganization following injury, especially during the first years of life. Knowledge of structural reorganization and its consequences following perinatal injury is sparse. Here we studied changes in brain tissue volume, morphology, perfusion, and integrity in children with hemiplegia compared to typically developing children, using MRI. Children with hemiplegia demonstrated reduced total cerebral volume, with increased cerebrospinal fluid (CSF) and reduced total white matter volumes, with no differences in total gray matter volume, compared to typically developing children. An increase in cortical thickness at the hemisphere contralateral to the lesion (CLH) was detected in motor and language areas, which may reflect compensation for the gray matter loss in the lesion area or retention of ipsilateral pathways. In addition, reduced cortical thickness, perfusion, and surface area were detected in limbic areas. Increased CSF volume and precentral cortical thickness and reduced white matter volume were correlated with worse motor performance. Brain reorganization of the gray matter within the CLH, while not necessarily indicating better outcome, is suggested as a response to neuronal deficits following injury early in life.
Collapse
|
119
|
Mohanty CB, Bhat D, Devi BI. Role of Central Plasticity in the Outcome of Peripheral Nerve Regeneration. Neurosurgery 2016; 77:418-23. [PMID: 26087003 DOI: 10.1227/neu.0000000000000851] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
The optimal refinement in nerve repair techniques has reached a plateau, making it imperative to continually explore newer avenues for improving the clinical outcome of peripheral nerve regeneration. The aim of this short review is to discuss the role and mechanism of brain plasticity in nerve regeneration, as well as to explore the possible application of this knowledge for improving the clinical outcome following nerve repair.
Collapse
Affiliation(s)
- Chandan B Mohanty
- Department of Neurosurgery, National Institute of Mental Health and Neurosciences, Bangalore, India
| | | | | |
Collapse
|
120
|
Mouthon AL, van Hedel HJA, Meyer-Heim A, Kurth S, Ringli M, Pugin F, Huber R. High-density electroencephalographic recordings during sleep in children with disorders of consciousness. NEUROIMAGE-CLINICAL 2016; 11:468-475. [PMID: 27104141 PMCID: PMC4827803 DOI: 10.1016/j.nicl.2016.03.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 02/17/2016] [Accepted: 03/17/2016] [Indexed: 11/20/2022]
Abstract
Introduction A large number of studies have investigated neural correlates of consciousness in adults. However, knowledge about brain function in children with disorders of consciousness (DOC) is very limited. We suggest that EEG recordings during sleep are a promising approach. In healthy adults as well as in children, it has been shown that the activity of sleep slow waves (EEG spectral power 1–4.5 Hz), the primary characteristic of deep sleep, is dependent on use during previous wakefulness. Thus the regulation of slow wave activity (SWA) provides indirect insights into brain function during wakefulness. Methods In the present study, we investigated high-density EEG recordings during sleep in ten healthy children and in ten children with acquired brain injury, including five children with DOC and five children with acquired brain injury without DOC. We used the build-up of SWA to quantify SWA regulation. Results Children with DOC showed a global reduction in the SWA build-up when compared to both, healthy children and children with acquired brain injury without DOC. This reduction was most pronounced over parietal brain areas. Comparisons within the group of children with DOC revealed that the parietal SWA build-up was the lowest in patients showing poor outcome. Longitudinal measurements during the recovery period showed an increase in parietal SWA build-up from the first to the second sleep recording. Conclusions Our results suggest that the reduced parietal SWA regulation may represent a characteristic topographical marker for brain network dysfunction in children with DOC. In the future, the regulation of SWA might be used as a complementary assessment in adult and paediatric patients with DOC. Longitudinal high-density EEG recording in children with disorders of consciousness Sleep electrophysiology provides a marker for brain network dysfunction. The sleep EEG might be used as a complementary assessment in paediatric patients.
Collapse
Affiliation(s)
- Anne-Laure Mouthon
- Child Development Centre and Paediatric Sleep Disorders Centre, University Children's Hospital Zurich, Switzerland; Rehabilitation Centre Affoltern am Albis, University Children's Hospital Zurich, Switzerland; Children's Research Centre, University Children's Hospital Zurich, Switzerland
| | - Hubertus J A van Hedel
- Rehabilitation Centre Affoltern am Albis, University Children's Hospital Zurich, Switzerland; Children's Research Centre, University Children's Hospital Zurich, Switzerland
| | - Andreas Meyer-Heim
- Rehabilitation Centre Affoltern am Albis, University Children's Hospital Zurich, Switzerland; Children's Research Centre, University Children's Hospital Zurich, Switzerland
| | - Salome Kurth
- Child Development Centre and Paediatric Sleep Disorders Centre, University Children's Hospital Zurich, Switzerland; Children's Research Centre, University Children's Hospital Zurich, Switzerland; Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Maya Ringli
- Child Development Centre and Paediatric Sleep Disorders Centre, University Children's Hospital Zurich, Switzerland; Children's Research Centre, University Children's Hospital Zurich, Switzerland
| | - Fiona Pugin
- Child Development Centre and Paediatric Sleep Disorders Centre, University Children's Hospital Zurich, Switzerland; Children's Research Centre, University Children's Hospital Zurich, Switzerland
| | - Reto Huber
- Child Development Centre and Paediatric Sleep Disorders Centre, University Children's Hospital Zurich, Switzerland; Children's Research Centre, University Children's Hospital Zurich, Switzerland; University Clinics for Child and Adolescent Psychiatry, University of Zurich, Switzerland.
| |
Collapse
|
121
|
Zepeda R, Contreras V, Pissani C, Stack K, Vargas M, Owen GI, Lazo OM, Bronfman FC. Venlafaxine treatment after endothelin-1-induced cortical stroke modulates growth factor expression and reduces tissue damage in rats. Neuropharmacology 2016; 107:131-145. [PMID: 26965219 DOI: 10.1016/j.neuropharm.2016.03.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 03/03/2016] [Accepted: 03/05/2016] [Indexed: 02/02/2023]
Abstract
Neuromodulators, such as antidepressants, may contribute to neuroprotection by modulating growth factor expression to exert anti-inflammatory effects and to support neuronal plasticity after stroke. Our objective was to study whether early treatment with venlafaxine, a serotonin-norepinephrine reuptake inhibitor, modulates growth factor expression and positively contributes to reducing the volume of infarcted brain tissue resulting in increased functional recovery. We studied the expression of BDNF, FGF2 and TGF-β1 by examining their mRNA and protein levels and cellular distribution using quantitative confocal microscopy at 5 days after venlafaxine treatment in control and infarcted brains. Venlafaxine treatment did not change the expression of these growth factors in sham rats. In infarcted rats, BDNF mRNA and protein levels were reduced, while the mRNA and protein levels of FGF2 and TGF-β1 were increased. Venlafaxine treatment potentiated all of the changes that were induced by cortical stroke alone. In particular, increased levels of FGF2 and TGF-β1 were observed in astrocytes at 5 days after stroke induction, and these increases were correlated with decreased astrogliosis (measured by GFAP) and increased synaptophysin immunostaining at twenty-one days after stroke in venlafaxine-treated rats. Finally, we show that venlafaxine reduced infarct volume after stroke resulting in increased functional recovery, which was measured using ladder rung motor tests, at 21 days after stroke. Our results indicate that the early oral administration of venlafaxine positively contributes to neuroprotection during the acute and late events that follow stroke.
Collapse
Affiliation(s)
- Rodrigo Zepeda
- MINREB and Center for Aging and Regeneration (CARE UC), Faculty of Biological Sciences, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Valentina Contreras
- MINREB and Center for Aging and Regeneration (CARE UC), Faculty of Biological Sciences, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Claudia Pissani
- MINREB and Center for Aging and Regeneration (CARE UC), Faculty of Biological Sciences, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Katherine Stack
- MINREB and Center for Aging and Regeneration (CARE UC), Faculty of Biological Sciences, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Macarena Vargas
- Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Gareth I Owen
- Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Oscar M Lazo
- MINREB and Center for Aging and Regeneration (CARE UC), Faculty of Biological Sciences, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | - Francisca C Bronfman
- MINREB and Center for Aging and Regeneration (CARE UC), Faculty of Biological Sciences, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
122
|
Kinoshita S, Kakuda W, Yamada N, Momosaki R, Okuma R, Watanabe S, Abo M. Therapeutic administration of atomoxetine combined with rTMS and occupational therapy for upper limb hemiparesis after stroke: a case series study of three patients. Acta Neurol Belg 2016; 116:31-7. [PMID: 26123130 DOI: 10.1007/s13760-015-0503-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 06/18/2015] [Indexed: 11/24/2022]
Abstract
Atomoxetine, a selective noradrenaline reuptake inhibitor, has been reported to enhance brain plasticity, but has not yet been used in stroke patients. We reported the feasibility and clinical benefits on motor functional recovery of the combination of repetitive transcranial magnetic stimulation (rTMS) and intensive occupational therapy (OT) in stroke patients. This pilot study was designed to evaluate the additive effects of oral atomoxetine to rTMS/OT in post-stroke hemiparetic patients. The study included three post-stroke patients with upper limb hemiparesis. Treatment with 40 mg/day atomoxetine commenced 2 weeks before admission. After confirming tolerance, the dose was increased to 120 mg/day. Low-frequency rTMS/OT was provided daily for 15 days during continued atomoxetine therapy. Motor function of the affected upper limb was evaluated with the Fugl-Meyer Assessment and Wolf Motor Function test. All patients completed the protocol and showed motor improvement up to 4 weeks after the treatment. No atomoxetine-related side effects were noted. Our protocol of triple therapy of atomoxetine, low-frequency rTMS, and OT is safe and feasible intervention for upper limb hemiparesis after stroke.
Collapse
Affiliation(s)
- Shoji Kinoshita
- Department of Rehabilitation Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-Ku, Tokyo, 105-8461, Japan
| | - Wataru Kakuda
- Department of Rehabilitation Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-Ku, Tokyo, 105-8461, Japan
| | - Naoki Yamada
- Department of Rehabilitation Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-Ku, Tokyo, 105-8461, Japan
| | - Ryo Momosaki
- Department of Rehabilitation Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-Ku, Tokyo, 105-8461, Japan
| | - Ryo Okuma
- Department of Rehabilitation Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-Ku, Tokyo, 105-8461, Japan
| | - Shu Watanabe
- Department of Rehabilitation Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-Ku, Tokyo, 105-8461, Japan
| | - Masahiro Abo
- Department of Rehabilitation Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-Ku, Tokyo, 105-8461, Japan.
| |
Collapse
|
123
|
|
124
|
Compensation through Functional Hyperconnectivity: A Longitudinal Connectome Assessment of Mild Traumatic Brain Injury. Neural Plast 2015; 2016:4072402. [PMID: 26819765 PMCID: PMC4706919 DOI: 10.1155/2016/4072402] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 08/07/2015] [Accepted: 08/11/2015] [Indexed: 11/18/2022] Open
Abstract
Mild traumatic brain injury (mTBI) is a major public health concern. Functional MRI has reported alterations in several brain networks following mTBI. However, the connectome-scale brain network changes are still unknown. In this study, sixteen mTBI patients were prospectively recruited from an emergency department and followed up at 4-6 weeks after injury. Twenty-four healthy controls were also scanned twice with the same time interval. Three hundred fifty-eight brain landmarks that preserve structural and functional correspondence of brain networks across individuals were used to investigate longitudinal brain connectivity. Network-based statistic (NBS) analysis did not find significant difference in the group-by-time interaction and time effects. However, 258 functional pairs show group differences in which mTBI patients have higher functional connectivity. Meta-analysis showed that "Action" and "Cognition" are the most affected functional domains. Categorization of connectomic signatures using multiview group-wise cluster analysis identified two patterns of functional hyperconnectivity among mTBI patients: (I) between the posterior cingulate cortex and the association areas of the brain and (II) between the occipital and the frontal lobes of the brain. Our results demonstrate that brain concussion renders connectome-scale brain network connectivity changes, and the brain tends to be hyperactivated to compensate the pathophysiological disturbances.
Collapse
|
125
|
Ramos-Languren LE, González-Piña R, Montes S, Chávez-García N, Ávila-Luna A, Barón-Flores V, Ríos C. Sensorimotor recovery from cortical injury is accompanied by changes on norepinephrine and serotonin levels in the dentate gyrus and pons. Behav Brain Res 2015; 297:297-306. [PMID: 26454240 DOI: 10.1016/j.bbr.2015.10.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 10/02/2015] [Accepted: 10/03/2015] [Indexed: 01/16/2023]
Abstract
Monoamines such as norepinephrine (NE) and serotonin (5-HT) have shown to play an important role in motor recovery after brain injury. The effects elicited by these neurotransmitters have been reported as distal from the area directly affected. Remote changes may take place over minutes to weeks and play an important role in post-stroke recovery. However, the mechanisms involved in spontaneous recovery have not been thoroughly delineated. Therefore, we determined the NE and 5-HT content, in the pons and hippocampal dentate gyrus (DG) as well as motor deficit and spontaneous activity in rats after 3, 10 and 20 days cortical iron injection. Three days post-lesion the pontine NE content diminished, this effect was accompanied by deficient spontaneous activity and impaired sensorimotor evaluation. Ten and twenty days after lesion the NE levels were similar to those of control group, and animals also showed behavioral recovery. Monoamines content on DG 3 days post-lesion showed no differences as compared to controls. Interestingly, ten and twenty days after cortical injury, animals showed increased NE and 5-HT. These results suggest that behavioral recovery after brain damage involve changes on monoamines levels on DG, an important structure to plastic processes. In addition, the results herein support evidence to propose these neurotransmitters as key molecules to functional recovery in the central nervous system.
Collapse
Affiliation(s)
- Laura E Ramos-Languren
- Depto. de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía, MVS, SSA, Mexico City, Mexico; Maestría en Ciencias Farmacéuticas, Universidad Autónoma Metropolitana, Unidad Xochimilco, Mexico City, Mexico; Laboratorio de Neuroplasticidad, División de Neurociencias, Torre de Investigación, Instituto Nacional de Rehabilitacion, Mexico City, Mexico
| | - Rigoberto González-Piña
- Laboratorio de Neuroplasticidad, División de Neurociencias, Torre de Investigación, Instituto Nacional de Rehabilitacion, Mexico City, Mexico
| | - Sergio Montes
- Depto. de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía, MVS, SSA, Mexico City, Mexico
| | - Norma Chávez-García
- Depto. de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía, MVS, SSA, Mexico City, Mexico
| | - Alberto Ávila-Luna
- Laboratorio de Neuroplasticidad, División de Neurociencias, Torre de Investigación, Instituto Nacional de Rehabilitacion, Mexico City, Mexico
| | - Verónica Barón-Flores
- Neurofarmalogía Molecular, Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana, Unidad Xochimilco, Mexico City, Mexico
| | - Camilo Ríos
- Depto. de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía, MVS, SSA, Mexico City, Mexico; Maestría en Ciencias Farmacéuticas, Universidad Autónoma Metropolitana, Unidad Xochimilco, Mexico City, Mexico; Neurofarmalogía Molecular, Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana, Unidad Xochimilco, Mexico City, Mexico.
| |
Collapse
|
126
|
Lake EMR, Chaudhuri J, Thomason L, Janik R, Ganguly M, Brown M, McLaurin J, Corbett D, Stanisz GJ, Stefanovic B. The effects of delayed reduction of tonic inhibition on ischemic lesion and sensorimotor function. J Cereb Blood Flow Metab 2015; 35:1601-9. [PMID: 25966952 PMCID: PMC4640317 DOI: 10.1038/jcbfm.2015.86] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 03/25/2015] [Accepted: 03/30/2015] [Indexed: 02/02/2023]
Abstract
To aid in development of chronic stage treatments for sensorimotor deficits induced by ischemic stroke, we investigated the effects of GABA antagonism on brain structure and fine skilled reaching in a rat model of focal ischemia induced via cortical microinjections of endothelin-1 (ET-1). Beginning 7 days after stroke, animals were administered a gamma-aminobutyric acid (GABAA) inverse agonist, L-655,708, at a dose low enough to afford α5-GABAA receptor specificity. A week after stroke, the ischemic lesion comprised a small hypointense necrotic core (6±1 mm(3)) surrounded by a large (62±11 mm(3)) hyperintense perilesional region; the skilled reaching ability on the Montoya staircase test was decreased to 34%±2% of the animals' prestroke performance level. On L-655,708 treatment, animals showed a progressive decrease in total stroke volume (13±4 mm(3) per week), with no change in animals receiving placebo. Concomitantly, treated animals' skilled reaching progressively improved by 9%±1% per week, so that after 2 weeks of treatment, these animals performed at 65%±6% of their baseline ability, which was 25%±11% better than animals given placebo. These data indicate beneficial effects of delayed, sustained low-dose GABAA antagonism on neuroanatomic injury and skilled reaching in the chronic stage of stroke recovery in an ET-1 rat model of focal ischemia.
Collapse
Affiliation(s)
- Evelyn M R Lake
- Department of Medical Biophysics, University of Toronto, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Joydeep Chaudhuri
- Department of Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Lynsie Thomason
- Department of Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Rafal Janik
- Department of Medical Biophysics, University of Toronto, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
- Department of Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Milan Ganguly
- Department of Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Mary Brown
- Department of Biological Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - JoAnne McLaurin
- Department of Biological Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Hurvitz Brain Sciences Research, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Dale Corbett
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Department of Cellular & Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Heart and Stroke Foundation Canadian Partnership for Stroke Recovery, Ottawa, Ontario, Canada
| | - Greg J Stanisz
- Department of Medical Biophysics, University of Toronto, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
- Department of Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Department of Neurosurgery and Paediatric Neurosurgery, Medical University Lublin, Lublin, Poland
| | - Bojana Stefanovic
- Department of Medical Biophysics, University of Toronto, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
- Department of Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Heart and Stroke Foundation Canadian Partnership for Stroke Recovery, Ottawa, Ontario, Canada
- Department of Neurosurgery and Paediatric Neurosurgery, Medical University Lublin, Lublin, Poland
- Neuropsychopharmacology Research Group, Sunnybrook Research Institute, Toronto, Ontario, Canada
| |
Collapse
|
127
|
Lesion-induced and activity-dependent structural plasticity of Purkinje cell dendritic spines in cerebellar vermis and hemisphere. Brain Struct Funct 2015; 221:3405-26. [PMID: 26420278 DOI: 10.1007/s00429-015-1109-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 09/09/2015] [Indexed: 10/23/2022]
Abstract
Neuroplasticity allows the brain to encode experience and learn behaviors, and also to re-acquire lost functions after damage. The cerebellum is a suitable structure to address this topic because of its strong involvement in learning processes and compensation of lesion-induced deficits. This study was aimed to characterize the effects of a hemicerebellectomy (HCb) combined or not with the exposition to environmental enrichment (EE) on dendritic spine density and size in Purkinje cell proximal and distal compartments of cerebellar vermian and hemispherical regions. Male Wistar rats were housed in enriched or standard environments from the 21st post-natal day (pnd) onwards. At the 75th pnd, rats were submitted to HCb or sham lesion. Neurological symptoms and spatial performance in the Morris water maze were evaluated. At the end of testing, morphological analyses assessed dendritic spine density, area, length, and head diameter on vermian and hemispherical Purkinje cells. All hemicerebellectomized (HCbed) rats showed motor compensation, but standard-reared HCbed animals exhibited cognitive impairment that was almost completely compensated in enriched HCbed rats. The standard-reared HCbed rats showed decreased density with augmented size of Purkinje cell spines in the vermis, and augmented both density and size in the hemisphere. Enriched HCbed rats almost completely maintained the spine density and size induced by EE. Both lesion-induced and activity-dependent cerebellar plastic changes may be interpreted as "beneficial" brain reactions, aimed to support behavioral performance rescuing.
Collapse
|
128
|
Sherwood MS, Kane JH, Weisend MP, Parker JG. Enhanced control of dorsolateral prefrontal cortex neurophysiology with real-time functional magnetic resonance imaging (rt-fMRI) neurofeedback training and working memory practice. Neuroimage 2015; 124:214-223. [PMID: 26348555 DOI: 10.1016/j.neuroimage.2015.08.074] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 07/20/2015] [Accepted: 08/22/2015] [Indexed: 12/15/2022] Open
Abstract
Real-time functional magnetic resonance imaging (rt-fMRI) neurofeedback can be used to train localized, conscious regulation of blood oxygen level-dependent (BOLD) signals. As a therapeutic technique, rt-fMRI neurofeedback reduces the symptoms of a variety of neurologic disorders. To date, few studies have investigated the use of self-regulation training using rt-fMRI neurofeedback to enhance cognitive performance. This work investigates the utility of rt-fMRI neurofeedback as a tool to enhance human cognition by training healthy individuals to consciously control activity in the left dorsolateral prefrontal cortex (DLPFC). A cohort of 18 healthy participants in the experimental group underwent rt-fMRI neurofeedback from the left DLPFC in five training sessions across two weeks while 7 participants in the control group underwent similar training outside the MRI and without rt-fMRI neurofeedback. Working memory (WM) performance was evaluated on two testing days separated by the five rt-fMRI neurofeedback sessions using two computerized tests. We investigated the ability to control the BOLD signal across training sessions and WM performance across the two testing days. The group with rt-fMRI neurofeedback demonstrated a significant increase in the ability to self-regulate the BOLD signal in the left DLPFC across sessions. WM performance showed differential improvement between testing days one and two across the groups with the highest increases observed in the rt-fMRI neurofeedback group. These results provide evidence that individuals can quickly gain the ability to consciously control the left DLPFC, and this training results in improvements of WM performance beyond that of training alone.
Collapse
Affiliation(s)
- Matthew S Sherwood
- Wright State Research Institute, Wright State University, 4035 Colonel Glenn Hwy, Dayton, OH 45431, USA; Department of Biomedical, Industrial, and Human Factors Engineering, Wright State University, 3640 Colonel Glenn Hwy, Dayton, OH 45435, USA.
| | - Jessica H Kane
- Wright State Research Institute, Wright State University, 4035 Colonel Glenn Hwy, Dayton, OH 45431, USA; Department of Neuroscience, Cell Biology, and Physiology, Wright State University, 3640 Colonel Glenn Hwy, Dayton, OH 45435, USA
| | - Michael P Weisend
- Wright State Research Institute, Wright State University, 4035 Colonel Glenn Hwy, Dayton, OH 45431, USA
| | - Jason G Parker
- Wright State Research Institute, Wright State University, 4035 Colonel Glenn Hwy, Dayton, OH 45431, USA; Department of Biomedical, Industrial, and Human Factors Engineering, Wright State University, 3640 Colonel Glenn Hwy, Dayton, OH 45435, USA
| |
Collapse
|
129
|
Yamada N, Kakuda W, Yamamoto K, Momosaki R, Abo M. Atomoxetine administration combined with intensive speech therapy for post-stroke aphasia: evaluation by a novel SPECT method. Int J Neurosci 2015; 126:829-38. [DOI: 10.3109/00207454.2015.1074226] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
130
|
Alawieh A, Elvington A, Tomlinson S. Complement in the Homeostatic and Ischemic Brain. Front Immunol 2015; 6:417. [PMID: 26322048 PMCID: PMC4533015 DOI: 10.3389/fimmu.2015.00417] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 07/30/2015] [Indexed: 11/29/2022] Open
Abstract
The complement system is a component of the immune system involved in both recognition and response to pathogens, and it is implicated in an increasing number of homeostatic and disease processes. It is well documented that reperfusion of ischemic tissue results in complement activation and an inflammatory response that causes post-reperfusion injury. This occurs following cerebral ischemia and reperfusion and triggers secondary damage that extends beyond the initial infarcted area, an outcome that has rationalized the use of complement inhibitors as candidate therapeutics after stroke. In the central nervous system, however, recent studies have revealed that complement also has essential roles in synaptic pruning, neurogenesis, and neuronal migration. In the context of recovery after stroke, these apparent divergent functions of complement may account for findings that the protective effect of complement inhibition in the acute phase after stroke is not always maintained in the subacute and chronic phases. The development of effective stroke therapies based on modulation of the complement system will require a detailed understanding of complement-dependent processes in both early neurodegenerative events and delayed neuro-reparatory processes. Here, we review the role of complement in normal brain physiology, the events initiating complement activation after cerebral ischemia-reperfusion injury, and the contribution of complement to both injury and recovery. We also discuss how the design of future experiments may better characterize the dual role of complement in recovery after ischemic stroke.
Collapse
Affiliation(s)
- Ali Alawieh
- Neuroscience Institute, Department of Neurosciences, Medical University of South Carolina , Charleston, SC , USA
| | - Andrew Elvington
- Department of Pathology and Immunology, Washington University School of Medicine , St. Louis, MO , USA
| | - Stephen Tomlinson
- Department of Microbiology and Immunology, Ralph H. Johnson Veteran Affairs Medical Center, Medical University of South Carolina , Charleston, SC , USA
| |
Collapse
|
131
|
How the cortico-thalamic feedback affects the EEG power spectrum over frontal and occipital regions during propofol-induced sedation. J Comput Neurosci 2015; 39:155-79. [PMID: 26256583 DOI: 10.1007/s10827-015-0569-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 07/05/2015] [Accepted: 07/13/2015] [Indexed: 12/16/2022]
Abstract
Increasing concentrations of the anaesthetic agent propofol initially induces sedation before achieving full general anaesthesia. During this state of anaesthesia, the observed specific changes in electroencephalographic (EEG) rhythms comprise increased activity in the δ- (0.5-4 Hz) and α- (8-13 Hz) frequency bands over the frontal region, but increased δ- and decreased α-activity over the occipital region. It is known that the cortex, the thalamus, and the thalamo-cortical feedback loop contribute to some degree to the propofol-induced changes in the EEG power spectrum. However the precise role of each structure to the dynamics of the EEG is unknown. In this paper we apply a thalamo-cortical neuronal population model to reproduce the power spectrum changes in EEG during propofol-induced anaesthesia sedation. The model reproduces the power spectrum features observed experimentally both in frontal and occipital electrodes. Moreover, a detailed analysis of the model indicates the importance of multiple resting states in brain activity. The work suggests that the α-activity originates from the cortico-thalamic relay interaction, whereas the emergence of δ-activity results from the full cortico-reticular-relay-cortical feedback loop with a prominent enforced thalamic reticular-relay interaction. This model suggests an important role for synaptic GABAergic receptors at relay neurons and, more generally, for the thalamus in the generation of both the δ- and the α- EEG patterns that are seen during propofol anaesthesia sedation.
Collapse
|
132
|
|
133
|
Stinear CM, Petoe MA, Byblow WD. Primary Motor Cortex Excitability During Recovery After Stroke: Implications for Neuromodulation. Brain Stimul 2015. [PMID: 26195321 DOI: 10.1016/j.brs.2015.06.015] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Non-invasive brain stimulation techniques may be useful adjuvants to promote recovery after stroke. They are typically used to facilitate ipsilesional cortical excitability directly, or indirectly by suppressing contralesional cortical excitability and reducing interhemispheric inhibition from the contralesional to ipsilesional hemisphere. However, most of the evidence for this approach comes from studies of patients at the chronic stage of recovery. HYPOTHESIS We hypothesized that corticomotor excitability and interhemispheric inhibition would initially be asymmetric, with greater interhemispheric inhibition from contralesional to ipsilesional M1. We also hypothesized that balancing of corticomotor excitability and interhemispheric inhibition would be associated with greater improvements in paretic upper-limb impairment and function. METHODS We conducted a retrospective analysis of longitudinal data collected from 46 patients during the first six months after stroke. Transcranial magnetic stimulation was used to measure rest motor threshold, stimulus-response curves, and ipsilateral silent periods from the extensor carpi radialis muscles of both upper limbs. Analyses of variance and linear regression modeling were used to evaluate the effect of time on corticomotor excitability and interhemispheric inhibition in both hemispheres, and associations between these effects and improvements in paretic upper-limb impairment and function. RESULTS All participants had subcortical damage and only two had motor cortex involvement. As expected, ipsilesional corticomotor excitability was initially suppressed and increased over time, and this increase was associated with improved upper-limb impairment and function. However, interhemispheric inhibition was symmetrical and stable over time, and there was no evidence for a decrease in contralesional corticomotor excitability. CONCLUSIONS Neuromodulation interventions applied during spontaneous recovery may be more beneficial if they facilitate ipsilesional corticomotor excitability directly.
Collapse
Affiliation(s)
- Cathy M Stinear
- Department of Medicine, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand; Centre for Brain Research, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Matthew A Petoe
- Department of Medicine, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand; Centre for Brain Research, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Winston D Byblow
- Centre for Brain Research, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand; Department of Sport & Exercise Science, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.
| |
Collapse
|
134
|
Abstract
PURPOSE OF REVIEW This review presents recent developments in the prediction of motor recovery after stroke; explores whether rehabilitation interventions delivered during the spontaneous recovery process can improve outcomes; and identifies the first trials to focus on the rate rather than extent of motor recovery (Supplementary Digital Content 1). RECENT FINDINGS Two recent studies have attempted to accelerate the rate of motor recovery during the first few weeks after stroke, with neuromodulation techniques designed to facilitate excitability of the ipsilesional motor cortex. One trial using transcranial direct current stimulation was negative, and the other trial using bilateral priming was positive. These contrasting results may be explained by important differences in trial design. This new focus on modifying rate, rather than extent, of motor recovery is in line with accumulating evidence that the motor recovery plateau is largely determined by the extent of damage to descending motor pathways, which is currently untreatable. SUMMARY Interventions that facilitate neural plasticity and reorganization may accelerate recovery of motor function during the spontaneous recovery period, without affecting final outcome. This may represent a useful new approach for future trials conducted during rehabilitation at the subacute stage of stroke.
Collapse
|
135
|
Fagerholm ED, Hellyer PJ, Scott G, Leech R, Sharp DJ. Disconnection of network hubs and cognitive impairment after traumatic brain injury. Brain 2015; 138:1696-709. [PMID: 25808370 PMCID: PMC4614120 DOI: 10.1093/brain/awv075] [Citation(s) in RCA: 134] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 01/21/2015] [Indexed: 01/06/2023] Open
Abstract
Traumatic brain injury (TBI) affects brain connectivity, which disrupts the large-scale networks that support cognitive function. Fagerholm et al. use graph analysis, with brain regions as nodes and white matter tracts as connections, to show that TBI particularly disrupts the connectivity of hubs, and that this disconnection predicts cognitive impairment. Traumatic brain injury affects brain connectivity by producing traumatic axonal injury. This disrupts the function of large-scale networks that support cognition. The best way to describe this relationship is unclear, but one elegant approach is to view networks as graphs. Brain regions become nodes in the graph, and white matter tracts the connections. The overall effect of an injury can then be estimated by calculating graph metrics of network structure and function. Here we test which graph metrics best predict the presence of traumatic axonal injury, as well as which are most highly associated with cognitive impairment. A comprehensive range of graph metrics was calculated from structural connectivity measures for 52 patients with traumatic brain injury, 21 of whom had microbleed evidence of traumatic axonal injury, and 25 age-matched controls. White matter connections between 165 grey matter brain regions were defined using tractography, and structural connectivity matrices calculated from skeletonized diffusion tensor imaging data. This technique estimates injury at the centre of tract, but is insensitive to damage at tract edges. Graph metrics were calculated from the resulting connectivity matrices and machine-learning techniques used to select the metrics that best predicted the presence of traumatic brain injury. In addition, we used regularization and variable selection via the elastic net to predict patient behaviour on tests of information processing speed, executive function and associative memory. Support vector machines trained with graph metrics of white matter connectivity matrices from the microbleed group were able to identify patients with a history of traumatic brain injury with 93.4% accuracy, a result robust to different ways of sampling the data. Graph metrics were significantly associated with cognitive performance: information processing speed (R2 = 0.64), executive function (R2 = 0.56) and associative memory (R2 = 0.25). These results were then replicated in a separate group of patients without microbleeds. The most influential graph metrics were betweenness centrality and eigenvector centrality, which provide measures of the extent to which a given brain region connects other regions in the network. Reductions in betweenness centrality and eigenvector centrality were particularly evident within hub regions including the cingulate cortex and caudate. Our results demonstrate that betweenness centrality and eigenvector centrality are reduced within network hubs, due to the impact of traumatic axonal injury on network connections. The dominance of betweenness centrality and eigenvector centrality suggests that cognitive impairment after traumatic brain injury results from the disconnection of network hubs by traumatic axonal injury.
Collapse
Affiliation(s)
- Erik D Fagerholm
- Computational, Cognitive, and Clinical Neuroimaging Laboratory, Division of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Peter J Hellyer
- Computational, Cognitive, and Clinical Neuroimaging Laboratory, Division of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Gregory Scott
- Computational, Cognitive, and Clinical Neuroimaging Laboratory, Division of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Robert Leech
- Computational, Cognitive, and Clinical Neuroimaging Laboratory, Division of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - David J Sharp
- Computational, Cognitive, and Clinical Neuroimaging Laboratory, Division of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, London, UK
| |
Collapse
|
136
|
In vivo analysis of neuroinflammation in the late chronic phase after experimental stroke. Neuroscience 2015; 292:71-80. [PMID: 25701708 DOI: 10.1016/j.neuroscience.2015.02.024] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2014] [Revised: 02/09/2015] [Accepted: 02/11/2015] [Indexed: 01/28/2023]
Abstract
BACKGROUND AND PURPOSE In vivo imaging of inflammatory processes is a valuable tool in stroke research. We here investigated the combination of two imaging modalities in the chronic phase after cerebral ischemia: magnetic resonance imaging (MRI) using intravenously applied ultra small supraparamagnetic iron oxide particles (USPIO), and positron emission tomography (PET) with the tracer [(11)C]PK11195. METHODS Rats were subjected to permanent middle cerebral artery occlusion (pMCAO) by the macrosphere model and monitored by MRI and PET for 28 or 56 days, followed by immunohistochemical endpoint analysis. To our knowledge, this is the first study providing USPIO-MRI data in the chronic phase up to 8 weeks after stroke. RESULTS Phagocytes with internalized USPIOs induced MRI-T2(∗) signal alterations in the brain. Combined analysis with [(11)C]PK11195-PET allowed quantification of phagocytic activity and other neuroinflammatory processes. From 4 weeks after induction of ischemia, inflammation was dominated by phagocytes. Immunohistochemistry revealed colocalization of Iba1+ microglia with [(11)C]PK11195 and ED1/CD68 with USPIOs. USPIO-related iron was distinguished from alternatively deposited iron by assessing MRI before and after USPIO application. Tissue affected by non-phagocytic inflammation during the first week mostly remained in a viably vital but remodeled state after 4 or 8 weeks, while phagocytic activity was associated with severe injury and necrosis accordingly. CONCLUSIONS We conclude that the combined approach of USPIO-MRI and [(11)C]PK11195-PET allows to observe post-stroke inflammatory processes in the living animal in an intraindividual and longitudinal fashion, predicting long-term tissue fate. The non-invasive imaging methods do not affect the immune system and have been applied to human subjects before. Translation into clinical applications is therefore feasible.
Collapse
|
137
|
Ishida A, Misumi S, Ueda Y, Shimizu Y, Cha-Gyun J, Tamakoshi K, Ishida K, Hida H. Early constraint-induced movement therapy promotes functional recovery and neuronal plasticity in a subcortical hemorrhage model rat. Behav Brain Res 2015; 284:158-66. [PMID: 25700666 DOI: 10.1016/j.bbr.2015.02.022] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Revised: 02/04/2015] [Accepted: 02/10/2015] [Indexed: 01/15/2023]
Abstract
Constraint-induced movement therapy (CIMT) promotes functional recovery of impaired forelimbs after hemiplegic strokes, including intracerebral hemorrhage (ICH). We used a rat model of subcortical hemorrhage to compare the effects of delivering early or late CIMT after ICH. The rat model was made by injecting collagenase into the globus pallidus near the internal capsule, and then forcing rats to use the affected forelimb for 7 days starting either 1 day (early CIMT) or 17 days (late CIMT) after the lesion. Recovery of forelimb function in the skilled reaching test and the ladder stepping test was found after early-CIMT, while no significant recovery was shown after late CIMT or in the non-CIMT controls. Early CIMT was associated with greater numbers of ΔFosB-positive cells in the ipsi-lesional sensorimotor cortex layers II-III and V. Additionally, we found expression of the growth-related genes brain-derived neurotrophic factor (BDNF) and growth-related protein 43 (GAP-43), and abundant dendritic arborization of pyramidal neurons in the sensorimotor area. Similar results were not detected in the contra-lesional cortex. In contrast to early CIMT, late CIMT failed to induce any changes in plasticity. We conclude that CIMT induces molecular and morphological plasticity in the ipsi-lesional sensorimotor cortex and facilitates better functional recovery when initiated immediately after hemorrhage.
Collapse
Affiliation(s)
- Akimasa Ishida
- Department of Neurophysiology and Brain Science, Nagoya City University Graduate School of Medicine, Nagoya, Aichi 467-8601, Japan; Department of Physical Therapy, Program in Physical and Occupational Therapy, Nagoya University Graduate School of Medicine, Nagoya, Aichi 461-8673, Japan
| | - Sachiyo Misumi
- Department of Neurophysiology and Brain Science, Nagoya City University Graduate School of Medicine, Nagoya, Aichi 467-8601, Japan
| | - Yoshitomo Ueda
- Department of Neurophysiology and Brain Science, Nagoya City University Graduate School of Medicine, Nagoya, Aichi 467-8601, Japan
| | - Yuko Shimizu
- Department of Neurophysiology and Brain Science, Nagoya City University Graduate School of Medicine, Nagoya, Aichi 467-8601, Japan
| | - Jung Cha-Gyun
- Department of Neurophysiology and Brain Science, Nagoya City University Graduate School of Medicine, Nagoya, Aichi 467-8601, Japan
| | - Keigo Tamakoshi
- Department of Physical Therapy, Program in Physical and Occupational Therapy, Nagoya University Graduate School of Medicine, Nagoya, Aichi 461-8673, Japan
| | - Kazuto Ishida
- Department of Physical Therapy, Program in Physical and Occupational Therapy, Nagoya University Graduate School of Medicine, Nagoya, Aichi 461-8673, Japan
| | - Hideki Hida
- Department of Neurophysiology and Brain Science, Nagoya City University Graduate School of Medicine, Nagoya, Aichi 467-8601, Japan.
| |
Collapse
|
138
|
Brizuela M, Blizzard CA, Chuckowree JA, Dawkins E, Gasperini RJ, Young KM, Dickson TC. The microtubule-stabilizing drug Epothilone D increases axonal sprouting following transection injury in vitro. Mol Cell Neurosci 2015; 66:129-40. [PMID: 25684676 DOI: 10.1016/j.mcn.2015.02.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Revised: 02/03/2015] [Accepted: 02/06/2015] [Indexed: 11/26/2022] Open
Abstract
Neuronal cytoskeletal alterations, in particular the loss and misalignment of microtubules, are considered a hallmark feature of the degeneration that occurs after traumatic brain injury (TBI). Therefore, microtubule-stabilizing drugs are attractive potential therapeutics for use following TBI. The best-known drug in this category is Paclitaxel, a widely used anti-cancer drug that has produced promising outcomes when employed in the treatment of various animal models of nervous system trauma. However, Paclitaxel is not ideal for the treatment of patients with TBI due to its limited blood-brain barrier (BBB) permeability. Herein we have characterized the effect of the brain penetrant microtubule-stabilizing agent Epothilone D (Epo D) on post-injury axonal sprouting in an in vitro model of CNS trauma. Epo D was found to modulate axonal sprout number in a dose dependent manner, increasing the number of axonal sprouts generated post-injury. Elevated sprouting was observed when analyzing the total population of injured neurons, as well as in selective analysis of Thy1-YFP-labeled excitatory neurons. However, we found no effect of Epo D on axonal sprout length or outgrowth speed. These findings indicate that Epo D specifically affects injury-induced axonal sprout generation, but not net growth. Our investigation demonstrates that primary cultures of cortical neurons are tolerant of Epo D exposure, and that Epo D significantly increases their regenerative response following structural injury. Therefore Epo D may be a potent therapeutic for enhancing regeneration following CNS injury. This article is part of a Special Issue entitled 'Traumatic Brain Injury'.
Collapse
Affiliation(s)
- Mariana Brizuela
- Menzies Institute for Medical Research Tasmania, University of Tasmania, Hobart, Tasmania 7000, Australia
| | - Catherine A Blizzard
- Menzies Institute for Medical Research Tasmania, University of Tasmania, Hobart, Tasmania 7000, Australia
| | - Jyoti A Chuckowree
- Menzies Institute for Medical Research Tasmania, University of Tasmania, Hobart, Tasmania 7000, Australia
| | - Edgar Dawkins
- Menzies Institute for Medical Research Tasmania, University of Tasmania, Hobart, Tasmania 7000, Australia
| | - Robert J Gasperini
- Menzies Institute for Medical Research Tasmania, University of Tasmania, Hobart, Tasmania 7000, Australia
| | - Kaylene M Young
- Menzies Institute for Medical Research Tasmania, University of Tasmania, Hobart, Tasmania 7000, Australia
| | - Tracey C Dickson
- Menzies Institute for Medical Research Tasmania, University of Tasmania, Hobart, Tasmania 7000, Australia.
| |
Collapse
|
139
|
Zheng Y, Zhong D, Chen H, Ma S, Sun Y, Wang M, Liu Q, Li G. Pivotal role of cerebral interleukin-23 during immunologic injury in delayed cerebral ischemia in mice. Neuroscience 2015; 290:321-31. [PMID: 25637493 DOI: 10.1016/j.neuroscience.2015.01.041] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 12/18/2014] [Accepted: 01/06/2015] [Indexed: 10/24/2022]
Abstract
BACKGROUND Interleukin-23 (IL-23) is required for T helper 17 (Th17) cell responses and IL-17 production in ischemic stroke. We previously showed that the IL-23/IL-17 axis aggravates immune injury after cerebral infarction in mice. However, IL-23 might activate other cytokines and transcription factor forkhead box P3 (Foxp3) production in cerebral ischemia. We aimed to determine whether IL-23p19 knockdown prevents cerebral ischemic injury by reducing ischemic-induced inflammation. METHODS Ischemic stroke models were established by permanent middle cerebral arterial occlusion (pMCAO) in male C57BL/6 mice. In vivo gene knockdown was achieved by intravenous delivery of lentiviral vectors (LVs) encoding IL-23p19 short hairpin RNA (LV-IL-23p19 shRNA). Enzyme-linked immunoassay (ELISA) and quantitative real-time polymerase chain reaction (qRT-PCR) confirmed inhibitory efficiency. Behavioral deficits were evaluated by adhesive-removal somatic-sensory test. Brain infarct volume was measured at day 5 after pMCAO by 2,3,5-triphenyltetrazolium chloride (TTC) staining. Expression of IL-17, IL-4, interferon (IFN)-γ and Foxp3 in ischemic brain tissues were detected by qRT-PCR and Western blotting, respectively. Additionally, immunohistochemical staining located cytokines in ischemic brain tissues. RESULTS RNA interference knockdown of IL-23p19 resulted in improved neurological function and reduced infarct volume. IL-23p19 knockdown suppressed IL-17 gene and protein expression. Moreover, IL-23p19 deficiency enhanced IFN-γ and Foxp3 expressions in delayed cerebral ischemic mice, and did not impact IL-4 expression. Immunohistochemical staining showed that IL-17, IL-4, IFN-γ and Foxp3-positive cells were located around ischemic lesions of the ipsilateral hemisphere. CONCLUSIONS IL-23p19 knockdown prevents delayed cerebral ischemic injury by dampening the ischemia-induced inflammation, and is a promising approach for clinically managing ischemic stroke.
Collapse
Affiliation(s)
- Y Zheng
- Department of Neurology, The First Affiliated Hospital, Harbin Medical University, 23 You Zheng Street, Harbin 150001, Heilong Jiang Province, PR China
| | - D Zhong
- Department of Neurology, The First Affiliated Hospital, Harbin Medical University, 23 You Zheng Street, Harbin 150001, Heilong Jiang Province, PR China
| | - H Chen
- Department of Neurology, The First Affiliated Hospital, Harbin Medical University, 23 You Zheng Street, Harbin 150001, Heilong Jiang Province, PR China
| | - S Ma
- Department of Neurology, The First Affiliated Hospital, Harbin Medical University, 23 You Zheng Street, Harbin 150001, Heilong Jiang Province, PR China
| | - Y Sun
- Department of Neurology, The First Affiliated Hospital, Harbin Medical University, 23 You Zheng Street, Harbin 150001, Heilong Jiang Province, PR China
| | - M Wang
- Department of Neurology, The First Affiliated Hospital, Harbin Medical University, 23 You Zheng Street, Harbin 150001, Heilong Jiang Province, PR China
| | - Q Liu
- Department of Neurology, The First Affiliated Hospital, Harbin Medical University, 23 You Zheng Street, Harbin 150001, Heilong Jiang Province, PR China
| | - G Li
- Department of Neurology, The First Affiliated Hospital, Harbin Medical University, 23 You Zheng Street, Harbin 150001, Heilong Jiang Province, PR China.
| |
Collapse
|
140
|
Kim J, Park SW, Lee Y, Seo H. Clinical Outcomes of Robot-assisted Arm Rehabilitation in Stroke Patients. BRAIN & NEUROREHABILITATION 2015. [DOI: 10.12786/bn.2015.8.1.46] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Affiliation(s)
- Jungsoo Kim
- Myongji Choonhey Rehabilitation Hospital, Korea
| | | | | | - Hyojin Seo
- Myongji Choonhey Rehabilitation Hospital, Korea
| |
Collapse
|
141
|
Ruscher K, Wieloch T. The involvement of the sigma-1 receptor in neurodegeneration and neurorestoration. J Pharmacol Sci 2015; 127:30-5. [PMID: 25704015 DOI: 10.1016/j.jphs.2014.11.011] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Revised: 11/19/2014] [Accepted: 11/27/2014] [Indexed: 02/07/2023] Open
Abstract
The sigma-1 receptor (Sig-1R) is a single 25 kD polypeptide and a chaperone protein immersed in lipid rafts of the endoplasmic reticulum (ER) where it interacts with mitochondria at the mitochondria-associated ER membrane domain (MAM). Upon activation, the Sig-1R binds to the inositol trisphosphate receptor (IP3R), and modulates cellular calcium (Ca(2+)) homeostasis. Also, the activated Sig-1R modulates plasma membrane receptor and ion channel functions, and may regulate cellular excitability. Further, the Sig-1R promotes trafficking of lipids and proteins essential for neurotransmission, cell growth and motility. Activation of the Sig-1R provides neuroprotection and is neurorestorative in cellular and animal models of neurodegenerative diseases and brain ischaemia. Neuroprotection appears to be due to inhibition of cellular Ca(2+) toxicity and/or inflammation, and neurorestoration may include balancing abberant neurotransmission or stimulation of synaptogenesis, thus remodelling brain connectivity. Single nucleotide polymorphisms and mutations of the SIGMAR1 gene worsen outcome in Alzheimer's disease and myotrophic lateral sclerosis supporting a role of Sig-1R in neurodegenerative disease. The combined neuroprotective and neurorestorative actions of the Sig-1R, provide a broad therapeutic time window of Sig-1R agonists. The Sig-1R is therefore a strong therapeutic target for the development of new treatments for neurodegenerative diseases and stroke.
Collapse
Affiliation(s)
- Karsten Ruscher
- Laboratory for Experimental Brain Research, Division of Neurosurgery, Department of Clinical Sciences, Lund University, BMC A13, S-22184 Lund, Sweden
| | - Tadeusz Wieloch
- Laboratory for Experimental Brain Research, Division of Neurosurgery, Department of Clinical Sciences, Lund University, BMC A13, S-22184 Lund, Sweden.
| |
Collapse
|
142
|
Li S, Zaninotto AL, Neville IS, Paiva WS, Nunn D, Fregni F. Clinical utility of brain stimulation modalities following traumatic brain injury: current evidence. Neuropsychiatr Dis Treat 2015; 11:1573-86. [PMID: 26170670 PMCID: PMC4494620 DOI: 10.2147/ndt.s65816] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Traumatic brain injury (TBI) remains the main cause of disability and a major public health problem worldwide. This review focuses on the neurophysiology of TBI, and the rationale and current state of evidence of clinical application of brain stimulation to promote TBI recovery, particularly on consciousness, cognitive function, motor impairments, and psychiatric conditions. We discuss the mechanisms of different brain stimulation techniques including major noninvasive and invasive stimulations. Thus far, most noninvasive brain stimulation interventions have been nontargeted and focused on the chronic phase of recovery after TBI. In the acute stages, there is limited available evidence of the efficacy and safety of brain stimulation to improve functional outcomes. Comparing the studies across different techniques, transcranial direct current stimulation is the intervention that currently has the higher number of properly designed clinical trials, though total number is still small. We recognize the need for larger studies with target neuroplasticity modulation to fully explore the benefits of brain stimulation to effect TBI recovery during different stages of recovery.
Collapse
Affiliation(s)
- Shasha Li
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China ; Spaulding Neuromodulation Center, Harvard Medical School, Boston, MA, USA
| | - Ana Luiza Zaninotto
- Spaulding Neuromodulation Center, Harvard Medical School, Boston, MA, USA ; Division of Psychology, Hospital das Clínicas, University of São Paulo, São Paulo, Brazil
| | - Iuri Santana Neville
- Division of Neurosurgery, University of São Paulo Medical School, São Paulo, São Paulo, Brazil
| | - Wellingson Silva Paiva
- Division of Neurosurgery, University of São Paulo Medical School, São Paulo, São Paulo, Brazil
| | - Danuza Nunn
- Spaulding Neuromodulation Center, Harvard Medical School, Boston, MA, USA
| | - Felipe Fregni
- Spaulding Neuromodulation Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
143
|
Abstract
Brain injury continues to be one of the leading causes of disability worldwide. Despite decades of research, there is currently no pharmacologically effective treatment for preventing neuronal loss and repairing the brain. As a result, novel therapeutic approaches, such as cell-based therapies, are being actively pursued to repair tissue damage and restore neurological function after injury. In this study, we examined the neuroprotective potential of amniotic fluid (AF) single cell clones, engineered to secrete glial cell derived neurotrophic factor (AF-GDNF), both in vitro and in a surgically induced model of brain injury. Our results show that pre-treatment with GDNF significantly increases cell survival in cultures of AF cells or cortical neurons exposed to hydrogen peroxide. Since improving the efficacy of cell transplantation depends on enhanced graft cell survival, we investigated whether AF-GDNF cells seeded on polyglycolic acid (PGA) scaffolds could enhance graft survival following implantation into the lesion cavity. Encouragingly, the AF-GDNF cells survived longer than control AF cells in serum-free conditions and continued to secrete GDNF both in vitro and following implantation into the injured motor cortex. AF-GDNF implantation in the acute period following injury was sufficient to activate the MAPK/ERK signaling pathway in host neural cells in the peri-lesion area, potentially boosting endogenous neuroprotective pathways. These results were complemented with promising trends in beam walk tasks in AF-GDNF/PGA animals during the 7 day timeframe. Further investigation is required to determine whether significant behavioural improvement can be achieved at a longer timeframe.
Collapse
|
144
|
Estradiol and Progesterone Administration After pMCAO Stimulates the Neurological Recovery and Reduces the Detrimental Effect of Ischemia Mainly in Hippocampus. Mol Neurobiol 2014; 52:1690-1703. [DOI: 10.1007/s12035-014-8963-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 10/23/2014] [Indexed: 10/24/2022]
|
145
|
Lövkvist H, Jönsson AC, Luthman H, Jood K, Jern C, Wieloch T, Lindgren A. Variations in apolipoprotein D and sigma non-opioid intracellular receptor 1 genes with relation to risk, severity and outcome of ischemic stroke. BMC Neurol 2014; 14:191. [PMID: 25261976 PMCID: PMC4186220 DOI: 10.1186/s12883-014-0191-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 09/23/2014] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND In experimental studies, the apolipoprotein D (APOD) and the sigma receptor type 1 (SIGMAR1) have been related to processes of brain damage, repair and plasticity. METHODS We examined blood samples from 3081 ischemic stroke (IS) patients and 1595 control subjects regarding 10 single nucleotide polymorphisms (SNPs) in the APOD (chromosomal location 3q29) and SIGMAR1 (chromosomal location 9p13) genes to find possible associations with IS risk, IS severity (NIHSS-score) and recovery after IS (modified Rankin Scale, mRS, at 90 days). Simple/multiple logistic regression and Spearman's rho were utilized for the analyses. RESULTS Among the SNPs analyzed, rs7659 within the APOD gene showed a possible association with stroke risk (OR = 1.12; 95% CI: 1.01-1.25; P = 0.029) and stroke severity (NIHSS ≥ 16) (OR = 0.70; 95% CI: 0.54-0.92; P = 0.009) when controlling for age, sex and vascular risk factors for stroke. No SNP showed an association with stroke recovery (mRS). CONCLUSIONS We conclude that the SNP rs7659 within the APOD gene might be related to risk and severity of ischemic stroke in patients.
Collapse
Affiliation(s)
- Håkan Lövkvist
- />Department of Clinical Sciences Lund, Neurology, Lund University, Lund, Sweden
- />Department of Neurology and Rehabilitation Medicine, Neurology, Skåne University Hospital, Lund, Sweden
- />R&D Centre Skåne, Skåne University Hospital, SE-221 85 Lund, Sweden
| | | | - Holger Luthman
- />Department of Clinical Sciences Malmö, Medical Genetics, Lund University, Malmö, Sweden
| | - Katarina Jood
- />Department of Clinical Neuroscience and Rehabilitation, The Sahlgrenska Academy at University of Gothenburg, Institute of Neuroscience and Physiology, Gothenburg, Sweden
| | - Christina Jern
- />Department of Clinical Neuroscience and Rehabilitation, The Sahlgrenska Academy at University of Gothenburg, Institute of Neuroscience and Physiology, Gothenburg, Sweden
| | - Tadeusz Wieloch
- />Department of Neurosurgery, Laboratory for Experimental Brain Research, Lund University, Lund, Sweden
| | - Arne Lindgren
- />Department of Clinical Sciences Lund, Neurology, Lund University, Lund, Sweden
- />Department of Neurology and Rehabilitation Medicine, Neurology, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
146
|
Wang Y, Yue X, Kiesewetter DO, Wang Z, Lu J, Niu G, Teng G, Chen X. [(18)F]DPA-714 PET imaging of AMD3100 treatment in a mouse model of stroke. Mol Pharm 2014; 11:3463-70. [PMID: 25157648 PMCID: PMC4186675 DOI: 10.1021/mp500234d] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
Chemokine
receptor 4 and stromal-cell-derived factor 1 have been
found to be related to the initiation of neuroinflammation in ischemic
brain. Herein, we aimed to monitor the changes of neuorinflammation
after AMD3100 treatment using a translocator protein (TSPO) specific
PET tracer in a mouse model of stroke. The transient MCAO model was
established with Balb/C mice. The success of the model was confirmed
by magnetic resonance imaging and FDG PET. The treatment started the
same day after surgery via daily intraperitoneal injection of 1 mg
of AMD3100/kg for three consecutive days. [18F]DPA-714
was used as the TSPO imaging tracer. In vivo PET
was performed at different time points after surgery in both control
and treated mice. Ex vivo histological and immunofluorescence
staining of brain slices was performed to confirm the lesion site
and inflammatory cell activation. The TSPO level was also evaluated
using Western blotting. Longitudinal PET scans revealed that the level
of [18F]DPA-714 uptake was significantly increased in the
ischemic brain area with a peak accumulation at around day 10 after
surgery, and the level of uptake remained high until day 16. The in vivo PET data were consistent with those from ex vivo immunofluorescence staining. After AMD3100 treatment,
the signal intensity was significantly decreased compared with that
of normal saline-treated control group. In conclusion, TSPO-targeted
PET imaging using [18F]DPA-714 can be used to monitor inflammatory
response after stroke and provide a useful method for evaluating the
efficacy of anti-inflammation treatment.
Collapse
Affiliation(s)
- Yu Wang
- Jiangsu Key Laboratory of Molecular Imaging and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University , Nanjing 210009, China
| | | | | | | | | | | | | | | |
Collapse
|
147
|
Lipsanen A, Parkkinen S, Khabbal J, Mäkinen P, Peräniemi S, Hiltunen M, Jolkkonen J. KB-R7943, an inhibitor of the reverse Na+/Ca2+ exchanger, does not modify secondary pathology in the thalamus following focal cerebral stroke in rats. Neurosci Lett 2014; 580:173-7. [DOI: 10.1016/j.neulet.2014.08.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 06/17/2014] [Accepted: 08/04/2014] [Indexed: 11/29/2022]
|
148
|
Skop NB, Calderon F, Cho CH, Gandhi CD, Levison SW. Improvements in biomaterial matrices for neural precursor cell transplantation. MOLECULAR AND CELLULAR THERAPIES 2014; 2:19. [PMID: 26056586 PMCID: PMC4452047 DOI: 10.1186/2052-8426-2-19] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 06/05/2014] [Indexed: 12/24/2022]
Abstract
Progress is being made in developing neuroprotective strategies for traumatic brain injuries; however, there will never be a therapy that will fully preserve neurons that are injured from moderate to severe head injuries. Therefore, to restore neurological function, regenerative strategies will be required. Given the limited regenerative capacity of the resident neural precursors of the CNS, many investigators have evaluated the regenerative potential of transplanted precursors. Unfortunately, these precursors do not thrive when engrafted without a biomaterial scaffold. In this article we review the types of natural and synthetic materials that are being used in brain tissue engineering applications for traumatic brain injury and stroke. We also analyze modifications of the scaffolds including immobilizing drugs, growth factors and extracellular matrix molecules to improve CNS regeneration and functional recovery. We conclude with a discussion of some of the challenges that remain to be solved towards repairing and regenerating the brain.
Collapse
Affiliation(s)
- Nolan B Skop
- Department of Neurology & Neurosciences, Rutgers University-New Jersey Medical School, NJMS-Cancer Center, H-1226, 205 South Orange Ave., Newark, NJ 07103 USA ; Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ 07102 USA
| | - Frances Calderon
- Department of Neurology & Neurosciences, Rutgers University-New Jersey Medical School, NJMS-Cancer Center, H-1226, 205 South Orange Ave., Newark, NJ 07103 USA
| | - Cheul H Cho
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ 07102 USA
| | - Chirag D Gandhi
- Department of Neurology & Neurosciences, Rutgers University-New Jersey Medical School, NJMS-Cancer Center, H-1226, 205 South Orange Ave., Newark, NJ 07103 USA ; Department of Neurological Surgery, Rutgers University-New Jersey Medical School, New Jersey Medical School, Newark, NJ 07103 USA
| | - Steven W Levison
- Department of Neurology & Neurosciences, Rutgers University-New Jersey Medical School, NJMS-Cancer Center, H-1226, 205 South Orange Ave., Newark, NJ 07103 USA
| |
Collapse
|
149
|
Allred RP, Kim SY, Jones TA. Use it and/or lose it-experience effects on brain remodeling across time after stroke. Front Hum Neurosci 2014; 8:379. [PMID: 25018715 PMCID: PMC4072969 DOI: 10.3389/fnhum.2014.00379] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Accepted: 05/14/2014] [Indexed: 01/29/2023] Open
Abstract
The process of brain remodeling after stroke is time- and neural activity-dependent, and the latter makes it inherently sensitive to behavioral experiences. This generally supports targeting early dynamic periods of post-stroke neural remodeling with rehabilitative training (RT). However, the specific neural events that optimize RT effects are unclear and, as such, cannot be precisely targeted. Here we review evidence for, potential mechanisms of, and ongoing knowledge gaps surrounding time-sensitivities in RT efficacy, with a focus on findings from animal models of upper extremity RT. The reorganization of neural connectivity after stroke is a complex multiphasic process interacting with glial and vascular changes. Behavioral manipulations can impact numerous elements of this process to affect function. RT efficacy varies both with onset time and its timing relative to the development of compensatory strategies with the less-affected (nonparetic) hand. Earlier RT may not only capitalize on a dynamic period of brain remodeling but also counter a tendency for compensatory strategies to stamp-in suboptimal reorganization patterns. However, there is considerable variability across injuries and individuals in brain remodeling responses, and some early behavioral manipulations worsen function. The optimal timing of RT may remain unpredictable without clarification of the cellular events underlying time-sensitivities in its effects.
Collapse
Affiliation(s)
- Rachel P Allred
- Department of Psychology and Institute for Neuroscience, University of Texas at Austin Austin, TX, USA
| | - Soo Young Kim
- Department of Integrative Biology, University of California Berkeley Berkeley, CA, USA
| | - Theresa A Jones
- Department of Psychology and Institute for Neuroscience, University of Texas at Austin Austin, TX, USA
| |
Collapse
|
150
|
Guo X, Jin Z, Feng X, Tong S. Enhanced effective connectivity in mild occipital stroke patients with hemianopia. IEEE Trans Neural Syst Rehabil Eng 2014; 22:1210-7. [PMID: 24876132 DOI: 10.1109/tnsre.2014.2325601] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Plasticity-based spontaneous recovery and rehabilitation intervention of stroke-induced hemianopia have drawn great attention in recent years. However, the underlying neural mechanism remains unknown. This study aims to investigate brain network disruption and reorganization in hemianopia patients due to mild occipital stroke. Resting-state networks were constructed from 12 hemianopia patients with right occipital infarct by partial directed coherence analysis of multi-channel electroencephalograms. Compared with control subjects, the patients presented enhanced connectivity owing to newly formed connections. Compensational connections mostly originated from the peri-infarct area and targeted contralesional frontal, central, and parietal cortices. These new ipsilesional-to-contralesional inter-hemispheric connections coordinately presented significant correlation with the extent of vision loss. The enhancement of connectivity might be the neural substrate for brain plasticity in stroke-induced hemianopia and may shed light on plasticity-based recovery or rehabilitation.
Collapse
|