Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Brain ; 139(Pt 2): 468-80, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26685158

ABSTRACT

Ischaemic stroke is the leading cause of severe long-term disability yet lacks drug therapies that promote the repair phase of recovery. This repair phase of stroke occurs days to months after stroke onset and involves brain remapping and plasticity within the peri-infarct zone. Elucidating mechanisms that promote this plasticity is critical for the development of new therapeutics with a broad treatment window. Inhibiting tonic (extrasynaptic) GABA signalling during the repair phase was reported to enhance functional recovery in mice suggesting that GABA plays an important function in modulating brain repair. While tonic GABA appears to suppress brain repair after stroke, less is known about the role of phasic (synaptic) GABA during the repair phase. We observed an increase in postsynaptic phasic GABA signalling in mice within the peri-infarct cortex specific to layer 5; we found increased numbers of α1 receptor subunit-containing GABAergic synapses detected using array tomography, and an associated increased efficacy of spontaneous and miniature inhibitory postsynaptic currents in pyramidal neurons. Furthermore, we demonstrate that enhancing phasic GABA signalling using zolpidem, a Food and Drug Administration (FDA)-approved GABA-positive allosteric modulator, during the repair phase improved behavioural recovery. These data identify potentiation of phasic GABA signalling as a novel therapeutic strategy, indicate zolpidem's potential to improve recovery, and underscore the necessity to distinguish the role of tonic and phasic GABA signalling in stroke recovery.


Subject(s)
Drug Delivery Systems , GABA-A Receptor Agonists/administration & dosage , Neural Inhibition/physiology , Pyridines/administration & dosage , Receptors, GABA-A/physiology , Stroke/drug therapy , Animals , Drug Delivery Systems/trends , Male , Mice , Mice, Inbred C57BL , Neocortex/drug effects , Neocortex/physiology , Neural Inhibition/drug effects , Organ Culture Techniques , Recovery of Function/drug effects , Recovery of Function/physiology , Stroke/pathology , Stroke/physiopathology , Zolpidem
2.
Am J Pathol ; 184(9): 2493-504, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25134760

ABSTRACT

Stroke is the leading cause of adult disability and the fourth most common cause of death in the United States. Inflammation is thought to play an important role in stroke pathology, but the factors that promote inflammation in this setting remain to be fully defined. An understudied but important factor is the role of meningeal-located immune cells in modulating brain pathology. Although different immune cells traffic through meningeal vessels en route to the brain, mature mast cells do not circulate but are resident in the meninges. With the use of genetic and cell transfer approaches in mice, we identified evidence that meningeal mast cells can importantly contribute to the key features of stroke pathology, including infiltration of granulocytes and activated macrophages, brain swelling, and infarct size. We also obtained evidence that two mast cell-derived products, interleukin-6 and, to a lesser extent, chemokine (C-C motif) ligand 7, can contribute to stroke pathology. These findings indicate a novel role for mast cells in the meninges, the membranes that envelop the brain, as potential gatekeepers for modulating brain inflammation and pathology after stroke.


Subject(s)
Mast Cells/immunology , Meninges/immunology , Stroke/immunology , Animals , Brain/immunology , Brain/pathology , Disease Models, Animal , Flow Cytometry , Gene Knock-In Techniques , Magnetic Resonance Imaging , Male , Mast Cells/pathology , Meninges/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Stroke/pathology
3.
Cell Rep ; 42(4): 112353, 2023 04 25.
Article in English | MEDLINE | ID: mdl-37043353

ABSTRACT

Stem cell therapy shows promise for multiple disorders; however, the molecular crosstalk between grafted cells and host tissue is largely unknown. Here, we take a step toward addressing this question. Using translating ribosome affinity purification (TRAP) with sequencing tools, we simultaneously decode the transcriptomes of graft and host for human neural stem cells (hNSCs) transplanted into the stroke-injured rat brain. Employing pathway analysis tools, we investigate the interactions between the two transcriptomes to predict molecular pathways linking host and graft genes; as proof of concept, we predict host-secreted factors that signal to the graft and the downstream molecular cascades they trigger in the graft. We identify a potential host-graft crosstalk pathway where BMP6 from the stroke-injured brain induces graft secretion of noggin, a known brain repair factor. Decoding the molecular interplay between graft and host is a critical step toward deciphering the molecular mechanisms of stem cell action.


Subject(s)
Neural Stem Cells , Stroke , Rats , Animals , Humans , Brain , Stroke/therapy , Stem Cell Transplantation , Cell Differentiation
4.
Stem Cells ; 29(2): 274-85, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21732485

ABSTRACT

Cell transplantation offers a novel therapeutic strategy for stroke; however, how transplanted cells function in vivo is poorly understood. We show for the first time that after subacute transplantation into the ischemic brain of human central nervous system stem cells grown as neurospheres (hCNS-SCns), the stem cell-secreted factor, human vascular endothelial growth factor (hVEGF), is necessary for cell-induced functional recovery. We correlate this functional recovery to hVEGF-induced effects on the host brain including multiple facets of vascular repair and its unexpected suppression of the inflammatory response. We found that transplanted hCNS-SCns affected multiple parameters in the brain with different kinetics: early improvement in blood-brain barrier integrity and suppression of inflammation was followed by a delayed spatiotemporal regulated increase in neovascularization. These events coincided with a bimodal pattern of functional recovery, with, an early recovery independent of neovascularization, and a delayed hVEGF-dependent recovery coincident with neovascularization. Therefore, cell transplantation therapy offers an exciting multimodal strategy for brain repair in stroke and potentially other disorders with a vascular or inflammatory component.


Subject(s)
Recovery of Function/physiology , Stem Cell Transplantation/methods , Stem Cells/metabolism , Stroke/therapy , Vascular Endothelial Growth Factors/metabolism , Animals , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/physiology , Brain/blood supply , Cell Differentiation , Cells, Cultured , Central Nervous System , Humans , Neovascularization, Physiologic , Rats , Rats, Nude , Wound Healing
5.
Brain ; 134(Pt 6): 1777-89, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21616972

ABSTRACT

Stem cell transplantation promises new hope for the treatment of stroke although significant questions remain about how the grafted cells elicit their effects. One hypothesis is that transplanted stem cells enhance endogenous repair mechanisms activated after cerebral ischaemia. Recognizing that bilateral reorganization of surviving circuits is associated with recovery after stroke, we investigated the ability of transplanted human neural progenitor cells to enhance this structural plasticity. Our results show the first evidence that human neural progenitor cell treatment can significantly increase dendritic plasticity in both the ipsi- and contralesional cortex and this coincides with stem cell-induced functional recovery. Moreover, stem cell-grafted rats demonstrated increased corticocortical, corticostriatal, corticothalamic and corticospinal axonal rewiring from the contralesional side; with the transcallosal and corticospinal axonal sprouting correlating with functional recovery. Furthermore, we demonstrate that axonal transport, which is critical for both proper axonal function and axonal sprouting, is inhibited by stroke and that this is rescued by the stem cell treatment, thus identifying another novel potential mechanism of action of transplanted cells. Finally, we established in vitro co-culture assays in which these stem cells mimicked the effects observed in vivo. Through immunodepletion studies, we identified vascular endothelial growth factor, thrombospondins 1 and 2, and slit as mediators partially responsible for stem cell-induced effects on dendritic sprouting, axonal plasticity and axonal transport in vitro. Thus, we postulate that human neural progenitor cells aid recovery after stroke through secretion of factors that enhance brain repair and plasticity.


Subject(s)
Axonal Transport/physiology , Brain Ischemia/surgery , Cerebral Cortex/cytology , Neural Stem Cells , Neuronal Plasticity/physiology , Analysis of Variance , Animals , Biotin/analogs & derivatives , Biotin/metabolism , Brain Infarction/etiology , Brain Infarction/pathology , Brain Ischemia/complications , Cell Survival , Cells, Cultured , Corpus Callosum/pathology , Dendrites/physiology , Dextrans/metabolism , Disease Models, Animal , Fetus , Gene Expression Regulation , Humans , Male , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/physiology , Neural Stem Cells/transplantation , Psychomotor Performance/physiology , RNA, Messenger/metabolism , Rats , Rats, Nude , Rats, Sprague-Dawley , Time Factors , Vibrissae/innervation
6.
Neurobiol Dis ; 37(2): 275-83, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19822211

ABSTRACT

Stem cell transplantation has evolved as a promising experimental treatment approach for stroke. In this review, we address the major hurdles for successful translation from basic research into clinical applications and discuss possible strategies to overcome these issues. We summarize the results from present pre-clinical and clinical studies and focus on specific areas of current controversy and research: (i) the therapeutic time window for cell transplantation; (ii) the selection of patients likely to benefit from such a therapy; (iii) the optimal route of cell delivery to the ischemic brain; (iv) the most suitable cell types and sources; (v) the potential mechanisms of functional recovery after cell transplantation; and (vi) the development of imaging techniques to monitor cell therapy.


Subject(s)
Brain/surgery , Stem Cell Transplantation/methods , Stroke/surgery , Animals , Brain/pathology , Brain/physiopathology , Cell Proliferation , Graft Survival/physiology , Humans , Microinjections/methods , Microinjections/trends , Patient Selection , Recovery of Function/physiology , Stem Cells/cytology , Stem Cells/physiology , Stroke/physiopathology , Time Factors
7.
Front Neurol ; 11: 236, 2020.
Article in English | MEDLINE | ID: mdl-32318016

ABSTRACT

Stroke is one of the major causes of chronic disability worldwide and increasing efforts have focused on studying brain repair and recovery after stroke. Following stroke, the primary injury site can disrupt functional connections in nearby and remotely connected brain regions, resulting in the development of secondary injuries that may impede long-term functional recovery. In particular, secondary degenerative injury occurs in the connected ipsilesional thalamus following a cortical stroke. Although secondary thalamic injury was first described decades ago, the underlying mechanisms still remain unclear. We performed a systematic literature review using the NCBI PubMed database for studies that focused on the secondary thalamic degeneration after cortical ischemic stroke. In this review, we discussed emerging studies that characterized the pathological changes in the secondary degenerative thalamus after stroke; these included excitotoxicity, apoptosis, amyloid beta protein accumulation, blood-brain-barrier breakdown, and inflammatory responses. In particular, we highlighted key findings of the dynamic inflammatory responses in the secondary thalamic injury and discussed the involvement of several cell types in this process. We also discussed studies that investigated the effects of blocking secondary thalamic injury on inflammatory responses and stroke outcome. Targeting secondary injuries after stroke may alleviate network-wide deficits, and ultimately promote stroke recovery.

8.
Stroke ; 40(1): 270-7, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18948613

ABSTRACT

BACKGROUND AND PURPOSE: Vascular endothelial growth factor (VEGF) and VEGF receptors (VEGFRs) play important roles during neurovascular repair after stroke. In this study, we imaged VEGFR expression with positron emission tomography (PET) to noninvasively analyze poststroke angiogenesis. METHODS: Female Sprague-Dawley rats after distal middle cerebral artery occlusion surgery were subjected to weekly MRI, (18)F-FDG PET, and (64)Cu-DOTA-VEGF(121) PET scans. Several control experiments were performed to confirm the VEGFR specificity of (64)Cu-DOTA-VEGF(121) uptake in the stroke border zone. VEGFR, BrdU, lectin staining, and (125)I-VEGF(165) autoradiography on stroke brain tissue slices were performed to validate the in vivo findings. RESULTS: T2-weighed MRI correlated with the "cold spot" on (18)F-FDG PET for rats undergoing distal middle cerebral artery occlusion surgery. The (64)Cu-DOTA-VEGF(121) uptake in the stroke border zone peaked at approximately 10 days after surgery, indicating neovascularization as confirmed by histology (VEGFR-2, BrdU, and lectin staining). VEGFR specificity of (64)Cu-DOTA-VEGF(121) uptake was confirmed by significantly lower uptake of (64)Cu-DOTA-VEGF(mutant) in vivo and intense (125)I-VEGF(165) uptake ex vivo in the stroke border zone. No appreciable uptake of (64)Cu-DOTA-VEGF(121) was observed in the brain of sham-operated rats. CONCLUSIONS: For the first time to our knowledge, we successfully evaluated the VEGFR expression kinetics noninvasively in a rat stroke model. In vivo imaging of VEGFR expression could become a significant clinical tool to plan and monitor therapies aimed at improving poststroke angiogenesis.


Subject(s)
Cerebral Arteries/diagnostic imaging , Infarction, Middle Cerebral Artery/diagnostic imaging , Neovascularization, Physiologic/physiology , Positron-Emission Tomography/methods , Receptors, Vascular Endothelial Growth Factor/analysis , Stroke/diagnostic imaging , Animals , Biomarkers/analysis , Biomarkers/metabolism , Bromodeoxyuridine , Cell Proliferation , Cerebral Arteries/metabolism , Copper Radioisotopes , Disease Models, Animal , Female , Fluorodeoxyglucose F18 , Infarction, Middle Cerebral Artery/physiopathology , Kinetics , Magnetic Resonance Imaging , Rats , Rats, Sprague-Dawley , Receptors, Vascular Endothelial Growth Factor/metabolism , Recovery of Function/physiology , Stroke/physiopathology , Vascular Endothelial Growth Factor A/analysis , Vascular Endothelial Growth Factor A/metabolism
9.
Front Cell Neurosci ; 13: 126, 2019.
Article in English | MEDLINE | ID: mdl-31001088

ABSTRACT

Stroke is the leading cause of adult disability in the United States. Because post-stroke inflammation is a critical determinant of damage and recovery after stroke, understanding the interplay between the immune system and the brain after stroke holds much promise for therapeutic intervention. An understudied, but important aspect of this interplay is the role of meninges that surround the brain. All blood vessels travel through the meningeal space before entering the brain parenchyma, making the meninges ideally located to act as an immune gatekeeper for the underlying parenchyma. Emerging evidence suggests that the actions of immune cells resident in the meninges are essential for executing this gatekeeper function. Mast cells (MCs), best known as proinflammatory effector cells, are one of the long-term resident immune cells in the meninges. Here, we discuss recent findings in the literature regarding the role of MCs located in the meningeal space and stroke pathology. We review the latest advances in mouse models to investigate the roles of MCs and MC-derived products in vivo, and the importance of using these mouse models. We examine the concept of the meninges playing a critical role in brain and immune interactions, reevaluate the perspectives on the key effectors of stroke pathology, and discuss the opportunities and challenges for therapeutic development.

10.
J Neurosci Res ; 86(4): 873-82, 2008 Mar.
Article in English | MEDLINE | ID: mdl-17975825

ABSTRACT

Endogenous neural stem cells normally reside in their niche, the subventricular zone, in the uninjured rodent brain. Upon stroke, these cells become more proliferative and migrate away from the subventricular zone into the surrounding parenchyma. It is not known whether this stroke-induced behavior is due to changes in the niche or introduction of attractive cues in the infarct zone, or both. A related question is how transplanted neural stem cells respond to subsequent insults, including whether exogenous stem cells have the plasticity to respond to subsequent injuries after engraftment. We addressed this issue by transplanting neural progenitor cells (NPCs) into the uninjured brain and then subjecting the animal to stroke. We were able to follow the transplanted NPCs in vivo by labeling them with superparamagnetic iron oxide particles and imaging them via high-resolution magnetic resonance imaging (MRI) during engraftment and subsequent to stroke. We find that transplanted NPCs that are latent can be activated in response to stroke and exhibit directional migration into the parenchyma, similar to endogenous neural NPCs, without a niche environment.


Subject(s)
Cell Movement/physiology , Neurons/transplantation , Stem Cell Transplantation , Stem Cells/physiology , Stroke/surgery , Animals , Cell Line , Immunohistochemistry , Magnetic Resonance Imaging , Male , Mice , Neurons/physiology , Rats , Rats, Sprague-Dawley , Staining and Labeling
11.
J Neurosci Methods ; 173(2): 286-90, 2008 Aug 30.
Article in English | MEDLINE | ID: mdl-18621079

ABSTRACT

Intracerebral injection of the vasoconstrictor peptide, endothelin-1 (ET-1), has been used as a method to induce focal ischemia in rats. The relative technical simplicity of this model makes it attractive for use in mice. However, the effect of ET-1 on mouse brains has not been firmly established. In this study, we determined the ability of ET-1 to induce focal cerebral ischemia in four different mouse strains (CD1, C57/BL6, NOD/SCID, and FVB). In contrast to rats, intracerebral injection of ET-1 did not produce a lesion in any mouse strain tested. A combination of ET-1 injection with either CCA occlusion or N(G)-nitro-l-arginine methyl ester (l-NAME) injection produced only a small infarct and its size was strain-dependent. A triple combination of CCA occlusion with co-injection of ET-1 and l-NAME produced a lesion in all mouse strains tested, and this resulted in a significant motor deficit. However, lesion size was still relatively small and strain-dependent. This study shows that ET-1 has a much less potent effect for producing an infarct in mice than rats.


Subject(s)
Brain Ischemia/chemically induced , Cerebral Arteries/drug effects , Disease Models, Animal , Endothelin-1/pharmacology , Vasoconstriction/drug effects , Animals , Brain Infarction/chemically induced , Brain Infarction/pathology , Brain Infarction/physiopathology , Brain Ischemia/pathology , Brain Ischemia/physiopathology , Carotid Artery Diseases/etiology , Carotid Artery Diseases/physiopathology , Cerebral Arteries/pathology , Cerebral Arteries/physiopathology , Enzyme Inhibitors/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, SCID , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase Type III/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Species Specificity , Vasoconstriction/physiology , Vasoconstrictor Agents/pharmacology
12.
Biomaterials ; 178: 63-72, 2018 09.
Article in English | MEDLINE | ID: mdl-29909038

ABSTRACT

Currently, no medical therapies exist to augment stroke recovery. Stem cells are an intriguing treatment option being evaluated, but cell-based therapies have several challenges including developing a stable cell product with long term reproducibility. Since much of the improvement observed from cellular therapeutics is believed to result from trophic factors the stem cells release over time, biomaterials are well-positioned to deliver these important molecules in a similar fashion. Here we show that essential trophic factors secreted from stem cells can be effectively released from a multi-component hydrogel system into the post-stroke environment. Using our polymeric system to deliver VEGF-A and MMP-9, we improved recovery after stroke to an equivalent degree as observed with traditional stem cell treatment in a rodent model. While VEGF-A and MMP-9 have many unique mechanisms of action, connective tissue growth factor (CTGF) interacts with both VEGF-A and MMP-9. With our hydrogel system as well as with stem cell delivery, the CTGF pathway is shown to be downregulated with improved stroke recovery.


Subject(s)
Neural Stem Cells/transplantation , Recovery of Function/physiology , Stem Cell Transplantation , Stroke/physiopathology , Stroke/therapy , Tissue Engineering , Animals , Connective Tissue Growth Factor/metabolism , Humans , Hydrogels/pharmacology , Injections , Male , Matrix Metalloproteinase 9/metabolism , Models, Biological , Neural Stem Cells/drug effects , Rats, Nude , Recovery of Function/drug effects , Vascular Endothelial Growth Factor A/metabolism
14.
Stroke ; 38(2 Suppl): 817-26, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17261746

ABSTRACT

No treatment currently exists to restore lost neurological function after stroke. A growing number of studies highlight the potential of stem cell transplantation as a novel therapeutic approach for stroke. In this review we summarize these studies, discuss potential mechanisms of action of the transplanted cells, and emphasize the need to determine parameters that are critical for transplantation success.


Subject(s)
Stem Cell Transplantation/methods , Stroke/surgery , Animals , Brain Tissue Transplantation/methods , Cell Transplantation/methods , Humans , Stroke/pathology
15.
Biomaterials ; 142: 31-40, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28719819

ABSTRACT

Exogenous human neural progenitor cells (hNPCs) are promising stroke therapeutics, but optimal delivery conditions and exact recovery mechanisms remain elusive. To further elucidate repair processes and improve stroke outcomes, we developed an electrically conductive, polymer scaffold for hNPC delivery. Electrical stimulation of hNPCs alters their transcriptome including changes to the VEGF-A pathway and genes involved in cell survival, inflammatory response, and synaptic remodeling. In our experiments, exogenous hNPCs were electrically stimulated (electrically preconditioned) via the scaffold 1 day prior to implantation. After in vitro stimulation, hNPCs on the scaffold are transplanted intracranially in a distal middle cerebral artery occlusion rat model. Electrically preconditioned hNPCs improved functional outcomes compared to unstimulated hNPCs or hNPCs where VEGF-A was blocked during in vitro electrical preconditioning. The ability to manipulate hNPCs via a conductive scaffold creates a new approach to optimize stem cell-based therapy and determine which factors (such as VEGF-A) are essential for stroke recovery.


Subject(s)
Electric Conductivity , Neural Stem Cells/metabolism , Neural Stem Cells/transplantation , Polymers/chemistry , Recovery of Function , Stroke/physiopathology , Stroke/therapy , Tissue Scaffolds/chemistry , Animals , Brain Infarction/pathology , Electric Stimulation , Gene Expression Regulation , Humans , Male , Pyrroles/chemistry , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Nude , Vascular Endothelial Growth Factor A/metabolism
16.
Front Neurol ; 7: 41, 2016.
Article in English | MEDLINE | ID: mdl-27047447

ABSTRACT

Stem cell therapies can promote neural repair and regeneration, yet controversy regarding optimal cell source and mechanism of action has slowed clinical translation, potentially due to undefined cellular heterogeneity. Single-cell resolution is needed to identify clinically relevant subpopulations with the highest therapeutic relevance. We combine single-cell microfluidic analysis with advanced computational modeling to study for the first time two common sources for cell-based therapies, human NSCs and MSCs. This methodology has the potential to logically inform cell source decisions for any clinical application.

17.
J Neurosci ; 24(27): 6202-8, 2004 Jul 07.
Article in English | MEDLINE | ID: mdl-15240812

ABSTRACT

Increasing evidence suggests that glutamate activates the generation of lactate from glucose in astrocytes; this lactate is shuttled to neurons that use it as a preferential energy source. We explore this multicellular "lactate shuttle" with a novel dual-cell, dual-gene therapy approach and determine the neuroprotective potential of enhancing this shuttle. Viral vector-driven overexpression of a glucose transporter in glia enhanced glucose uptake, lactate efflux, and the glial capacity to protect neurons from excitotoxicity. In parallel, overexpression of a lactate transporter in neurons enhanced lactate uptake and neuronal resistance to excitotoxicity. Finally, overexpression of both transgenes in the respective cell types provided more protection than either therapy alone, demonstrating that a dual-cell, dual-gene therapy approach gives greater neuroprotection than the conventional single-cell, single-gene strategy.


Subject(s)
Energy Metabolism/genetics , Genetic Therapy/methods , Neuroglia/drug effects , Neurons/drug effects , Neurons/metabolism , Neurotoxins/toxicity , Animals , Cell Survival/drug effects , Cell Survival/genetics , Cells, Cultured , Cytoprotection/drug effects , Cytoprotection/genetics , Drug Synergism , Energy Metabolism/drug effects , Feasibility Studies , Glucose/metabolism , Glucose/pharmacokinetics , Glucose Transporter Type 1 , Lactic Acid/metabolism , Lactic Acid/pharmacokinetics , Monocarboxylic Acid Transporters/biosynthesis , Monocarboxylic Acid Transporters/genetics , Monocarboxylic Acid Transporters/pharmacology , Monosaccharide Transport Proteins/biosynthesis , Monosaccharide Transport Proteins/genetics , Monosaccharide Transport Proteins/pharmacology , Neuroglia/metabolism , Plasmids/genetics , Plasmids/pharmacology , Rats , Rats, Sprague-Dawley , Transfection/methods , Transgenes
18.
J Neurosci Methods ; 196(2): 247-57, 2011 Mar 30.
Article in English | MEDLINE | ID: mdl-21256866

ABSTRACT

Middle cerebral artery occlusion (MCAO) in rats is a well-studied experimental model for ischemic stroke leading to brain infarction and functional deficits. Many preclinical studies have focused on a small time window after the ischemic episode to evaluate functional outcome for screening therapeutic candidates. Short evaluation periods following injury have led to significant setbacks due to lack of information on the delayed effects of treatments, as well as short-lived and reversible neuroprotection, so called false-positive results. In this report, we evaluated long-term functional deficit for 90 days after MCAO in two rat strains with two durations of ischemic insult, in order to identify the best experimental paradigm to assess injury and subsequent recovery. Behavioral outcomes were measured pre-MCAO followed by weekly assessment post-stroke. Behavioral tests included the 18-point composite neurological score, 28-point neuroscore, rearing test, vibrissae-evoked forelimb placing test, foot fault test and the CatWalk. Brain lesions were assessed to correlate injury to behavior outcomes at the end of study. Our results indicate that infarction volume in Sprague-Dawley rats was dependent on occlusion duration. In contrast, the infarction volume in Wistar rats did not correlate with the duration of ischemic episode. Functional outcomes were not dependent on occlusion time in either strain; however, measurable deficits were detectable long-term in limb asymmetry, 18- and 28-point neuroscores, forelimb placing, paw swing speed, and gait coordination. In conclusion, these behavioral assays, in combination with an extended long-term assessment period, can be used for evaluating therapeutic candidates in preclinical models of ischemic stroke.


Subject(s)
Behavior, Animal/physiology , Brain Ischemia/diagnosis , Brain Ischemia/physiopathology , Disability Evaluation , Stroke/diagnosis , Stroke/physiopathology , Animals , Brain Ischemia/pathology , Disease Models, Animal , Male , Neurologic Examination/methods , Rats , Rats, Sprague-Dawley , Rats, Wistar , Recovery of Function/physiology , Stroke/pathology
19.
Cell Transplant ; 18(7): 691-3, 2009.
Article in English | MEDLINE | ID: mdl-19796499

ABSTRACT

A consortium of translational stem cell and stroke experts from multiple academic institutes and biotechnology companies, under the guidance of the government (FDA/NIH), is missing. Here, we build a case for the establishment of this consortium if cell therapy for stroke is to advance from the laboratory to the clinic.


Subject(s)
Cell- and Tissue-Based Therapy , Stroke/therapy , Translational Research, Biomedical/organization & administration , Academies and Institutes , Animals , Clinical Trials as Topic/standards , Drug Evaluation, Preclinical/standards , Humans , National Institutes of Health (U.S.) , Private Sector , Stem Cell Transplantation , United States , United States Food and Drug Administration
20.
J Cereb Blood Flow Metab ; 28(10): 1722-32, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18594557

ABSTRACT

Thrombospondins 1 and 2 (TSP-1/2) belong to a family of extracellular glycoproteins with angiostatic and synaptogenic properties. Although TSP-1/2 have been postulated to drive the resolution of postischemic angiogenesis, their role in synaptic and functional recovery is unknown. We investigated whether TSP-1/2 are necessary for synaptic and motor recovery after stroke. Focal ischemia was induced in 8- to 12-week-old wild-type (WT) and TSP-1/2 knockout (KO) mice by unilateral occlusion of the distal middle cerebral artery and the common carotid artery (CCA). Thrombospondins 1 and 2 increased after stroke, with both TSP-1 and TSP-2 colocalizing mostly to astrocytes. Wild-type and TSP-1/2 KO mice were compared in angiogenesis, synaptic density, axonal sprouting, infarct size, and functional recovery at different time points after stroke. Using the tongue protrusion test of motor function, we observed that TSP-1/2 KO mice exhibited significant deficit in their ability to recover function (P<0.05) compared with WT mice. No differences were found in infarct size and blood vessel density between the two groups after stroke. However, TSP-1/2 KO mice exhibited significant synaptic density and axonal sprouting deficits. Deficiency of TSP-1/2 leads to impaired recovery after stroke mainly due to the role of these proteins in synapse formation and axonal outgrowth.


Subject(s)
Neuronal Plasticity/physiology , Recovery of Function/physiology , Stroke/physiopathology , Thrombospondin 1/metabolism , Thrombospondins/metabolism , Animals , Astrocytes/pathology , Astrocytes/physiology , Axons/pathology , Axons/physiology , Behavior, Animal/physiology , Cerebrovascular Circulation/physiology , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/physiopathology , Mice , Mice, Inbred Strains , Mice, Knockout , Neovascularization, Physiologic/physiology , Stroke/metabolism , Stroke/pathology , Synapses/physiology , Thrombospondin 1/genetics , Thrombospondins/genetics , Up-Regulation/physiology
SELECTION OF CITATIONS
SEARCH DETAIL