Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
ASN Neuro ; 10: 1759091418781921, 2018.
Article in English | MEDLINE | ID: mdl-29932344

ABSTRACT

An estimated 5.3 million Americans are living with a disability from a traumatic brain injury (TBI). There is emerging evidence of the detrimental effects from repeated mild TBIs (rmTBIs). rmTBI manifests its own unique set of behavioral and neuropathological changes. A subset of individuals exposed to rmTBI develop permanent behavioral and pathological consequences, defined postmortem as chronic traumatic encephalopathy. We have combined components of two classic rodent models of TBI, the controlled cortical impact model and the weight drop model, to develop a repeated mild closed head injury (rmCHI) that produces long-term deficits in several behaviors that correlate with neuropathological changes. Mice receiving rmCHI performed differently from 1-hit or sham controls on the elevated plus maze; these deficits persist up to 6 months postinjury (MPI). rmCHI mice performed worse than 1-hit and control sham mice at 2 MPI and 6 MPI on the Morris water maze. Mice receiving rmCHI exhibited significant atrophy of the corpus callosum at both 2 MPI and 6 MPI, as assessed by stereological volume analysis. Stereological analysis also revealed significant loss of cortical neurons in comparison with 1-hit and controls. Moreover, both of these pathological changes correlated with behavioral impairments. In human tau transgenic mice, rmCHI induced increases in hyperphosphorylated paired helical filament 1 tau in the hippocampus. This suggests that strategies to restore myelination or reduce neuronal loss may ameliorate the behavioral deficits observed following rmCHI and that rmCHI may model chronic traumatic encephalopathy in human tau mice.


Subject(s)
Head Injuries, Closed/complications , Head Injuries, Closed/pathology , Mental Disorders/etiology , Neurons/pathology , White Matter/pathology , Animals , Calcium-Binding Proteins/metabolism , Disease Models, Animal , Hindlimb Suspension , Longitudinal Studies , Male , Maze Learning , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microfilament Proteins/metabolism , Nerve Tissue Proteins/metabolism , Swimming , tau Proteins/genetics , tau Proteins/metabolism
2.
Cell Transplant ; 26(7): 1247-1261, 2017 07.
Article in English | MEDLINE | ID: mdl-28933218

ABSTRACT

Traumatic brain injury (TBI) is one of the leading causes of death and disability in the population worldwide, with a broad spectrum of symptoms and disabilities. Posttraumatic hyperexcitability is one of the most common neurological disorders that affect people after a head injury. A reliable animal model of posttraumatic hyperexcitability induced by TBI which allows one to test effective treatment strategies is yet to be developed. To address these issues, in the present study, we tested human embryonic stem cell-derived neural stem cell (NSC) transplantation in an animal model of posttraumatic hyperexcitability in which the brain injury was produced in one hemisphere of immunodeficient athymic nude rats by controlled cortical impact, and spontaneous seizures were produced by repeated electrical stimulation (kindling) in the contralateral hemisphere. At 14 wk posttransplantation, we report human NSC (hNSC) survival and differentiation into all 3 neural lineages in both sham and injured animals. We observed twice as many surviving hNSCs in the injured versus sham brain, and worse survival on the kindled side in both groups, indicating that kindling/seizures are detrimental to survival or proliferation of hNSCs. We also replicated our previous finding that hNSCs can ameliorate deficits on the novel place recognition task,33 but such improvements are abolished following kindling. We found no significant differences pre- or post-kindling on the elevated plus maze. No significant correlations were observed between hNSC survival and cognitive performance on either task. Together these findings suggest that Shef6-derived hNSCs may be beneficial as a therapy for TBI, but not in animals or patients with posttraumatic hyperexcitability.


Subject(s)
Brain Injuries, Traumatic/physiopathology , Brain Injuries, Traumatic/therapy , Human Embryonic Stem Cells/cytology , Neural Stem Cells/transplantation , Stem Cell Transplantation , Animals , Brain Injuries, Traumatic/pathology , Cell Count , Cell Differentiation , Cell Lineage , Cell Survival , Cognition , Disease Models, Animal , Humans , Kindling, Neurologic , Male , Maze Learning , Neural Stem Cells/cytology , Rats, Nude , Task Performance and Analysis
3.
Exp Neurol ; 281: 1-16, 2016 07.
Article in English | MEDLINE | ID: mdl-27079998

ABSTRACT

Traumatic brain injury (TBI) in humans can result in permanent tissue damage and has been linked to cognitive impairment that lasts years beyond the initial insult. Clinically effective treatment strategies have yet to be developed. Transplantation of human neural stem cells (hNSCs) has the potential to restore cognition lost due to injury, however, the vast majority of rodent TBI/hNSC studies to date have evaluated cognition only at early time points, typically <1month post-injury and cell transplantation. Additionally, human cell engraftment and long-term survival in rodent models of TBI has been difficult to achieve due to host immunorejection of the transplanted human cells, which confounds conclusions pertaining to transplant-mediated behavioral improvement. To overcome these shortfalls, we have developed a novel TBI xenotransplantation model that utilizes immunodeficient athymic nude (ATN) rats as the host recipient for the post-TBI transplantation of human embryonic stem cell (hESC) derived NSCs and have evaluated cognition in these animals at long-term (≥2months) time points post-injury. We report that immunodeficient ATN rats demonstrate hippocampal-dependent spatial memory deficits (Novel Place, Morris Water Maze), but not non-spatial (Novel Object) or emotional/anxiety-related (Elevated Plus Maze, Conditioned Taste Aversion) deficits, at 2-3months post-TBI, confirming that ATN rats recapitulate some of the cognitive deficits found in immunosufficient animal strains. Approximately 9-25% of transplanted hNSCs survived for at least 5months post-transplantation and differentiated into mature neurons (NeuN, 18-38%), astrocytes (GFAP, 13-16%), and oligodendrocytes (Olig2, 11-13%). Furthermore, while this model of TBI (cortical impact) targets primarily cortex and the underlying hippocampus and generates a large lesion cavity, hNSC transplantation facilitated cognitive recovery without affecting either lesion volume or total spared cortical or hippocampal tissue volume. Instead, we have found an overall increase in host hippocampal neuron survival in hNSC transplanted animals and demonstrate that a correlation exists between hippocampal neuron survival and cognitive performance. Together, these findings support the use of immunodeficient rodents in models of TBI that involve the transplantation of human cells, and suggest that hNSC transplantation may be a viable, long-term therapy to restore cognition after brain injury.


Subject(s)
Brain Injuries, Traumatic/complications , Cognition Disorders/etiology , Cognition Disorders/surgery , Neural Stem Cells/transplantation , Animals , Antigens, CD/metabolism , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/surgery , Cell Differentiation , Conditioning, Classical , Disease Models, Animal , Escape Reaction/physiology , Exploratory Behavior/physiology , Hippocampus/pathology , Humans , Male , Maze Learning/physiology , Nerve Tissue Proteins/metabolism , Neural Stem Cells/metabolism , Neurogenesis , Neurons/metabolism , Neurons/pathology , Rats , Rats, Nude , Recognition, Psychology/physiology , Spatial Behavior
4.
PLoS One ; 10(5): e0126551, 2015.
Article in English | MEDLINE | ID: mdl-25946089

ABSTRACT

Mesenchymal stromal cells secrete a variety of anti-inflammatory factors and may provide a regenerative medicine option for the treatment of traumatic brain injury. The present study investigates the efficacy of multiple intravenous or intracardiac administrations of rat mesenchymal stromal cells or human mesenchymal stromal cells in female rats after controlled cortical impact by in vivo MRI, neurobehavior, and histopathology evaluation. Neither intravenous nor intracardiac administration of mesenchymal stromal cells derived from either rats or humans improved MRI measures of lesion volume or neurobehavioral outcome compared to saline treatment. Few mesenchymal stromal cells (<0.0005% of injected dose) were found within 3 days of last dosage at the site of injury after either delivery route, with no mesenchymal stromal cells being detectable in brain at 30 or 56 days post-injury. These findings suggest that non-autologous mesenchymal stromal cells therapy via intravenous or intracardiac administration is not a promising treatment after focal contusion traumatic brain injury in this female rodent model.


Subject(s)
Brain Injuries/therapy , Brain/pathology , Cell- and Tissue-Based Therapy/methods , Mesenchymal Stem Cell Transplantation/methods , Administration, Intravenous , Animals , Brain/cytology , Cardiac Catheters , Cells, Cultured , Disease Models, Animal , Female , Humans , Image Processing, Computer-Assisted , Magnetic Resonance Imaging , Mesenchymal Stem Cells/cytology , Rats , Rats, Wistar , Treatment Failure
5.
Stem Cell Res ; 13(2): 214-26, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25082219

ABSTRACT

Common methods for the generation of human embryonic-derived neural stem cells (hNSCs) result in cells with potentially compromised safety profiles due to maintenance of cells in conditions containing non-human proteins (e.g. in bovine serum or on mouse fibroblast feeders). Additionally, sufficient expansion of resulting hNSCs for scaling out or up in a clinically relevant time frame has proven to be difficult. Here, we report a strategy that produces hNSCs in completely "Xeno-Free" culture conditions. Furthermore, we have enriched the hNSCs for the cell surface marker CD133 via magnetic sorting, which has led to an increase in the expansion rate and neuronal fate specification of the hNSCs in vitro. Critically, we have also confirmed neural lineage specificity upon sorted hNSC transplantation into the immunodeficient NOD-scid mouse brain. The future use or adaptation of these protocols has the potential to better facilitate the advancement of pre-clinical strategies from the bench to the bedside.


Subject(s)
Antigens, CD/metabolism , Cell Proliferation , Embryonic Stem Cells/metabolism , Glycoproteins/metabolism , Neural Stem Cells/metabolism , Neurogenesis , Peptides/metabolism , Teratoma/immunology , AC133 Antigen , Animals , Biomarkers/metabolism , Cell Culture Techniques , Cell Line , Cell Lineage , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Embryonic Stem Cells/immunology , Embryonic Stem Cells/pathology , Embryonic Stem Cells/transplantation , Flow Cytometry , Heterografts , Humans , Immunomagnetic Separation/methods , Mice, Inbred NOD , Mice, SCID , Neural Stem Cells/immunology , Neural Stem Cells/pathology , Neural Stem Cells/transplantation , Phenotype , Teratoma/metabolism , Teratoma/pathology , Time Factors
6.
Regen Med ; 8(4): 483-516, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23826701

ABSTRACT

Traumatic brain injury (TBI) ranks as the leading cause of mortality and disability in the young population worldwide. The annual US incidence of TBI in the general population is estimated at 1.7 million per year, with an estimated financial burden in excess of US$75 billion a year in the USA alone. Despite the prevalence and cost of TBI to individuals and society, no treatments have passed clinical trial to clinical implementation. The rapid expansion of stem cell research and technology offers an alternative to traditional pharmacological approaches targeting acute neuroprotection. However, preclinical testing of these approaches depends on the selection and characterization of appropriate animal models. In this article we consider the underlying pathophysiology for the focal and diffuse TBI subtypes, discuss the existing preclinical TBI models and functional outcome tasks used for assessment of injury and recovery, identify criteria particular to preclinical animal models of TBI in which stem cell therapies can be tested for safety and efficacy, and review these criteria in the context of the existing TBI literature. We suggest that 2 months post-TBI is the minimum period needed to evaluate human cell transplant efficacy and safety. Comprehensive review of the published TBI literature revealed that only 32% of rodent TBI papers evaluated functional outcome ≥1 month post-TBI, and only 10% evaluated functional outcomes ≥2 months post-TBI. Not all published papers that evaluated functional deficits at a minimum of 2 months post-TBI reported deficits; hence, only 8.6% of overall TBI papers captured in this review demonstrated functional deficits at 2 months or more postinjury. A 2-month survival and assessment period would allow sufficient time for differentiation and integration of human neural stem cells with the host. Critically, while trophic effects might be observed at earlier time points, it will also be important to demonstrate the sustainability of such an effect, supporting the importance of an extended period of in vivo observation. Furthermore, regulatory bodies will likely require at least 6 months survival post-transplantation for assessment of toxicology/safety, particularly in the context of assessing cell abnormalities.


Subject(s)
Behavior, Animal , Brain Injuries/physiopathology , Disease Models, Animal , Animals , Brain Injuries/etiology , Humans , Phenotype , Rodentia
7.
NMR Biomed ; 26(4): 468-79, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23225324

ABSTRACT

Serial MRI facilitates the in vivo analysis of the intra- and intersubject evolution of traumatic brain injury lesions. Despite the availability of MRI, the natural history of experimental focal contusion lesions in the controlled cortical impact (CCI) rat model has not been well described. We performed CCI on rats and MRI during the acute to chronic stages of cerebral injury to investigate the time course of changes in the brain. Female Wistar rats underwent CCI of their left motor cortex with a flat impact tip driven by an electromagnetic piston. In vivo MRI was performed at 7 T serially over 6 weeks post-CCI. The appearances of CCI-induced lesions and lesion-associated cortical volumes were variable on MRI, with the percentage change in cortical volume of the CCI ipsilateral side relative to the contralateral side ranging from 18% within 2 h of injury on day 0 to a peak of 35% on day 1, and a trough of -28% by week 5/6, with an average standard deviation of ± 14% at any given time point. In contrast, the percentage change in cortical volume of the ipsilateral side relative to the contralateral side in control rats was not significant (1 ± 2%). Hemorrhagic conversion within and surrounding the CCI lesion occurred between days 2 and 9 in 45% of rats, with no hemorrhage noted on the initial scan. Furthermore, hemorrhage and hemosiderin within the lesion were positive for Prussian blue and highly autofluorescent on histological examination. Although some variation in injuries may be technique related, the divergence of similar lesions between initial and final scans demonstrates the inherent biological variability of the CCI rat model.


Subject(s)
Brain Injuries/complications , Brain Injuries/pathology , Contusions/complications , Contusions/pathology , Animals , Behavior, Animal , Cerebral Cortex/pathology , Cerebral Hemorrhage/complications , Cerebral Hemorrhage/pathology , Disease Models, Animal , Female , Magnetic Resonance Imaging , Microscopy, Fluorescence , Organ Size , Rats, Wistar
8.
Stem Cells ; 30(6): 1216-27, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22593018

ABSTRACT

Bone marrow stromal cells (BMSCs) have shown significant promise in the treatment of disease, but their therapeutic efficacy is often limited by inefficient homing of systemically administered cells, which results in low number of cells accumulating at sites of pathology. BMSC home to areas of inflammation where local expression of integrins and chemokine gradients is present. We demonstrated that nondestructive pulsed focused ultrasound (pFUS) exposures that emphasize the mechanical effects of ultrasound-tissue interactions induced local and transient elevations of chemoattractants (i.e., cytokines, integrins, and growth factors) in the murine kidney. pFUS-induced upregulation of cytokines occurred through approximately 1 day post-treatment and returned to contralateral kidney levels by day 3. This window of significant increases in cytokine expression was accompanied by local increases of other trophic factors and integrins that have been shown to promote BMSC homing. When BMSCs were intravenously administered following pFUS treatment to a single kidney, enhanced homing, permeability, and retention of BMSC was observed in the treated kidney versus the contralateral kidney. Histological analysis revealed up to eight times more BMSC in the peritubular regions of the treated kidneys on days 1 and 3 post-treatment. Furthermore, cytokine levels in pFUS-treated kidneys following BMSC administration were found to be similar to controls, suggesting modulation of cytokine levels by BMSC. pFUS could potentially improve cell-based therapies as a noninvasive modality to target homing by establishing local chemoattractant gradients and increasing expression of integrins to enhance tropism of cells toward treated tissues.


Subject(s)
Bone Marrow Cells/cytology , Bone Marrow Cells/diagnostic imaging , Bone Marrow Transplantation/methods , Kidney/cytology , Kidney/diagnostic imaging , Stromal Cells/transplantation , Ultrasonics/methods , Animals , Bone Marrow Cells/metabolism , Bone Marrow Transplantation/diagnostic imaging , Cell Culture Techniques , Cytokines/metabolism , Female , Humans , Immunohistochemistry , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/diagnostic imaging , Mice , Mice, Inbred BALB C , Mice, Nude , Stromal Cells/cytology , Ultrasonography
9.
Nanotechnology ; 20(35): 355102, 2009 Sep 02.
Article in English | MEDLINE | ID: mdl-19671960

ABSTRACT

For cellular MRI there is a need to label cells with superparamagnetic iron oxide nanoparticles (SPION) that have multiple imaging moieties that are nontoxic and have increased NMR relaxation properties to improve the detection and tracking of therapeutic cells. Although increases in the relaxation properties of SPION have been accomplished, detection of tagged cells is limited by either poor cell labeling efficiency or low intracellular iron content. A strategy via a complex formation with transfection agents to overcome these obstacles has been reported. In this paper, we report a complex formation between negatively charged fluorescent monodisperse SPION and positively charged peptides and use the complex formation to improve the MR properties of labeled stem cells. As a result, labeled stem cells exhibited a strong fluorescent signal and enhanced T 2*-weighted MR imaging in vitro and in vivo in a flank tumor model.


Subject(s)
Ferric Compounds/metabolism , Magnetics , Mesenchymal Stem Cells/cytology , Peptides/metabolism , Static Electricity , Animals , Cell Line , Chickens , Humans , Iron/metabolism , Magnetic Resonance Imaging , Mice , Microscopy, Fluorescence , Phantoms, Imaging , Staining and Labeling , Whole Body Imaging
SELECTION OF CITATIONS
SEARCH DETAIL
...