Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Transplant ; 26(7): 1247-1261, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28933218

RESUMO

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.


Assuntos
Lesões Encefálicas Traumáticas/fisiopatologia , Lesões Encefálicas Traumáticas/terapia , Células-Tronco Embrionárias Humanas/citologia , Células-Tronco Neurais/transplante , Transplante de Células-Tronco , Animais , Lesões Encefálicas Traumáticas/patologia , Contagem de Células , Diferenciação Celular , Linhagem da Célula , Sobrevivência Celular , Cognição , Modelos Animais de Doenças , Humanos , Excitação Neurológica , Masculino , Aprendizagem em Labirinto , Células-Tronco Neurais/citologia , Ratos Nus , Análise e Desempenho de Tarefas
2.
Exp Neurol ; 281: 1-16, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27079998

RESUMO

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.


Assuntos
Lesões Encefálicas Traumáticas/complicações , Transtornos Cognitivos/etiologia , Transtornos Cognitivos/cirurgia , Células-Tronco Neurais/transplante , Animais , Antígenos CD/metabolismo , Lesões Encefálicas Traumáticas/patologia , Lesões Encefálicas Traumáticas/cirurgia , Diferenciação Celular , Condicionamento Clássico , Modelos Animais de Doenças , Reação de Fuga/fisiologia , Comportamento Exploratório/fisiologia , Hipocampo/patologia , Humanos , Masculino , Aprendizagem em Labirinto/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Células-Tronco Neurais/metabolismo , Neurogênese , Neurônios/metabolismo , Neurônios/patologia , Ratos , Ratos Nus , Reconhecimento Psicológico/fisiologia , Comportamento Espacial
3.
Stem Cell Res ; 13(2): 214-26, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25082219

RESUMO

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.


Assuntos
Antígenos CD/metabolismo , Proliferação de Células , Células-Tronco Embrionárias/metabolismo , Glicoproteínas/metabolismo , Células-Tronco Neurais/metabolismo , Neurogênese , Peptídeos/metabolismo , Teratoma/imunologia , Antígeno AC133 , Animais , Biomarcadores/metabolismo , Técnicas de Cultura de Células , Linhagem Celular , Linhagem da Célula , Transformação Celular Neoplásica/imunologia , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Células-Tronco Embrionárias/imunologia , Células-Tronco Embrionárias/patologia , Células-Tronco Embrionárias/transplante , Citometria de Fluxo , Xenoenxertos , Humanos , Separação Imunomagnética/métodos , Camundongos Endogâmicos NOD , Camundongos SCID , Células-Tronco Neurais/imunologia , Células-Tronco Neurais/patologia , Células-Tronco Neurais/transplante , Fenótipo , Teratoma/metabolismo , Teratoma/patologia , Fatores de Tempo
4.
Cell Rep ; 8(1): 217-28, 2014 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-25001280

RESUMO

Redox-mediated posttranslational modifications represent a molecular switch that controls major mechanisms of cell function. Nitric oxide (NO) can mediate redox reactions via S-nitrosylation, representing transfer of an NO group to a critical protein thiol. NO is known to modulate neurogenesis and neuronal survival in various brain regions in disparate neurodegenerative conditions. However, a unifying molecular mechanism linking these phenomena remains unknown. Here, we report that S-nitrosylation of myocyte enhancer factor 2 (MEF2) transcription factors acts as a redox switch to inhibit both neurogenesis and neuronal survival. Structure-based analysis reveals that MEF2 dimerization creates a pocket, facilitating S-nitrosylation at an evolutionally conserved cysteine residue in the DNA binding domain. S-Nitrosylation disrupts MEF2-DNA binding and transcriptional activity, leading to impaired neurogenesis and survival in vitro and in vivo. Our data define a molecular switch whereby redox-mediated posttranslational modification controls both neurogenesis and neurodegeneration via a single transcriptional signaling cascade.


Assuntos
Apoptose , Fatores de Transcrição MEF2/metabolismo , Células-Tronco Neurais/metabolismo , Neurogênese , Óxido Nítrico/metabolismo , Processamento de Proteína Pós-Traducional , Ativação Transcricional , Animais , Sítios de Ligação , Células Cultivadas , DNA/metabolismo , Células HEK293 , Humanos , Fatores de Transcrição MEF2/química , Fatores de Transcrição MEF2/genética , Camundongos , Células-Tronco Neurais/citologia , Oxirredução , Ligação Proteica
5.
J Comp Neurol ; 522(12): 2767-83, 2014 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-24715528

RESUMO

Human embryonic stem cells (hESC) and induced pluripotent stem cells (hiPSC) can differentiate into many cell types and are important for regenerative medicine; however, further work is needed to reliably differentiate hESC and hiPSC into neural-restricted multipotent derivatives or specialized cell types under conditions that are free from animal products. Toward this goal, we tested the transition of hESC and hiPSC lines onto xeno-free (XF) / feeder-free conditions and evaluated XF substrate preference, pluripotency, and karyotype. Critically, XF transitioned H9 hESC, Shef4 hESC, and iPS6-9 retained pluripotency (Oct-4 and NANOG), proliferation (MKI67 and PCNA), and normal karyotype. Subsequently, XF transitioned hESC and hiPSC were induced with epidermal growth factor (EGF) and basic fibroblast growth factor (bFGF) to generate neuralized spheres containing primitive neural precursors, which could differentiate into astrocytes and neurons, but not oligoprogenitors. Further neuralization of spheres via LIF supplementation and attachment selection on CELLstart substrate generated adherent human neural stem cells (hNSC) with normal karyotype and high proliferation potential under XF conditions. Interestingly, adherent hNSC derived from H9, Shef4, and iPS6-9 differentiated into significant numbers of O4+ oligoprogenitors (∼20-30%) with robust proliferation; however, very few GalC+ cells were observed (∼2-4%), indicative of early oligodendrocytic lineage commitment. Overall, these data demonstrate the transition of multiple hESC and hiPSC lines onto XF substrate and media conditions, and a reproducible neuralization method that generated neural derivatives with multipotent cell fate potential and normal karyotype.


Assuntos
Diferenciação Celular/fisiologia , Compômeros/metabolismo , Células-Tronco Embrionárias/fisiologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Adesão Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células , Células-Tronco Embrionárias/efeitos dos fármacos , Fator de Crescimento Epidérmico/farmacologia , Fator 2 de Crescimento de Fibroblastos/farmacologia , Citometria de Fluxo , Expressão Gênica , Proteínas de Homeodomínio/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intracelular , Cariótipo , Proteína Homeobox Nanog , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Oligodendroglia/fisiologia , Antígeno Nuclear de Célula em Proliferação/genética , Antígeno Nuclear de Célula em Proliferação/metabolismo
6.
Regen Med ; 8(4): 483-516, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23826701

RESUMO

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.


Assuntos
Comportamento Animal , Lesões Encefálicas/fisiopatologia , Modelos Animais de Doenças , Animais , Lesões Encefálicas/etiologia , Humanos , Fenótipo , Roedores
7.
Stem Cell Res ; 7(3): 256-63, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21775237

RESUMO

Accurate automated cell fate analysis of immunostained human stem cells from 2- and 3-dimensional (2D-3D) images would improve efficiency in the field of stem cell research. Development of an accurate and precise tool that reduces variability and the time needed for human stem cell fate analysis will improve productivity and interpretability of the data across research groups. In this study, we have created protocols for high performance image analysis software Volocity® to classify and quantify cytoplasmic and nuclear cell fate markers from 2D-3D images of human neural stem cells after in vitro differentiation. To enhance 3D image capture efficiency, we optimized the image acquisition settings of an Olympus FV10i® confocal laser scanning microscope to match our quantification protocols and improve cell fate classification. The methods developed in this study will allow for a more time efficient and accurate software based, operator validated, stem cell fate classification and quantification from 2D and 3D images, and yield the highest ≥94.4% correspondence with human recognized objects.


Assuntos
Linhagem da Célula , Processamento de Imagem Assistida por Computador/métodos , Software , Células-Tronco/citologia , Biomarcadores/metabolismo , Diferenciação Celular , Citoplasma/metabolismo , Humanos , Microscopia Confocal , Reprodutibilidade dos Testes , Células-Tronco/metabolismo , Fatores de Tempo
8.
Regen Med ; 6(3): 367-406, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21548741

RESUMO

There is potential for a variety of stem cell populations to mediate repair in the diseased or injured CNS; in some cases, this theoretical possibility has already transitioned to clinical safety testing. However, careful consideration of preclinical animal models is essential to provide an appropriate assessment of stem cell safety and efficacy, as well as the basic biological mechanisms of stem cell action. This article examines the lessons learned from early tissue, organ and hematopoietic grafting, the early assumptions of the stem cell and CNS fields with regard to immunoprivilege, and the history of success in stem cell transplantation into the CNS. Finally, we discuss strategies in the selection of animal models to maximize the predictive validity of preclinical safety and efficacy studies.


Assuntos
Sistema Nervoso Central/patologia , Sobrevivência de Enxerto/imunologia , Medicina Regenerativa/métodos , Transplante de Células-Tronco , Células-Tronco/citologia , Células-Tronco/imunologia , Animais , Sobrevivência Celular/imunologia , Modelos Animais de Doenças , Rejeição de Enxerto/imunologia , Hematopoese , Humanos , Doenças do Sistema Nervoso/patologia , Doenças do Sistema Nervoso/terapia , Células-Tronco/metabolismo , Transplante Heterólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...