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











Base de dados
Intervalo de ano de publicação
1.
Exp Neurol ; 374: 114694, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38272159

RESUMO

Parkinson's disease (PD) is a relentlessly progressive and currently incurable neurodegenerative disease with significant unmet medical needs. Since PD stems from the degeneration of midbrain dopaminergic (DA) neurons in a defined brain location, PD patients are considered optimal candidates for cell replacement therapy. Clinical trials for cell transplantation in PD are beginning to re-emerge worldwide with a new focus on induced pluripotent stem cells (iPSCs) as a source of DA neurons since they can be derived from adult somatic cells and produced in large quantities under current good manufacturing practices. However, for this therapeutic strategy to be realized as a viable clinical option, fundamental translational challenges need to be addressed including the manufacturing process, purity and efficacy of the cells, the method of delivery, the extent of host reinnervation and the impact of patient-centered adjunctive interventions. In this study we report on the impact of physical and cognitive training (PCT) on functional recovery in the nonhuman primate (NHP) model of PD after cell transplantation. We observed that at 6 months post-transplant, the PCT group returned to normal baseline in their daily activity measured by actigraphy, significantly improved in their sensorimotor and cognitive tasks, and showed enhanced synapse formation between grafted cells and host cells. We also describe a robust, simple, efficient, scalable, and cost-effective manufacturing process of engraftable DA neurons derived from iPSCs. This study suggests that integrating PCT with cell transplantation therapy could promote optimal graft functional integration and better outcome for patients with PD.


Assuntos
Células-Tronco Pluripotentes Induzidas , Doenças Neurodegenerativas , Doença de Parkinson , Adulto , Animais , Humanos , Neurônios Dopaminérgicos/fisiologia , Células-Tronco Pluripotentes Induzidas/transplante , Callithrix , Treino Cognitivo , Doença de Parkinson/cirurgia , Transplante de Células-Tronco/métodos , Diferenciação Celular/fisiologia
2.
Methods Mol Biol ; 2389: 165-175, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34558010

RESUMO

Neural stem cell therapy has been galvanized by the discovery of pluripotent stem cells. The possibility to generate specialized central nervous system-specific differentiated cells using human somatic cells engineered to become induced pluripotent stem cells (iPSCs) was a game changer. This technology has broad applications in the field of regenerative medicine, in vitro disease modeling, targeted drug discovery, and precision medicine. Currently, iPSCs are one of the most promising cell sources amenable for commercialization and off-the-shelf neural stem cell therapy products. iPSCs exhibit a strong self-renewable ability that supports the development of a virtually unlimited source of neural cells for structural repair in neurological disorders. However, along with this strong proliferative capacity of iPSCs comes the tumorigenic potential of these cells after transplantation. Thus, the isolation and purification of a homogeneous population of human neural stem cells (hNSCs) are of paramount importance to ensure consistency in the composition of the cellular product and to avoid tumor formation in the host brain. This chapter describes the isolation, neuralization, and long-term perpetuation of hNSCs derived from iPSCs through the use of specific growth medium and the preparation of hNSCs for transplantation in an experimental model of stroke. Additionally, we will describe methods to analyze the ischemic stroke and size of grafts using magnetic resonance imaging and OsiriX software and neuroanatomical tracing procedures to study axonal remodeling after ischemic stroke and cell transplantation.


Assuntos
Isquemia Encefálica , Autorrenovação Celular , Células-Tronco Pluripotentes Induzidas , AVC Isquêmico , Células-Tronco Neurais , Diferenciação Celular , Terapia Baseada em Transplante de Células e Tecidos , Humanos , AVC Isquêmico/terapia , Modelos Teóricos
3.
Methods Mol Biol ; 1919: 175-186, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30656629

RESUMO

Limited access to primary tissue from various nonhuman primate (NHP) species represents a significant unmet need that hampers progress in understanding unique cellular diversity and gene regulation of specific tissues and organs in stem cell translational research. Most comparative biology studies have been limited to using postmortem tissue usually frozen specimens with limited utility for research. The generation of induced pluripotent stem cell (iPSC) lines from somatic cells, such as adult skin or blood cells, offers an alternative to invasive and ethically controversial interventions for acquiring tissue. Pluripotent iPSCs have virtually an unlimited capacity to proliferate and differentiate into all cell types of the body. We are generating high-quality validated NHP iPSC lines to offer to scientific community and facilitate their research programs. We use the non-integrative episomal vector system to generate iPSCs from NHP skin biopsies. In this chapter we describe the validation of NHP iPSC lines by confirming pluripotency and their propensity to differentiate into all three germ layers ectoderm, mesoderm, and endoderm according to established standards and measurable limits for a set of marker genes incorporated into a scorecard.


Assuntos
Perfilação da Expressão Gênica , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Medicina Regenerativa , Transcriptoma , Animais , Biópsia , Callithrix , Linhagem Celular , Corpos Embrioides/metabolismo , Perfilação da Expressão Gênica/métodos , Pele/citologia , Pele/metabolismo , Fluxo de Trabalho
4.
IUBMB Life ; 69(9): 745-755, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28762248

RESUMO

Ultrasound-targeted microbubble destruction (UTMD) is a novel means of tissue-specific gene delivery. This approach systemically infuses transgenes precoupled to gas-filled lipid microbubbles that are burst within the microvasculature of target tissues via an ultrasound signal resulting in release of DNA and transfection of neighboring cells within the tissue. Previous work has shown that adenovirus containing cDNA of UCP-1, injected into the epididymal fat pads in mice, induced localized fat depletion, improving glucose tolerance, and decreasing food intake in obese diabetic mice. Our group recently demonstrated that gene therapy by UTMD achieved beta cell regeneration in streptozotocin (STZ)-treated mice and baboons. We hypothesized that gene therapy with BMP7/PRDM16/PPARGC1A in skeletal muscle (SKM) of obese Zucker diabetic fatty (fa/fa) rats using UTMD technology would produce a brown adipose tissue (BAT) phenotype with UCP-1 overexpression. This study was designed as a proof of concept (POC) project. Obese Zucker rats were administered plasmid cDNA contructs encoding a gene cocktail with BMP7/PRDM16/PPARGC1A incorporated within microbubbles and intravenously delivered into their left thigh. Controls received UTMD with plasmids driving a DsRed reporter gene. An ultrasound transducer was directed to the thigh to disrupt the microbubbles within the microcirculation. Blood samples were drawn at baseline, and after treatment to measure glucose, insulin, and free fatty acids levels. SKM was harvested for immunohistochemistry (IHC). Our IHC results showed a reliable pattern of effective UTMD-based gene delivery in enhancing SKM overexpression of the UCP-1 gene. This clearly indicates that our plasmid DNA construct encoding the gene combination of PRDM16, PPARGC1A, and BMP7 reprogrammed adult SKM tissue into brown adipose cells in vivo. Our pilot established POC showing that the administration of the gene cocktail to SKM in this rat model of genetic obesity using UTMD gene therapy, engineered a BAT phenotype with UCP-1 over-expression. © 2017 IUBMB Life, 69(9):745-755, 2017.


Assuntos
Reprogramação Celular/genética , Diabetes Mellitus Experimental/terapia , Técnicas de Transferência de Genes , Terapia Genética , Obesidade/terapia , Tecido Adiposo Marrom/metabolismo , Animais , Proteína Morfogenética Óssea 7/genética , Diferenciação Celular/genética , Diabetes Mellitus Experimental/genética , Modelos Animais de Doenças , Humanos , Microbolhas/uso terapêutico , Músculo Esquelético/metabolismo , Músculo Esquelético/transplante , Obesidade/genética , Obesidade/fisiopatologia , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/genética , Plasmídeos/genética , Plasmídeos/uso terapêutico , Ratos , Ratos Zucker , Fatores de Transcrição/genética
5.
Stem Cells Transl Med ; 6(3): 877-885, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28297573

RESUMO

Optimal stem cell delivery procedures are critical to the success of the cell therapy approach. Variables such as flow rate, suspension solution, needle diameter, cell density, and tissue mechanics affect tissue penetration, backflow along the needle, and the dispersion and survival of injected cells during delivery. Most cell transplantation centers engaged in human clinical trials use custom-designed cannula needles, syringes, or catheters, sometimes precluding the use of magnetic resonance imaging (MRI)-guided delivery to target tissue. As a result, stem cell therapies may be hampered because more than 80% of grafted cells do not survive the delivery-for example, to the heart, liver/pancreas, and brain-which translates to poor patient outcomes. We developed a minimally invasive interventional MRI (iMRI) approach for intraoperatively imaging neural stem cell (NSC) delivery procedures. We used NSCs prelabeled with a contrast agent and real-time magnetic resonance imaging to guide the injection cannula to the target and to track the delivery of the cells into the putamen of baboons. We provide evidence that cell injection into the brain parenchyma follows a novel pulsatile mode of cellular discharge from the delivery catheter despite a constant infusion flow rate. The rate of cell infusion significantly affects the dispersion and viability of grafted cells. We report on our investigational use of a frameless navigation system for image-guided NSC transplantation using a straight cannula. Through submillimeter accuracy and real-time imaging, iMRI approaches may improve the safety and efficacy of neural cell transplantation therapies. Stem Cells Translational Medicine 2017;6:877-885.


Assuntos
Gânglios da Base/citologia , Imageamento por Ressonância Magnética , Células-Tronco Neurais/transplante , Transplante de Células-Tronco/métodos , Animais , Sobrevivência Celular , Sistemas Computacionais , Dextranos/química , Humanos , Nanopartículas de Magnetita/química , Células-Tronco Neurais/citologia , Papio , Imagens de Fantasmas
6.
Cell Transplant ; 25(7): 1371-80, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26132738

RESUMO

Compelling evidence suggests that transplantation of neural stem cells (NSCs) from multiple sources ameliorates motor deficits after stroke. However, it is currently unknown to what extent the electrophysiological activity of grafted NSC progeny participates in the improvement of motor deficits and whether excitatory phenotypes of the grafted cells are beneficial or deleterious to sensorimotor performances. To address this question, we used optogenetic tools to drive the excitatory outputs of the grafted NSCs and assess the impact on local circuitry and sensorimotor performance. We genetically engineered NSCs to express the Channelrhodopsin-2 (ChR2), a light-gated cation channel that evokes neuronal depolarization and initiation of action potentials with precise temporal control to light stimulation. To test the function of these cells in a stroke model, rats were subjected to an ischemic stroke and grafted with ChR2-NSCs. The grafted NSCs identified with a human-specific nuclear marker survived in the peri-infarct tissue and coexpressed the ChR2 transgene with the neuronal markers TuJ1 and NeuN. Gene expression analysis in stimulated versus vehicle-treated animals showed a differential upregulation of transcripts involved in neurotransmission, neuronal differentiation, regeneration, axonal guidance, and synaptic plasticity. Interestingly, genes involved in the inflammatory response were significantly downregulated. Behavioral analysis demonstrated that chronic optogenetic stimulation of the ChR2-NSCs enhanced forelimb use on the stroke-affected side and motor activity in an open field test. Together these data suggest that excitatory stimulation of grafted NSCs elicits beneficial effects in experimental stroke model through cell replacement and non-cell replacement, anti-inflammatory/neurotrophic effects.


Assuntos
Regulação para Baixo , Células-Tronco Neurais/transplante , Optogenética/métodos , Acidente Vascular Cerebral/terapia , Transmissão Sináptica , Animais , Separação Celular , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Células-Tronco Embrionárias Humanas/citologia , Humanos , Inflamação/complicações , Inflamação/genética , Inflamação/terapia , Masculino , Neostriado/metabolismo , Células-Tronco Neurais/citologia , Análise de Sequência com Séries de Oligonucleotídeos , Ratos Sprague-Dawley , Rodopsina/genética , Acidente Vascular Cerebral/complicações , Acidente Vascular Cerebral/genética , Transdução Genética , Transgenes
7.
Stem Cells Dev ; 23 Suppl 1: 83-7, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25457970

RESUMO

Humans and nonhuman primates (NHPs) are similar in size, behavior, physiology, biochemistry, structure and function of organs, and complexity of the immune system. Research on NHPs generates complementary data that bridge translational research from small animal models to humans. NHP models of human disease offer unique opportunities to develop stem cell-based therapeutic interventions that directly address relevant and challenging translational aspects of cell transplantation therapy. These include the use of autologous induced pluripotent stem cell-derived cellular products, issues related to the immune response in autologous and allogeneic setting, pros and cons of delivery techniques in a clinical setting, as well as the safety and efficacy of candidate cell lines. The NHP model allows the assessment of complex physiological, biochemical, behavioral, and imaging end points, with direct relevance to human conditions. At the same time, the value of using primates in scientific research must be carefully evaluated and timed due to expense and the necessity for specialized equipment and highly trained personnel. Often it is more efficient and useful to perform initial proof-of-concept studies for new therapeutics in rodents and/or other species before the pivotal studies in NHPs that may eventually lead to first-in-human trials. In this report, we present how the Southwest National Primate Research Center, one of seven NIH-funded National Primate Research Centers, may help the global community in translating promising technologies to the clinical arena.


Assuntos
Transplante de Células/métodos , Modelos Animais , Medicina Regenerativa/tendências , Pesquisa Translacional Biomédica/tendências , Animais , Financiamento Governamental , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Primatas , Desenvolvimento de Programas , Pesquisa com Células-Tronco , Texas , Estados Unidos
8.
Methods Mol Biol ; 1059: 157-67, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23934842

RESUMO

Human embryonic stem cells (hESCs) are pluripotent with a strong self-renewable ability making them a virtually unlimited source of neural cells for structural repair in neurological disorders. Currently, hESCs are one of the most promising cell sources amenable for commercialization of off-shelf cell therapy products. However, along with this strong proliferative capacity of hESCs comes the tumorigenic potential of these cells after transplantation. Thus, the isolation and purification of a homogeneous, population of neural stem cells (hNSCs) are of paramount importance to avoid tumor formation in the host brain. This chapter describes the isolation, neuralization, and long-term perpetuation of hNSCs derived from hESCs through use of specific mitogenic growth factors and the preparation of hNSCs for transplantation in an experimental model of stroke. Additionally, we describe methods to analyze the stroke and size of grafts using magnetic resonance imaging and Osirix software, and neuroanatomical tracing procedures to study axonal remodeling after stroke and cell transplantation.


Assuntos
Separação Celular , Infarto da Artéria Cerebral Média/terapia , Células-Tronco Neurais/transplante , Animais , Técnicas de Cultura de Células , Meios de Cultura , Modelos Animais de Doenças , Células-Tronco Embrionárias/fisiologia , Humanos , Ratos , Esferoides Celulares/fisiologia
9.
Cell Transplant ; 22(5): 881-92, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23044338

RESUMO

Stem cell therapy ameliorates motor deficits in experimental stroke model. Multimodal molecular imaging enables real-time longitudinal monitoring of infarct location, size, and transplant survival. In the present study, we used magnetic resonance imaging (MRI) and positron emission tomography (PET) to track the infarct evolution,tissue repair, and the fate of grafted cells. We genetically engineered embryonic stem cell-derived neural stem cells (NSCs) with a triple fusion reporter gene to express monomeric red fluorescence protein and herpes simplex virus-truncated thymidine kinase for multimodal molecular imaging and SPIO labeled for MRI. The infarct size as well as fate and function of grafted cells were tracked in real time for 3 months using MRI and PET. We report that grafted NSCs reduced the infarct size in animals with less than 0.1 cm(3) initial infarct in a dose-dependent manner, while larger stroke was not amenable to such beneficial effects. PET imaging revealed increased metabolic activity in grafted animals and visualized functioning grafted cells in vivo. Immunohistopathological analysis demonstrated that, after a 3-month survival period, grafted NSCs dispersed in the stroke-lesioned parenchyma and differentiated into neurons, astrocytes, and oligodendrocytes. Longitudinal multimodal imaging provides insights into time course dose-dependent interactions between NSC grafts and structural changes in infarcted tissue.


Assuntos
Células-Tronco Neurais/citologia , Acidente Vascular Cerebral/terapia , Animais , Diferenciação Celular , Linhagem da Célula , Células-Tronco Embrionárias/citologia , Compostos Férricos/química , Humanos , Lentivirus/genética , Imageamento por Ressonância Magnética , Masculino , Células-Tronco Neurais/transplante , Tomografia por Emissão de Pósitrons , Proteínas Tirosina Quinases/metabolismo , Radiografia , Ratos , Ratos Sprague-Dawley , Acidente Vascular Cerebral/diagnóstico por imagem , Acidente Vascular Cerebral/patologia , Transplante Heterólogo
10.
PLoS One ; 7(7): e41120, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22815935

RESUMO

The use of human embryonic stem cells (hESCs) to repair diseased or injured brain is promising technology with significant humanitarian, societal and economic impact. Parkinson's disease (PD) is a neurological disorder characterized by the loss of midbrain dopaminergic (DA) neurons. The generation of this cell type will fulfill a currently unmet therapeutic need. We report on the isolation and perpetuation of a midbrain-specified self-renewable human neural stem cell line (hNSCs) from hESCs. These hNSCs grew as a monolayer and uniformly expressed the neural precursor markers nestin, vimentin and a radial glial phenotype. We describe a process to direct the differentiation of these hNSCs towards the DA lineage. Glial conditioned media acted synergistically with fibroblastic growth factor and leukemia inhibitory factor to induce the expression of the DA marker, tyrosine hydroxylase (TH), in the hNSC progeny. The glial-derived neurotrophic factor did not fully mimic the effects of conditioned media. The hNSCs expressed the midbrain-specific transcription factors Nurr1 and Pitx3. The inductive effects did not modify the level of the glutamic acid decarboxylase (GAD) transcript, a marker for GABAergic neurons, while the TH transcript increased 10-fold. Immunocytochemical analysis demonstrated that the TH-expressing cells did not co-localize with GAD. The transplantation of these DA-induced hNSCs into the non-human primate MPTP model of PD demonstrated that the cells maintain their DA-induced phenotype, extend neurite outgrowths and express synaptic markers.


Assuntos
Neurônios Dopaminérgicos/metabolismo , Células-Tronco Embrionárias/citologia , Mesencéfalo/metabolismo , Células-Tronco Neurais/citologia , Doença de Parkinson/metabolismo , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/farmacologia , Animais , Transplante de Células , Modelos Animais de Doenças , Eletrofisiologia/métodos , Fatores de Crescimento de Fibroblastos/metabolismo , Glutamato Descarboxilase/metabolismo , Haplorrinos , Humanos , Masculino , Neurônios/metabolismo , Fenótipo , Sinapses
11.
Cell Stem Cell ; 6(3): 238-50, 2010 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-20207227

RESUMO

We investigated a strategy to ameliorate the motor symptoms of rats that received 6-hydroxydopamine (6-OHDA) lesions, a rodent model of Parkinson's disease, through transplantation of embryonic medial ganglionic eminence (MGE) cells into the striatum. During brain development, embryonic MGE cells migrate into the striatum and neocortex where they mature into GABAergic interneurons and play a key role in establishing the balance between excitation and inhibition. Unlike most other embryonic neurons, MGE cells retain the capacity for migration and integration when transplanted into the postnatal and adult brain. We performed MGE cell transplantation into the basal ganglia of control and 6-OHDA-lesioned rats. Transplanted MGE cells survived, differentiated into GABA(+) neurons, integrated into host circuitry, and modified motor behavior in both lesioned and control rats. Our data suggest that MGE cell transplantation into the striatum is a promising approach to investigate the potential benefits of remodeling basal ganglia circuitry in neurodegenerative diseases.


Assuntos
Envelhecimento , Corpo Estriado/citologia , Corpo Estriado/metabolismo , Eminência Mediana/citologia , Eminência Mediana/metabolismo , Atividade Motora , Oxidopamina/metabolismo , Animais , Comportamento Animal , Diferenciação Celular , Movimento Celular , Sobrevivência Celular , Transplante de Células , Corpo Estriado/cirurgia , Feminino , Eminência Mediana/embriologia , Ratos , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/metabolismo , Ácido gama-Aminobutírico/metabolismo
12.
Stroke ; 41(3): 516-23, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20075340

RESUMO

BACKGROUND AND PURPOSE: Hypoxic-ischemic (HI) brain injury in newborn infants represents a major cause of cerebral palsy, development delay, and epilepsy. Stem cell-based therapy has the potential to rescue and replace the ischemic tissue caused by HI and to restore function. However, the mechanisms by which stem cell transplants induce functional recovery are yet to be elucidated. In the present study, we sought to investigate the efficacy of human neural stem cells derived from human embryonic stem cells in a rat model of neonatal HI and the mechanisms enhancing brain repair. METHODS: The human neural stem cells were genetically engineered for in vivo molecular imaging and for postmortem histological tracking. Twenty-four hours after the induction of HI, animals were grafted with human neural stem cells into the forebrain. Motor behavioral tests were performed the fourth week after transplantation. We used immunocytochemistry and neuroanatomical tracing to analyze neural differentiation, axonal sprouting, and microglia response. Treatment-induced changes in gene expression were investigated by microarray and quantitative polymerase chain reaction. RESULTS: Bioluminescence imaging permitted real time longitudinal tracking of grafted human neural stem cells. HI transplanted animals significantly improved in their use of the contralateral impeded forelimb and in the Rotorod test. The grafts showed good survival, dispersion, and differentiation. We observed an increase of uniformly distributed microglia cells in the grafted side. Anterograde neuroanatomical tracing demonstrated significant contralesional sprouting. Microarray analysis revealed upregulation of genes involved in neurogenesis, gliogenesis, and neurotrophic support. CONCLUSIONS: These results suggest that human neural stem cell transplants enhance endogenous brain repair through multiple modalities in response to HI.


Assuntos
Axônios/fisiologia , Células-Tronco Embrionárias/transplante , Hipóxia-Isquemia Encefálica/cirurgia , Microglia/fisiologia , Neurônios/transplante , Transplante de Células-Tronco , Animais , Animais Recém-Nascidos , Humanos , Hipóxia-Isquemia Encefálica/patologia , Plasticidade Neuronal/fisiologia , Ratos , Ratos Sprague-Dawley , Transplante de Células-Tronco/métodos , Transplantes
14.
Cell Transplant ; 18(7): 815-26, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19500468

RESUMO

Currently there are no effective treatments targeting residual anatomical and behavioral deficits resulting from stroke. Evidence suggests that cell transplantation therapy may enhance functional recovery after stroke through multiple mechanisms. We used a syngeneic model of neural transplantation to explore graft-host communications that enhance cellular engraftment.The medial ganglionic eminence (MGE) cells were derived from 15-day-old transgenic rat embryos carrying green fluorescent protein (GFP), a marker, to easily track the transplanted cells. Adult rats were subjected to transient intraluminal occlusion of the medial cerebral artery. Two weeks after stroke, the grafts were deposited into four sites, along the rostro-caudal axis and medially to the stroke in the penumbra zone. Control groups included vehicle and fibroblast transplants. Animals were subjected to motor behavioral tests at 4 week posttransplant survival time. Morphological analysis demonstrated that the grafted MGE cells differentiated into multiple neuronal subtypes, established synaptic contact with host cells, increased the expression of synaptic markers, and enhanced axonal reorganization in the injured area. Initial patch-clamp recording demonstrated that the MGE cells received postsynaptic currents from host cells. Behavioral analysis showed reduced motor deficits in the rotarod and elevated body swing tests. These findings suggest that graft-host interactions influence the fate of grafted neural precursors and that functional recovery could be mediated by neurotrophic support, new synaptic circuit elaboration, and enhancement of the stroke-induced neuroplasticity.


Assuntos
Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/transplante , Infarto da Artéria Cerebral Média/terapia , Neurônios/patologia , Animais , Comunicação Celular/fisiologia , Modelos Animais de Doenças , Atividade Motora , Neurogênese , Plasticidade Neuronal , Ratos , Teste de Desempenho do Rota-Rod , Sinapses/fisiologia , Potenciais Sinápticos , Telencéfalo/citologia , Transplante Isogênico
15.
Mol Ther ; 17(7): 1282-91, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19436269

RESUMO

Real-time imaging of transplanted stem cells is essential for understanding their interactions in vivo with host environments, for tracking cell fate and function and for successful delivery and safety monitoring in the clinical setting. In this study, we used bioluminescence (BLI) and magnetic resonance imaging (MRI) to visualize the fate of grafted human embryonic stem cell (hESC)-derived human neural stem cells (hNSCs) in stroke-damaged rat brain. The hNSCs were genetically engineered with a lentiviral vector carrying a double fusion (DF) reporter gene that stably expressed enhanced green fluorescence protein (eGFP) and firefly luciferase (fLuc) reporter genes. The hNSCs were self-renewable, multipotent, and expressed markers for neural stem cells. Cell survival was tracked noninvasively by MRI and BLI for 2 months after transplantation and confirmed histologically. Electrophysiological recording from grafted GFP(+) cells and immuno-electronmicroscopy demonstrated connectivity. Grafted hNSCs differentiated into neurons, into oligodendrocytes in stroke regions undergoing remyelination and into astrocytes extending processes toward stroke-damaged vasculatures. Our data suggest that the combination of BLI and MRI modalities provides reliable real-time monitoring of cell fate.


Assuntos
Isquemia Encefálica/patologia , Isquemia Encefálica/terapia , Células-Tronco Embrionárias/citologia , Imageamento por Ressonância Magnética/métodos , Transplante de Células-Tronco/métodos , Animais , Linhagem Celular , Células-Tronco Embrionárias/fisiologia , Citometria de Fluxo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Imuno-Histoquímica , Luciferases de Vaga-Lume/genética , Luciferases de Vaga-Lume/metabolismo , Masculino , Técnicas de Patch-Clamp , Ratos , Ratos Sprague-Dawley
16.
Regen Med ; 4(2): 251-63, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19317644

RESUMO

Demographic trends, particularly those related to longer life expectancy, suggest that the demand for tissue and organ transplants will further increase since many disorders result from degeneration, injury or organ failure. The most urgent problem in transplantation medicine is the shortage or lack of suitable donor organs and tissue, leading to ethical and societal problems such as organ trafficking. The discovery of stem cells in the inner cell mass of developing embryos and in adult tissue has revolutionized the medical field by introducing new therapeutic dimensions to consider for previously untreatable diseases and injuries. The unlimited self-renewal ability and pluripotent capacity to become any cell type of the organism make human embryonic stem cells (hESCs) a compelling source of cells to study tissue histogenesis and to apply in a wide array of tissue engineering, cell transplantation therapy and drug discovery applications. In this article, we will focus on hESCs and address the derivation of therapeutic neural stem cell lines from hESCs, as well as the biological and regulatory aspects to developing a safe cellular product for stroke cell therapy.


Assuntos
Transplante de Células/métodos , Células-Tronco Embrionárias/citologia , Neurônios/citologia , Acidente Vascular Cerebral/terapia , Técnicas de Cultura de Células , Diferenciação Celular , Humanos
17.
Methods Mol Biol ; 438: 205-12, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18369760

RESUMO

Parkinson's disease (PD) is a neurodegenerative disorder characterized by the loss of dopaminergic neurons in the substantia nigra. Symptoms include tremors, rigidity, bradykinesia, and instability. Neural transplantation is a promising strategy for improving dopaminergic dysfunction in PD provided that the dopaminergic neurons are consistently generated from a renewable source of cells. Neural stem cells (NSCs) have the ability to self-renew, generate a large number of progeny, and differentiate into the principal nervous system cell types. As such, stem cells provide an exciting opportunity to understand the basic mechanisms involved in cell differentiation and histogenesis. These mechanisms have translational applications in tissue engineering and biomedicine in general. In vitro differentiation assays are important in cell characterization, in assaying for novel instructive molecules, and in generating specific cell types. We describe differentiation techniques to test NSCs for multipotency and to induce the dopaminergic phenotype in neural stem cell progeny by coculturing them with astrocytes and treating them with conditioned media and basic fibroblastic growth factor.


Assuntos
Bioensaio/métodos , Diferenciação Celular , Dopamina/metabolismo , Neurônios/citologia , Células-Tronco/citologia , Animais , Separação Celular , Técnicas de Cocultura , Camundongos , Fenótipo
18.
PLoS One ; 3(2): e1644, 2008 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-18286199

RESUMO

BACKGROUND: Human embryonic stem cells (hESCs) offer a virtually unlimited source of neural cells for structural repair in neurological disorders, such as stroke. Neural cells can be derived from hESCs either by direct enrichment, or by isolating specific growth factor-responsive and expandable populations of human neural stem cells (hNSCs). Studies have indicated that the direct enrichment method generates a heterogeneous population of cells that may contain residual undifferentiated stem cells that could lead to tumor formation in vivo. METHODS/PRINCIPAL FINDINGS: We isolated an expandable and homogenous population of hNSCs (named SD56) from hESCs using a defined media supplemented with epidermal growth factor (EGF), basic fibroblast growth factor (bFGF) and leukemia inhibitory growth factor (LIF). These hNSCs grew as an adherent monolayer culture. They were fully neuralized and uniformly expressed molecular features of NSCs, including nestin, vimentin and radial glial markers. These hNSCs did not express the pluripotency markers Oct4 or Nanog, nor did they express markers for the mesoderm or endoderm lineages. The self-renewal property of the hNSCs was characterized by a predominant symmetrical mode of cell division. The SD56 hNSCs differentiated into neurons, astrocytes and oligodendrocytes throughout multiple passages in vitro, as well as after transplantation. Together, these criteria confirm the definitive NSC identity of the SD56 cell line. Importantly, they exhibited no chromosome abnormalities and did not form tumors after implantation into rat ischemic brains and into naïve nude rat brains and flanks. Furthermore, hNSCs isolated under these conditions migrated toward the ischemia-injured adult brain parenchyma and improved the independent use of the stroke-impaired forelimb two months post-transplantation. CONCLUSIONS/SIGNIFICANCE: The SD56 human neural stem cells derived under the reported conditions are stable, do not form tumors in vivo and enable functional recovery after stroke. These properties indicate that this hNSC line may offer a renewable, homogenous source of neural cells that will be valuable for basic and translational research.


Assuntos
Células-Tronco Embrionárias/citologia , Neurônios/citologia , Transplante de Células-Tronco , Acidente Vascular Cerebral/terapia , Animais , Biomarcadores/análise , Técnicas de Cultura de Células , Diferenciação Celular , Divisão Celular , Linhagem Celular , Movimento Celular , Modelos Animais de Doenças , Humanos , Neurônios/transplante , Ratos , Células-Tronco , Acidente Vascular Cerebral/patologia , Transplante Heterólogo
19.
Neuroreport ; 17(2): 201-4, 2006 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-16407771

RESUMO

The present report describes for the first time, the stability of recombinant adeno-associated virus serotype 2 (AAV2) human aromatic L-amino acid decarboxylase (hAADC) gene transfer after 3-year survival time in a non-human primate model of Parkinson's disease. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine-lesioned monkeys were treated with six injections of 30 microl/site of AAV2-hAADC at a concentration of 2 x 10(12) vg/ml into the caudate and putamen. Stereological analysis revealed a 46.6% increase in the total number of AAV2-hAADC-transduced cells in the striatum between 8 weeks and 3 years after gene transfer survival time. In the 8-week animals, the distribution of the AADC+ cells was dispersed and heterogeneous, whereas in the 3-year animals it was widespread and homogenous. Confocal analysis demonstrated that approximately 85% of the AADC+ cells were neuronal nuclei immunoreactive.


Assuntos
Descarboxilases de Aminoácido-L-Aromático/uso terapêutico , Núcleo Caudado/patologia , Neurônios/metabolismo , Transtornos Parkinsonianos/patologia , Transdução Genética/métodos , Animais , Descarboxilases de Aminoácido-L-Aromático/genética , Descarboxilases de Aminoácido-L-Aromático/metabolismo , Núcleo Caudado/fisiopatologia , Contagem de Células/métodos , Dependovirus/genética , Modelos Animais de Doenças , Humanos , Imuno-Histoquímica/métodos , Macaca mulatta , Masculino , Transtornos Parkinsonianos/terapia , Fosfopiruvato Hidratase/metabolismo , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA