Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Stem Cells ; 38(8): 936-947, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32374064

RESUMEN

Huntington's disease (HD) is a devastating, autosomal-dominant neurodegenerative disease, for which there are currently no disease-modifying therapies. Clinical trials to replace the damaged striatal medium spiny neurons (MSNs) have been attempted in the past two decades but have met with only limited success. In this study, we investigated whether a clonal, conditionally immortalized neural stem cell line (CTX0E03), which has already shown safety and signals of efficacy in chronic ischemic stroke patients, could rescue deficits seen in an animal model of HD. After CTX0E03 transplantation into the quinolinic acid-lesioned rat model of HD, behavioral changes were measured using the rotarod, stepping, and staircase tests. In vivo differentiation and neuronal connections of the transplanted CTX0E03 cells were evaluated with immunohistochemical staining and retrograde tracing with Fluoro-Gold. We found that transplantation of CTX0E03 gave rise to a significant behavioral improvement compared with the sham- or fibroblast-transplanted group. Transplanted CTX0E03 formed MSNs (DARPP-32) and GABAergic neurons (GABA, GAD65/67) with BDNF expression in the striatum, while cortically transplanted cells formed Tbr1-positive neurons. Using a retrograde label, we also found stable engraftment and connection of the transplanted cells with host brain tissues. CTX0E03 transplantation also reduced glial scar formation and inflammation, as well as increasing endogenous neurogenesis and angiogenesis. Overall, our results demonstrate that CTX0E03, a clinical-grade neural stem cell line, is effective for preclinical test in HD, and, therefore, will be useful for clinical development in the treatment of HD patients.


Asunto(s)
Enfermedad de Huntington/metabolismo , Células-Madre Neurales/metabolismo , Ácido Quinolínico/metabolismo , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Humanos , Ratones , Clasificación del Tumor
2.
Cytopathology ; 30(6): 620-627, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31461195

RESUMEN

OBJECTIVE: The study set out to assess the feasibility of using ParsortixTM circulating tumour cell (CTC) extraction and CytoFoam Disc cell-block immunohistochemistry to diagnose metastatic carcinoma from blood samples in a National Health Service district general hospital. METHODS: Blood samples were taken from 50 patients with metastatic carcinoma and 50 healthy volunteers and processed, using a previously published method, to extract CTCs and collect them in a cell-block for routine formalin-fixed paraffin sectioning and immunohistochemistry. The extracted cells were compared with the patients' routine diagnostic samples. RESULTS: The samples from the 50 carcinoma patients showed cytokeratin-positive cells in 19 cases. In eight of these, the cytokeratin-positive cells had a similar immunoprofile to the carcinoma in the conventional biopsy or cytology specimen. Some carcinoma patients also had circulating cytokeratin-positive cells that were probably benign epithelial cells and circulating megakaryocytes. Both of these types of cells were also found in healthy volunteers. Processing and initial examination could be completed in 2 days. The full processing cost was approximately £316 per case. CONCLUSIONS: CTCs could be extracted from the blood of some patients with metastatic carcinoma and formed into a formalin-fixed cell-block for routine paraffin processing and immunohistochemistry. The specificity of this approach is constrained by the observation that some patients with metastatic carcinoma had circulating cytokeratin-positive cells that were probably benign, and these were also found in healthy volunteers. Circulating megakaryocytes were present in carcinoma patients and healthy volunteers.


Asunto(s)
Carcinoma/sangre , ADN Tumoral Circulante/sangre , Citodiagnóstico , Neoplasias/sangre , Carcinoma/genética , Carcinoma/patología , Diagnóstico Diferencial , Femenino , Humanos , Masculino , Megacariocitos/patología , Neoplasias/genética , Neoplasias/patología , Células Neoplásicas Circulantes/patología
4.
Tissue Eng Part C Methods ; 23(10): 575-582, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28877649

RESUMEN

This study explored different approaches to preserve engineered neural tissue (EngNT), a stabilized, cellular collagen hydrogel containing columns of aligned Schwann cells for nervous system repair. The ability to preserve EngNT without disrupting cellular and extracellular components and structures is important for clinical translation and commercialization. Stabilized cellular gels and EngNT constructs were preserved under various conditions and cell survival assessed using live/dead microscopy and metabolic assay. Optimal survival was recorded in hypothermic (4°C) conditions for 2-3 days using Hibernate®-A media and, for longer-term cryogenic storage (liquid nitrogen), using a mixture of 60% Dulbecco's modified Eagle's medium, 30% fetal bovine serum, and 10% dimethyl sulfoxide. Functionality and structure of preserved EngNT were assessed in coculture with dorsal root ganglion neurons, which indicated that alignment of Schwann cells and the ability of EngNT to support and guide neuronal regeneration were not disrupted. The identification of conditions that preserve EngNT will inform development of storage and transport methodologies to support clinical and commercial translation of this technology and other therapies based on cellular hydrogels.


Asunto(s)
Criopreservación , Hipotermia Inducida , Tejido Nervioso/fisiología , Ingeniería de Tejidos/métodos , Animales , Bovinos , Muerte Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Colágeno/farmacología , Geles , Tejido Nervioso/efectos de los fármacos , Neuritas/efectos de los fármacos , Neuritas/metabolismo , Ratas Wistar , Células de Schwann/citología , Células de Schwann/efectos de los fármacos , Células de Schwann/metabolismo , Células de Schwann/ultraestructura
5.
Transl Vis Sci Technol ; 5(4): 6, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27486556

RESUMEN

PURPOSE: We assessed the long-term efficacy and safety of human retinal progenitor cells (hRPC) using established rodent models. METHODS: Efficacy of hRPC was tested initially in Royal College of Surgeons (RCS) dystrophic rats immunosuppressed with cyclosporine/dexamethasone. Due to adverse effects of dexamethasone, this drug was omitted from a subsequent dose-ranging study, where different hRPC doses were tested for their ability to preserve visual function (measured by optokinetic head tracking) and retinal structure in RCS rats at 3 to 6 months after grafting. Safety of hRPC was assessed by subretinal transplantation into wild type (WT) rats and NIH-III nude mice, with analysis at 3 to 6 and 9 months after grafting, respectively. RESULTS: The optimal dose of hRPC for preserving visual function/retinal structure in dystrophic rats was 50,000 to 100,000 cells. Human retinal progenitor cells integrated/survived in dystrophic and WT rat retina up to 6 months after grafting and expressed nestin, vimentin, GFAP, and ßIII tubulin. Vision and retinal structure remained normal in WT rats injected with hRPC and there was no evidence of tumors. A comparison between dexamethasone-treated and untreated dystrophic rats at 3 months after grafting revealed an unexpected reduction in the baseline visual acuity of dexamethasone-treated animals. CONCLUSIONS: Human retinal progenitor cells appear safe and efficacious in the preclinical models used here. TRANSLATIONAL RELEVANCE: Human retinal progenitor cells could be deployed during early stages of retinal degeneration or in regions of intact retina, without adverse effects on visual function. The ability of dexamethasone to reduce baseline visual acuity in RCS dystrophic rats has important implications for the interpretation of preclinical and clinical cell transplant studies.

6.
PLoS One ; 11(1): e0146353, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26752061

RESUMEN

Exosomes are small (30-100 nm) membrane vesicles secreted by a variety of cell types and only recently have emerged as a new avenue for cell-to-cell communication. They are natural shuttles of RNA and protein cargo, making them attractive as potential therapeutic delivery vehicles. MicroRNAs (miRNAs) are short non-coding RNAs which regulate biological processes and can be found in exosomes. Here we characterized the miRNA contents of exosomes derived from human neural stem cells (hNSCs). Our investigated hNSC line is a clonal, conditionally immortalized cell line, compliant with good manufacturing practice (GMP), and in clinical trials for stroke and critical limb ischemia in the UK (clinicaltrials.gov: NCT01151124, NCT02117635, and NCT01916369). By using next generation sequencing (NGS) technology we identified the presence of a variety of miRNAs in both exosomal and cellular preparations. Many of these miRNAs were enriched in exosomes indicating that cells specifically sort them for extracellular release. Although exosomes have been proven to contain miRNAs, the copy number quantification per exosome of a given miRNA remains unclear. Herein we quantified by real-time PCR a highly shuttled exosomal miRNA subtype (hsa-miR-1246) in order to assess its stoichiometry per exosome. Furthermore, we utilized an in vitro system to confirm its functional transfer by measuring the reduction in luciferase expression using a 3' untranslated region dual luciferase reporter assay. In summary, NGS analysis allowed the identification of a unique set of hNSC derived exosomal miRNAs. Stoichiometry and functional transfer analysis of one of the most abundant identified miRNA, hsa-miR-1246, were measured to support biological relevance of exosomal miRNA delivery.


Asunto(s)
Exosomas/metabolismo , MicroARNs/genética , Células-Madre Neurales/metabolismo , Células HeLa , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , MicroARNs/metabolismo
7.
Am J Physiol Endocrinol Metab ; 309(6): E511-22, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26126685

RESUMEN

The development of hepatocyte cell models that represent fatty acid partitioning within the human liver would be beneficial for the study of the development and progression of nonalcoholic fatty liver disease (NAFLD). We sought to develop and characterize a novel human liver cell line (LIV0APOLY) to establish a model of lipid accumulation using a physiological mixture of fatty acids under low- and high-glucose conditions. LIV0APOLY cells were compared with a well-established cell line (HepG2) and, where possible, primary human hepatocytes. LIV0APOLY cells were found to proliferate and express some mature liver markers and were wild type for the PNPLA3 (rs738409) gene, whereas HepG2 cells carried the Ile(148)Met variant that is positively associated with liver fat content. Intracellular triglyceride content was higher in HepG2 than in LIV0APOLY cells; exposure to high glucose and/or exogenous fatty acids increased intracellular triglyceride in both cell lines. Triglyceride concentrations in media were higher from LIV0APOLY compared with HepG2 cells. Culturing LIV0APOLY cells in high glucose increased a marker of endoplasmic reticulum stress and attenuated insulin-stimulated Akt phosphorylation whereas low glucose and exogenous fatty acids increased AMPK phosphorylation. Although LIV0APOLY cells and primary hepatocytes stored similar amounts of exogenous fatty acids as triglyceride, more exogenous fatty acids were partitioned toward oxidation in the LIV0APOLY cells than in primary hepatocytes. LIV0APOLY cells offer the potential to be a renewable cellular model for studying the effects of exogenous metabolic substrates on fatty acid partitioning; however, their usefulness as a model of lipoprotein metabolism needs to be further explored.


Asunto(s)
Ácidos Grasos/metabolismo , Glucosa/metabolismo , Hepatocitos/metabolismo , Metabolismo de los Lípidos , Hígado/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Triglicéridos/metabolismo , Línea Celular , Estrés del Retículo Endoplásmico , Células Hep G2 , Humanos , Insulina/metabolismo , Lipasa/genética , Proteínas de la Membrana/genética , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo
8.
J Vis Exp ; (98)2015 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-25938519

RESUMEN

Neural stem cells (NSCs) are capable of self-renewal and differentiation into neurons, astrocytes and oligodendrocytes under specific local microenvironments. In here, we present a set of methods used for three dimensional (3D) differentiation and miRNA analysis of a clonal human neural stem cell (hNSC) line, currently in clinical trials for stroke disability (NCT01151124 and NCT02117635, Clinicaltrials.gov). HNSCs were derived from an ethical approved first trimester human fetal cortex and conditionally immortalized using retroviral integration of a single copy of the c-mycER(TAM)construct. We describe how to measure axon process outgrowth of hNSCs differentiated on 3D scaffolds and how to quantify associated changes in miRNA expression using PCR array. Furthermore we exemplify computational analysis with the aim of selecting miRNA putative targets. SOX5 and NR4A3 were identified as suitable miRNA putative target of selected significantly down-regulated miRNAs in differentiated hNSC. MiRNA target validation was performed on SOX5 and NR4A3 3'UTRs by dual reporter plasmid transfection and dual luciferase assay.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , MicroARNs/análisis , Células-Madre Neurales/citología , Diferenciación Celular/fisiología , Línea Celular , Humanos , MicroARNs/biosíntesis , MicroARNs/genética , Células-Madre Neurales/química , Neuronas/química , Neuronas/citología , Oligodendroglía/química , Oligodendroglía/citología , Transfección
9.
Stem Cell Res Ther ; 5(2): 49, 2014 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-24725992

RESUMEN

INTRODUCTION: Stem cells have the ability to self-renew or to differentiate into numerous cell types; however, our understanding of how to control and exploit this potential is currently limited. An emerging hypothesis is that microRNAs (miRNAs) play a central role in controlling stem cell-fate determination. Herein, we have characterized the effects of miRNAs in differentiated human neural stem cells (hNSCs) by using a cell line currently being tested in clinical trials for stroke disability (NCT01151124, Clinicaltrials.gov). METHODS: HNSCs were differentiated on 2- (2D) and 3-dimensional (3D) cultures for 1 and 3 weeks. Quantification of hNSC differentiation was measured with real-time PCR and axon outgrowth. The miRNA PCR arrays were implemented to investigate differential expression profiles in differentiated hNSCs. Evaluation of miRNA effects on hNSCs was performed by using transfection of miRNA mimics, real-time PCR, Western blot, and immunocytochemistry. RESULTS: The 3D substrate promoted enhanced hNSC differentiation coupled with a loss of cell proliferation. Differentiated hNSCs exhibited a similar miRNA profiling. However, in 3D samples, the degree and timing of regulation were significantly different in miRNA members of cluster mi-R17 and miR-96-182, and hsa-miR-302a. Overall, hNSC 3D cultures demonstrated differential regulation of miRNAs involved in hNSC stemness, cell proliferation, and differentiation. The miRNA mimic analysis of hsa-miR-146b-5p and hsa-miR-99a confirmed induction of lineage-committed progenitors. Downregulated miRNAs were more abundant; those most significantly downregulated were selected, and their putative target mRNAs analyzed with the aim of unraveling their functionality. In differentiated hNSCs, downregulated hsa-miR-96 correlated with SOX5 upregulation of gene and protein expression; similar results were obtained for hsa-miR-302a, hsa-miR-182, hsa-miR-7, hsa-miR-20a/b, and hsa-miR-17 and their target NR4A3. Moreover, SOX5 was identified as a direct target gene of hsa-miR-96, and NR43A, a direct target of hsa-miR-7 and hsa-mir-17 by luciferase reporter assays. Therefore, the regulatory role of these miRNAs may occur through targeting NR4A3 and SOX5, both reported as modulators of cell-cycle progression and axon length. CONCLUSIONS: The results provide new insight into the identification of specific miRNAs implicated in hNSC differentiation. These strategies may be exploited to optimize in vitro hNSC differentiation potential for use in preclinical studies and future clinical applications.


Asunto(s)
MicroARNs/genética , MicroARNs/metabolismo , Células-Madre Neurales/fisiología , Técnicas de Cultivo de Célula , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Perfilación de la Expresión Génica , Humanos , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Accidente Cerebrovascular/genética , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/patología , Transfección
10.
Arterioscler Thromb Vasc Biol ; 34(2): 408-18, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24202301

RESUMEN

OBJECTIVE: CTX0E03 (CTX) is a clinical-grade human neural stem cell (hNSC) line that promotes angiogenesis and neurogenesis in a preclinical model of stroke and is now under clinical development for stroke disability. We evaluated the therapeutic activity of intramuscular CTX hNSC implantation in murine models of hindlimb ischemia for potential translation to clinical studies in critical limb ischemia. APPROACH AND RESULTS: Immunodeficient (CD-1 Fox(nu/nu)) mice acutely treated with hNSCs had overall significantly increased rates and magnitude of recovery of surface blood flow (laser Doppler), limb muscle perfusion (fluorescent microspheres, P<0.001), and capillary and small arteriole densities in the ischemic limb (fluorescence immunohistochemistry, both P<0.001) when compared with the vehicle-treated group. Hemodynamic and anatomic improvements were dose related and optimal at a minimum dose of 3×10(5) cells. Dose-dependent improvements in blood flow and increased vessel densities by hNSC administration early after ischemia were confirmed in immunocompetent CD-1 and streptozotocin-induced diabetic mice, together with marked reductions in the incidence of necrotic toes (P<0.05). Delayed administration of hNSCs, 7 days after occlusion, produced restorative effects when comparable with acute treatment of 35 days after hindlimb ischemia. Histological studies in hindlimb ischemia immunocompetent mice for the first 7 days after treatment revealed short-term hNSC survival, transient elevation of early host muscle inflammatory, and angiogenic responses and acceleration of myogenesis. CONCLUSIONS: hNSC therapy represents a promising treatment option for critical limb ischemia.


Asunto(s)
Pie Diabético/cirugía , Isquemia/cirugía , Músculo Esquelético/irrigación sanguínea , Neovascularización Fisiológica , Células-Madre Neurales/trasplante , Animales , Arteriolas/fisiopatología , Velocidad del Flujo Sanguíneo , Capilares/fisiopatología , Línea Celular , Supervivencia Celular , Pie Diabético/inmunología , Pie Diabético/fisiopatología , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Miembro Posterior , Humanos , Inmunocompetencia , Isquemia/genética , Isquemia/inmunología , Isquemia/fisiopatología , Flujometría por Láser-Doppler , Ratones , Ratones Noqueados , Ratones Desnudos , Células-Madre Neurales/inmunología , Flujo Sanguíneo Regional , Factores de Tiempo
11.
Cell Transplant ; 22(9): 1541-52, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23067568

RESUMEN

CTX0E03 is a human neural stem cell line previously reported to reduce sensory motor deficits in a middle cerebral artery occlusion (MCAo) model of stroke. The objective of this study was to investigate if CTX0E03 treatment promotes angiogenesis. As stroke leads to damage of the vasculature in the brain, angiogenesis may contribute to the functional recovery. To test this hypothesis, the angiogenic activity of CTX0E03 was assessed both in vitro and in vivo. In vitro, CTX0E03 expression of trophic and proangiogenic factors was determined by real-time RT-PCR, Western blot, and ELISA, and its angiogenic activity was investigated in well-established angiogenesis assays. In vivo, angiogenesis was investigated in naive mice and MCAo rat brain and was evaluated by immunohistochemistry (IHC) using Von Willebrand factor (VWF), a marker of blood vessel formation, and BrdU/CD31 double labeling in naive mice only. In vitro results showed that CTX0E03-conditioned medium and coculture significantly increased total tubule formation compared with controls (p=0.002 and p=0.0008, respectively). Furthermore, CTX0E03 cells were found to be in direct association with the tubules by ICC. In vivo CTX0E03-treated brains demonstrated a significant increase in areas occupied by VWF-positive microvessels compared with vehicle-treated naive mice (two-way ANOVA, Interaction p<0.05, Treatment p<0.0001, Time p<0.0) and MCAo rat (p=0.001 unpaired t test, Welch's correction). CTX0E03-treated naive mouse brains showed an increase in BrdU/CD31 colabeling. In conclusion, in vitro CTX0E03 cells express proangiogenic factors and may promote angiogenesis by both release of paracrine factors and direct physical interaction. Furthermore, in vivo CTX0E03-treated rodent brains exhibited a significant increase in microvessels at the site of implantation compared with vehicle-injected groups. Taken together these data suggest that CTX0E03 cell therapy may provide significant benefit to stroke patients through upregulation of angiogenesis in the ischemic brain.


Asunto(s)
Células-Madre Neurales/fisiología , Neuronas/fisiología , Trasplante de Células Madre/métodos , Proteínas Angiogénicas/biosíntesis , Animales , Western Blotting , Diferenciación Celular/fisiología , Línea Celular , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Células Endoteliales de la Vena Umbilical Humana , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Neovascularización Fisiológica/fisiología , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neurogénesis/fisiología , Neuronas/citología , Neuronas/metabolismo , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa
12.
Discov Med ; 14(78): 327-33, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23200064

RESUMEN

Metastasis is one of the cancer hallmarks described by Hanahan and Weinberg. Emerging evidence shows that it requires interplays between cancer cells and micro-environmental biofactors. Indoleamine 3,5-dioxygenase-1 (IDO-1) produced by cancer, local lymph nodes, and satellite cells have been demonstrated as one of the biofactors. Aberrant IDO-1 activity has partially contributed to immunosuppressive environment by repressing T lymphocyte and natural killer cell activities, and activating regulatory T cells (Treg, CD4+CD25+). Clinical investigations further show a negative correlation between the enzyme activity and prognosis in patients with various cancer types. The findings suggest a possible role of IDO-1 inhibitor in restoring host anti-tumor immunity and attenuating cancer metastasis. Data from preclinical and phase I/II clinical studies with IDO-1 inhibitors support this hypothesis. Polyphenols as antioxidants are shown to exhibit anticancer activities. However, the underlying mechanism has not been entirely characterized. We recently found that certain flavone molecules profoundly inhibit the enzymatic activity of IDO-1 but not mRNA expression in human neuronal stem cells (hNSC) confirmed by cell-based assay and qRT-PCR. To further the investigation, we studied additional anti-cancer phytochemicals including chalcone, flavonol, isoflavone, and diterpene. Here we summarize the results and show that the inhibitory sensitivity depends on the molecular structure in the following order: apigenin > wogonin > chrysin > biacalein ~ genistein > quercetin. Curcumin and isoliquiritigenin (a chalcone) exhibited toxicity to hNSCs. Although oridonin (a diterpene) showed a null toxicity toward hNSCs, it repressed the enzymatic function only marginally in contrast to its potent cytotoxicity in various cancer cell lines. While the mode of action of the enzyme-polyphenol complex awaits to be investigated, the sensitivity of enzyme inhibition was compared to the anti-proliferative activities toward three cancer cell lines. The IC50s obtained from both sets of the experiments indicate that they are in the vicinity of micromolar concentration with the enzyme inhibition slightly more active. These results suggest that attenuation of immune suppression via inhibition of IDO-1 enzyme activity may be one of the important mechanisms of polyphenols in chemoprevention or combinatorial cancer therapy.


Asunto(s)
Dioxigenasas/metabolismo , Inmunomodulación/efectos de los fármacos , Polifenoles/farmacología , Apigenina/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Quimioprevención , Flavanonas/farmacología , Genisteína/farmacología , Humanos , Quercetina/farmacología
13.
Stem Cells ; 30(4): 785-96, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22213183

RESUMEN

Stroke remains one of the most promising targets for cell therapy. Thorough preclinical efficacy testing of human neural stem cell (hNSC) lines in a rat model of stroke (transient middle cerebral artery occlusion) is, however, required for translation into a clinical setting. Magnetic resonance imaging (MRI) here confirmed stroke damage and allowed the targeted injection of 450,000 hNSCs (CTX0E03) into peri-infarct tissue, rather than the lesion cyst. Intraparenchymal cell implants improved sensorimotor dysfunctions (bilateral asymmetry test) and motor deficits (footfault test and rotameter). Importantly, analyses based on lesion topology (striatal vs. striatal + cortical damage) revealed a more significant improvement in animals with a stroke confined to the striatum. However, no improvement in learning and memory (water maze) was evident. An intracerebroventricular injection of cells did not result in any improvement. MRI-based lesion, striatal and cortical volumes were unchanged in treated animals compared to those with stroke that received an intraparenchymal injection of suspension vehicle. Grafted cells only survived after intraparenchymal injection with a striatal + cortical topology resulting in better graft survival (16,026 cells) than in animals with smaller striatal lesions (2,374 cells). Almost 20% of cells differentiated into glial fibrillary acidic protein+ astrocytes, but <2% turned into FOX3+ neurons. These results indicate that CTX0E03 implants robustly recover behavioral dysfunction over a 3-month time frame and that this effect is specific to their site of implantation. Lesion topology is potentially an important factor in the recovery, with a stroke confined to the striatum showing a better outcome compared to a larger area of damage.


Asunto(s)
Células-Madre Neurales/citología , Células-Madre Neurales/trasplante , Trasplante de Células Madre , Accidente Cerebrovascular/patología , Accidente Cerebrovascular/terapia , Animales , Conducta Animal , Vasos Sanguíneos/patología , Diferenciación Celular , Línea Celular , Enfermedad Crónica , Modelos Animales de Enfermedad , Supervivencia de Injerto , Humanos , Imagen por Resonancia Magnética , Neurogénesis , Ratas , Resultado del Tratamiento
14.
Neurochem Int ; 59(3): 421-31, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21624409

RESUMEN

Expression of group III metabotropic glutamate receptors (mGluR) was established by RT-PCR and immunocytochemistry on a cultured clonal human neural stem/progenitor cell (hNSPC) line derived from fetal ventral mesencephalon (VM). Selective activation of these receptors by the group III mGluR agonist L-(+)-2-amino-4-phosphonobutyric acid (L-AP4) prevented increases in cAMP levels following forskolin stimulation, suggesting these receptors are coupled to their canonical G-protein coupled signal transduction pathway. Tonic exposure of undifferentiated cultures to L-AP4 resulted in a decrease in cellular metabolism and proliferation in the absence of toxicity, as measured by MTT and LDH assays, in a dose-dependent manner. This was confirmed by a reduction in BrdU incorporation into nuclear DNA, suggestive of an anti-proliferative effect of L-AP4. This effect was rescued by co-addition of the broad-spectrum group III mGluR competitive antagonist (RS)-a-cyclopropyl-4-phosphonophenylglycine (CPPG), demonstrating a receptor-mediated mechanism, but not mimicked by application of the cell permeable cAMP analogue dibutyrl cAMP (db-cAMP). The potency of these effects of L-AP4 indicates that this is an mGlu7 subtype-mediated effect. Tonic exposure of undifferentiated cultures to the mGlu7 selective allosteric agonist N,N'-bis(diphenylmethyl)-1,2-ethanediamine dihydrochloride (AMN082), but not the mGlu4 selective allosteric agonist (±)-cis-2-(3,5-dicholorphenylcarbamoyl)cyclohexanecarboxylic acid (VU0155041), or the mGlu8 selective agonist (S)-3,4-dicarboxyphenylglycine ((S)-3,4-DCPG) resulted in an identical anti-proliferative effect to L-AP4, confirming the involvement of the mGlu7 subtype. In differentiating cultures, tonic exposure to L-AP4 or AMN082 resulted in a significant shift towards an astrocyte cell fate. The mGlu7 receptor therefore provides a new opportunity to influence the proliferation and differentiation of ventral mesencephalon-derived hNSPC.


Asunto(s)
Astrocitos/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Mesencéfalo/efectos de los fármacos , Receptores de Glutamato Metabotrópico/agonistas , Células Madre/citología , Astrocitos/citología , Secuencia de Bases , Proliferación Celular , Cartilla de ADN , Humanos , Inmunohistoquímica , Mesencéfalo/citología , Reacción en Cadena de la Polimerasa
15.
BMC Neurosci ; 10: 86, 2009 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-19622162

RESUMEN

BACKGROUND: The human neural stem cell line CTX0E03 was developed for the cell based treatment of chronic stroke disability. Derived from fetal cortical brain tissue, CTX0E03 is a clonal cell line that contains a single copy of the c-mycERTAM transgene delivered by retroviral infection. Under the conditional regulation by 4-hydroxytamoxifen (4-OHT), c-mycERTAM enabled large-scale stable banking of the CTX0E03 cells. In this study, we investigated the fate of this transgene following growth arrest (EGF, bFGF and 4-OHT withdrawal) in vitro and following intracerebral implantation into a mid-cerebral artery occluded (MCAo) rat brain. In vitro, 4-weeks after removing growth factors and 4-OHT from the culture medium, c-mycERTAM transgene transcription is reduced by ~75%. Furthermore, immunocytochemistry and western blotting demonstrated a concurrent decrease in the c-MycERTAM protein. To examine the transcription of the transgene in vivo, CTX0E03 cells (450,000) were implanted 4-weeks post MCAo lesion and analysed for human cell survival and c-mycERTAM transcription by qPCR and qRT-PCR, respectively. RESULTS: The results show that CTX0E03 cells were present in all grafted animal brains ranging from 6.3% to 39.8% of the total cells injected. Prior to implantation, the CTX0E03 cell suspension contained 215.7 (SEM = 13.2) copies of the c-mycERTAM transcript per cell. After implantation the c-mycERTAM transcript copy number per CTX0E03 cell had reduced to 6.9 (SEM = 3.4) at 1-week and 7.7 (SEM = 2.5) at 4-weeks. Bisulfite genomic DNA sequencing of the in vivo samples confirmed c-mycERTAM silencing occurred through methylation of the transgene promoter sequence. CONCLUSION: In conclusion the results confirm that CTX0E03 cells downregulated c-mycERTAM transgene expression both in vitro following EGF, bFGF and 4-OHT withdrawal and in vivo following implantation in MCAo rat brain. The silencing of the c-mycERTAM transgene in vivo provides an additional safety feature of CTX0E03 cells for potential clinical application.


Asunto(s)
Corteza Cerebral/trasplante , Células Madre Fetales/trasplante , Silenciador del Gen , Infarto de la Arteria Cerebral Media/genética , Animales , Línea Celular , Células Cultivadas , Corteza Cerebral/irrigación sanguínea , Células Madre Fetales/citología , Humanos , Masculino , Neuronas/citología , Ratas , Ratas Sprague-Dawley , Transgenes , Trasplante Heterólogo
16.
Circ Res ; 104(9): 1095-102, 2009 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-19342601

RESUMEN

We evaluated the healing potential of human fetal aorta-derived CD133(+) progenitor cells and their conditioned medium (CD133(+) CCM) in a new model of ischemic diabetic ulcer. Streptozotocin-induced diabetic mice underwent bilateral limb ischemia and wounding. One wound was covered with collagen containing 2x10(4) CD133(+) or CD133(-) cells or vehicle. The contralateral wound, covered with only collagen, served as control. Fetal CD133(+) cells expressed high levels of wingless (Wnt) genes, which were downregulated following differentiation into CD133(-) cells along with upregulation of Wnt antagonists secreted frizzled-related protein (sFRP)-1, -3, and -4. CD133(+) cells accelerated wound closure as compared with CD133(-) or vehicle and promoted angiogenesis through stimulation of endothelial cell proliferation, migration, and survival by paracrine effects. CD133(+) cells secreted high levels of vascular endothelial growth factor (VEGF)-A and interleukin (IL)-8. Consistently, CD133(+) CCM accelerated wound closure and reparative angiogenesis, with this action abrogated by co-administering the Wnt antagonist sFRP-1 or neutralizing antibodies against VEGF-A or IL-8. In vitro, these effects were recapitulated following exposure of high-glucose-primed human umbilical vein endothelial cells to CD133(+) CCM, resulting in stimulation of migration, angiogenesis-like network formation and induction of Wnt expression. The promigratory and proangiogenic effect of CD133(+) CCM was blunted by sFRP-1, as well as antibodies against VEGF-A or IL-8. CD133(+) cells stimulate wound healing by paracrine mechanisms that activate Wnt signaling pathway in recipients. These preclinical findings open new perspectives for the cure of diabetic ulcers.


Asunto(s)
Diabetes Mellitus Experimental/complicaciones , Pie Diabético/cirugía , Células Madre Fetales/trasplante , Isquemia/complicaciones , Extremidad Inferior/irrigación sanguínea , Neovascularización Fisiológica , Trasplante de Células Madre , Proteínas Wnt/metabolismo , Cicatrización de Heridas , Antígeno AC133 , Animales , Antígenos CD/análisis , Aorta/embriología , Diferenciación Celular , Movimiento Celular , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Medios de Cultivo Condicionados/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/fisiopatología , Diabetes Mellitus Experimental/cirugía , Pie Diabético/etiología , Pie Diabético/metabolismo , Pie Diabético/fisiopatología , Células Madre Fetales/inmunología , Células Madre Fetales/metabolismo , Glicoproteínas/análisis , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Interleucina-8/metabolismo , Isquemia/metabolismo , Isquemia/fisiopatología , Isquemia/cirugía , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Comunicación Paracrina , Péptidos/análisis , Transducción de Señal , Factores de Tiempo , Factor A de Crecimiento Endotelial Vascular/metabolismo
17.
Cell Transplant ; 16(2): 101-15, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17474292

RESUMEN

Human stem cells, progenitor cells, and cell lines have been derived from embryonic, fetal, and adult sources in the search for graft tissue suitable for the treatment of CNS disorders. An increasing number of experimental studies have shown that grafts from several sources survive, differentiate into distinct cell types, and exert positive functional effects in experimental animal models, but little attention has been given to developing cells under conditions of good manufacturing practice (GMP) that can be scaled up for mass treatment. The capacity for continued division of stem cells in culture offers the opportunity to expand their production to meet the widespread clinical demands posed by neurodegenerative diseases. However, maintaining stem cell division in culture long term, while ensuring differentiation after transplantation, requires genetic and/ or oncogenetic manipulations, which may affect the genetic stability and in vivo survival of cells. This review outlines the stages, selection criteria, problems, and ultimately the successes arising in the development of conditionally immortal clinical grade stem cell lines, which divide in vitro, differentiate in vivo, and exert positive functional effects. These processes are specifically exemplified by the murine MHP36 cell line, conditionally immortalized by a temperature-sensitive mutant of the SV40 large T antigen, and cell lines transfected with the c-myc protein fused with a mutated estrogen receptor (c-mycERTAM), regulated by a tamoxifen metabolite, but the issues raised are common to all routes for the development of effective clinical grade cells.


Asunto(s)
Trasplante de Células Madre/métodos , Células Madre/citología , Células Madre/metabolismo , Animales , División Celular , Humanos , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Receptores de Estrógenos/metabolismo , Cicatrización de Heridas
18.
Exp Neurol ; 199(1): 143-55, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16464451

RESUMEN

Transplantation of neural stem cells into the brain is a novel approach to the treatment of chronic stroke disability. For clinical application, safety and efficacy of defined, stable cell lines produced under GMP conditions are required. To this end, a human neural stem cell line, CTX0E03, was derived from human somatic stem cells following genetic modification with a conditional immortalizing gene, c-mycER(TAM). This transgene generates a fusion protein that stimulates cell proliferation in the presence of a synthetic drug 4-hydroxy-tamoxifen (4-OHT). The cell line is clonal, expands rapidly in culture (doubling time 50-60 h) and has a normal human karyotype (46 XY). In the absence of growth factors and 4-OHT, the cells undergo growth arrest and differentiate into neurons and astrocytes. Transplantation of CTX0E03 in a rat model of stroke (MCAo) caused statistically significant improvements in both sensorimotor function and gross motor asymmetry at 6-12 weeks post-grafting. In addition, cell migration and long-term survival in vivo were not associated with significant cell proliferation. These data indicate that CTX0E03 has the appropriate biological and manufacturing characteristics necessary for development as a therapeutic cell line.


Asunto(s)
Corteza Cerebral/citología , Infarto de la Arteria Cerebral Media/cirugía , Células Neuroepiteliales/fisiología , Trasplante de Células Madre , Células Madre/fisiología , Análisis de Varianza , Animales , Conducta Animal , Diferenciación Celular/fisiología , Movimiento Celular , Corteza Cerebral/embriología , Células Clonales , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Feto , Humanos , Hidroxitestosteronas/farmacología , Masculino , Actividad Motora/fisiología , Células Neuroepiteliales/citología , ARN Mensajero/biosíntesis , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Trasplante de Células Madre/métodos , Telomerasa/efectos de los fármacos , Telomerasa/metabolismo , Factores de Tiempo , Transducción Genética/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...