Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Genome Biol ; 19(1): 43, 2018 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-29587824

RESUMO

BACKGROUND: Glioma stem cells (GSCs) are a subpopulation of stem-like cells that contribute to glioblastoma (GBM) aggressiveness, recurrence, and resistance to radiation and chemotherapy. Therapeutically targeting the GSC population may improve patient survival, but unique vulnerabilities need to be identified. RESULTS: We isolate GSCs from well-characterized GBM patient-derived xenografts (PDX), characterize their stemness properties using immunofluorescence staining, profile their epigenome including 5mC, 5hmC, 5fC/5caC, and two enhancer marks, and define their transcriptome. Fetal brain-derived neural stem/progenitor cells are used as a comparison to define potential unique and common molecular features between these different brain-derived cells with stem properties. Our integrative study reveals that abnormal expression of ten-eleven-translocation (TET) family members correlates with global levels of 5mC and 5fC/5caC and may be responsible for the distinct levels of these marks between glioma and neural stem cells. Heterogenous transcriptome and epigenome signatures among GSCs converge on several genes and pathways, including DNA damage response and cell proliferation, which are highly correlated with TET expression. Distinct enhancer landscapes are also strongly associated with differential gene regulation between glioma and neural stem cells; they exhibit unique co-localization patterns with DNA epigenetic mark switching events. Upon differentiation, glioma and neural stem cells exhibit distinct responses with regard to TET expression and DNA mark changes in the genome and GSCs fail to properly remodel their epigenome. CONCLUSIONS: Our integrative epigenomic and transcriptomic characterization reveals fundamentally distinct yet potentially targetable biologic features of GSCs that result from their distinct epigenomic landscapes.


Assuntos
Epigênese Genética , Regulação Neoplásica da Expressão Gênica , Glioma/genética , Células-Tronco Neoplásicas/metabolismo , Animais , Diferenciação Celular , Metilação de DNA , Reparo do DNA , Proteínas de Ligação a DNA/metabolismo , Elementos Facilitadores Genéticos , Perfilação da Expressão Gênica , Redes Reguladoras de Genes , Glioma/metabolismo , Código das Histonas , Humanos , Camundongos , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Regiões Promotoras Genéticas
2.
PLoS One ; 11(3): e0150850, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26986213

RESUMO

BACKGROUND: Cardiovascular disease, a progressive manifestation of α-L-iduronidase deficiency or mucopolysaccharidosis type I, continues in patients both untreated and treated with hematopoietic stem cell transplantation or intravenous enzyme replacement. Few studies have examined the effects of α-L-iduronidase deficiency and subsequent glycosaminoglycan storage upon arterial gene expression to understand the pathogenesis of cardiovascular disease. METHODS: Gene expression in carotid artery, ascending, and descending aortas from four non-tolerized, non-enzyme treated 19 month-old mucopolysaccharidosis type I dogs was compared with expression in corresponding vascular segments from three normal, age-matched dogs. Data were analyzed using R and whole genome network correlation analysis, a bias-free method of categorizing expression level and significance into discrete modules. Genes were further categorized based on module-trait relationships. Expression of clusterin, a protein implicated in other etiologies of cardiovascular disease, was assessed in canine and murine mucopolysaccharidosis type I aortas via Western blot and in situ immunohistochemistry. RESULTS: Gene families with more than two-fold, significant increased expression involved lysosomal function, proteasome function, and immune regulation. Significantly downregulated genes were related to cellular adhesion, cytoskeletal elements, and calcium regulation. Clusterin gene overexpression (9-fold) and protein overexpression (1.3 to 1.62-fold) was confirmed and located specifically in arterial plaques of mucopolysaccharidosis-affected dogs and mice. CONCLUSIONS: Overexpression of lysosomal and proteasomal-related genes are expected responses to cellular stress induced by lysosomal storage in mucopolysaccharidosis type I. Upregulation of immunity-related genes implicates the potential involvement of glycosaminoglycan-induced inflammation in the pathogenesis of mucopolysaccharidosis-related arterial disease, for which clusterin represents a potential biomarker.


Assuntos
Doenças Cardiovasculares/etiologia , Doenças Cardiovasculares/patologia , Regulação da Expressão Gênica , Inflamação/complicações , Mucopolissacaridose I/complicações , Animais , Aorta/metabolismo , Aorta/patologia , Doenças Cardiovasculares/genética , Artérias Carótidas/imunologia , Artérias Carótidas/patologia , Clusterina/análise , Cães , Feminino , Redes Reguladoras de Genes , Inflamação/genética , Camundongos Endogâmicos C57BL , Mucopolissacaridose I/genética
3.
Mol Ther Methods Clin Dev ; 2: 14068, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26052536

RESUMO

Mucopolysaccharidosis type I (MPS I) is an inherited α-L-iduronidase (IDUA, I) deficiency in which glycosaminoglycan (GAG) accumulation causes progressive multisystem organ dysfunction, neurological impairment, and death. Current MPS I mouse models, based on a NOD/SCID (NS) background, are short-lived, providing a very narrow window to assess the long-term efficacy of therapeutic interventions. They also develop thymic lymphomas, making the assessment of potential tumorigenicity of human stem cell transplantation problematic. We therefore developed a new MPS I model based on a NOD/SCID/Il2rγ (NSG) background. This model lives longer than 1 year and is tumor-free during that time. NSG MPS I (NSGI) mice exhibit the typical phenotypic features of MPS I including coarsened fur and facial features, reduced/abnormal gait, kyphosis, and corneal clouding. IDUA is undetectable in all tissues examined while GAG levels are dramatically higher in most tissues. NSGI brain shows a significant inflammatory response and prominent gliosis. Neurological MPS I manifestations are evidenced by impaired performance in behavioral tests. Human neural and hematopoietic stem cells were found to readily engraft, with human cells detectable for at least 1 year posttransplantation. This new MPS I model is thus suitable for preclinical testing of novel pluripotent stem cell-based therapy approaches.

4.
J Neurosci Res ; 91(10): 1247-62, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23893392

RESUMO

Robust strategies for developing patient-specific, human, induced pluripotent stem cell (iPSC)-based therapies of the brain require an ability to derive large numbers of highly defined neural cells. Recent progress in iPSC culture techniques includes partial-to-complete elimination of feeder layers, use of defined media, and single-cell passaging. However, these techniques still require embryoid body formation or coculture for differentiation into neural stem cells (NSCs). In addition, none of the published methodologies has employed all of the advances in a single culture system. Here we describe a reliable method for long-term, single-cell passaging of PSCs using a feeder-free, defined culture system that produces confluent, adherent PSCs that can be differentiated into NSCs. To provide a basis for robust quality control, we have devised a system of cellular nomenclature that describes an accurate genotype and phenotype of the cells at specific stages in the process. We demonstrate that this protocol allows for the efficient, large-scale, cGMP-compliant production of transplantable NSCs from all lines tested. We also show that NSCs generated from iPSCs produced with the process described are capable of forming both glia defined by their expression of S100ß and neurons that fire repetitive action potentials.


Assuntos
Técnicas de Cultura de Células/métodos , Diferenciação Celular , Proliferação de Células , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Neurais/citologia , Diferenciação Celular/fisiologia , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Neurônios/citologia , Neurônios/fisiologia , Neurônios/transplante , Técnicas de Patch-Clamp
5.
Cancer Cell Int ; 12(1): 41, 2012 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-22995409

RESUMO

BACKGROUND: The cancer stem cell (CSC) hypothesis posits that deregulated neural stem cells (NSCs) form the basis of brain tumors such as glioblastoma multiforme (GBM). GBM, however, usually forms in the cerebral white matter while normal NSCs reside in subventricular and hippocampal regions. We attempted to characterize CSCs from a rare form of glioblastoma multiforme involving the neurogenic ventricular wall. METHODS: We described isolating CSCs from a GBM involving the lateral ventricles and characterized these cells with in vitro molecular biomarker profiling, cellular behavior, ex vivo and in vivo techniques. RESULTS: The patient's MRI revealed a heterogeneous mass with associated edema, involving the left subventricular zone. Histological examination of the tumor established it as being a high-grade glial neoplasm, characterized by polygonal and fusiform cells with marked nuclear atypia, amphophilic cytoplasm, prominent nucleoli, frequent mitotic figures, irregular zones of necrosis and vascular hyperplasia. Recurrence of the tumor occurred shortly after the surgical resection. CD133-positive cells, isolated from the tumor, expressed stem cell markers including nestin, CD133, Ki67, Sox2, EFNB1, EFNB2, EFNB3, Cav-1, Musashi, Nucleostemin, Notch 2, Notch 4, and Pax6. Biomarkers expressed in differentiated cells included Cathepsin L, Cathepsin B, Mucin18, Mucin24, c-Myc, NSE, and TIMP1. Expression of unique cancer-related transcripts in these CD133-positive cells, such as caveolin-1 and -2, do not appear to have been previously reported in the literature. Ex vivo organotypic brain slice co-culture showed that the CD133+ cells behaved like tumor cells. The CD133-positive cells also induced tumor formation when they were stereotactically transplanted into the brains of the immune-deficient NOD/SCID mice. CONCLUSIONS: This brain tumor involving the neurogenic lateral ventricular wall was comprised of tumor-forming, CD133-positive cancer stem cells, which are likely the driving force for the rapid recurrence of the tumor in the patient.

6.
Acta Biochim Biophys Sin (Shanghai) ; 44(7): 597-605, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22659494

RESUMO

Immunofluorescence assay (IFA) is one of the most frequently used methods in the biological sciences and clinic diagnosis, but it is expensive and time-consuming. To overcome these limitations, we developed a faster and more cost-effective IFA (f-IFA) by modifying the standard IFA, and applied this method to track the progression of human cytomegalovirus (HCMV) infection in different cells. The f-IFA that we developed not only saves time, but also dramatically reduces the quantity of antibody (Ab), which will facilitate the application of IFA in clinic diagnosis. f-IFA requires only 15 min for blocking, 10 min incubation for each primary and secondary Abs, followed by 1 min extensive wash after each incubation. Only 25 µl of diluted Ab solution was needed for each coverslip at the primary and secondary Ab incubation steps. In addition, all steps were performed at room temperature. This f-IFA has been applied successfully to follow virion entry (pp65) and expression of viral genes (IE1, UL44, and pp65) in order to track the details of HCMV infection process. We found that ∼0.5% HCMV-infected T98G cells formed multiple-micronuclei (IE1 and nucleus staining) and had virus shedding (pp65 staining) by f-IFA, which could not be detected by the traditional IFA. Our results indicated that f-IFA is a sensitive, convenient, fast, and cost-effective method for investigating the details of virus infection progress, especially HCMV infection. The faster and cost-effective feature with higher sensitivity and specificity implies that f-IFA has potential applications in clinical diagnosis.


Assuntos
Citomegalovirus/metabolismo , Imunofluorescência/métodos , Proteínas Virais/metabolismo , Internalização do Vírus , Linhagem Celular Tumoral , Células Cultivadas , Análise Custo-Benefício , Citomegalovirus/crescimento & desenvolvimento , Proteínas de Ligação a DNA/metabolismo , Fibroblastos/virologia , Imunofluorescência/economia , Glioblastoma/patologia , Glioblastoma/virologia , Humanos , Proteínas Imediatamente Precoces/metabolismo , Pulmão/citologia , Pulmão/embriologia , Microscopia de Fluorescência , Células-Tronco Neurais/virologia , Fosfoproteínas/metabolismo , Reprodutibilidade dos Testes , Fatores de Tempo , Proteínas da Matriz Viral/metabolismo
8.
Neurology ; 76(13): 1126-34, 2011 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-21346220

RESUMO

OBJECTIVES: New data suggest that glioma stem-like cells (GSCs) and neural stem/progenitor cells (NSCs) may share common origins. GSCs drive tumor proliferation and appear to be resistant to classic chemotherapy, while the effects of chemotherapy on NSCs are not well studied. As the role of NSCs in learning and memory is increasingly recognized, we need to identify drugs that reduce neurotoxicity but are still effective against glial tumors. METHODS: We treated 3 human NSC cultures and multiple low- and high-grade GSC cultures with the commonly used agents temozolomide (TMZ) and cisplatin (CIS), and with 2 newer, promising drugs: the proteasome inhibitor bortezomib (BTZ) and the epidermal growth factor receptor tyrosine kinase inhibitor erlotinib (ERL). We measured cell survival, proliferation, cell death induction, and drug resistance markers. RESULTS: TMZ decreased NSC viability, while minimally affecting GSCs. TMZ induced NSC death, which was partially compensated for by increased proliferation. CIS had similar effects. The NSC's sensitivity to TMZ and CIS correlated with low expression of the multidrug resistance gene ABCG2, but not of MGMT or MSH1/MLH2. BTZ caused an 80%decrease in GSCs, while minimally affecting NSCs. GSCs had lower proteasome levels and activity after BTZ treatment. ERL treatment also decreased GSC numbers, but not NSC viability, which correlated with low EGFR expression in NSCs compared to GSCs. CONCLUSIONS: Newer chemotherapy agents ERL and BTZ are effective against GSCs yet produce minimal effects on NSCs, while the older drugs TMZ and CIS are more toxic for NSCs than for GSCs. The identification and testing of more selective drugs is clearly warranted.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Encefálicas , Resistencia a Medicamentos Antineoplásicos , Glioma , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neurais/efeitos dos fármacos , Animais , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Ácidos Borônicos/farmacologia , Bortezomib , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Caspase 3/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Cisplatino/farmacologia , Dacarbazina/análogos & derivados , Dacarbazina/farmacologia , Receptores ErbB/metabolismo , Cloridrato de Erlotinib , Expressão Gênica , Glioma/tratamento farmacológico , Glioma/patologia , Humanos , Células-Tronco Neoplásicas/citologia , Células-Tronco Neoplásicas/fisiologia , Células-Tronco Neurais/citologia , Células-Tronco Neurais/fisiologia , Inibidores de Proteínas Quinases/farmacologia , Pirazinas/farmacologia , Quinazolinas/farmacologia , Temozolomida
9.
Mol Cancer Res ; 7(10): 1622-34, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19825994

RESUMO

DNA methylation is an epigenetic mark essential for mammalian development, genomic stability, and imprinting. DNA methylation patterns are established and maintained by three DNA methyltransferases: DNMT1, DNMT3A, and DNMT3B. Interestingly, all three DNMTs make use of alternative splicing. DNMT3B has nearly 40 known splice variants expressed in a tissue- and disease-specific manner, but very little is known about the role of these splice variants in modulating DNMT3B function. We describe here the identification and characterization of a novel alternatively spliced form of DNMT3B lacking exon 5 within the NH(2)-terminal regulatory domain. This variant, which we term DNMT3B3Delta5 because it is closely related in structure to the ubiquitously expressed DNMT3B3 isoform, is highly expressed in pluripotent cells and brain tissue, is downregulated during differentiation, and is conserved in the mouse. Creation of pluripotent iPS cells from fibroblasts results in marked induction of DNMT3B3Delta5. DNMT3B3Delta5 expression is also altered in human disease, with tumor cell lines displaying elevated or reduced expression depending on their tissue of origin. We then compared the DNA binding and subcellular localization of DNMT3B3Delta5 versus DNMT3B3, revealing that DNMT3B3Delta5 possessed significantly enhanced DNA binding affinity and displayed an altered nuclear distribution. Finally, ectopic overexpression of DNMT3B3Delta5 resulted in repetitive element hypomethylation and enhanced cell growth in a colony formation assay. Taken together, these results show that DNMT3B3Delta5 may play an important role in stem cell maintenance or differentiation and suggest that sequences encoded by exon 5 influence the functional properties of DNMT3B.


Assuntos
DNA (Citosina-5-)-Metiltransferases/genética , Metilação de DNA/genética , Proteínas de Ligação a DNA/genética , Neoplasias/genética , Neoplasias/metabolismo , Células-Tronco Pluripotentes/metabolismo , Processamento Alternativo/genética , Animais , Sequência de Bases/genética , Linhagem Celular Tumoral , Núcleo Celular/genética , Núcleo Celular/metabolismo , Proliferação de Células , DNA (Citosina-5-)-Metiltransferases/química , DNA (Citosina-5-)-Metiltransferases/isolamento & purificação , Epigênese Genética/genética , Éxons/genética , Instabilidade Genômica/genética , Humanos , Camundongos , Células-Tronco Pluripotentes/citologia , Isoformas de Proteínas , Estrutura Terciária de Proteína/genética , Ensaio Tumoral de Célula-Tronco , DNA Metiltransferase 3B
10.
Mol Genet Metab ; 98(4): 406-11, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19748810

RESUMO

BACKGROUND: The mucopolysaccharidoses (MPSs) are a family of lysosomal storage disorders caused by impaired glycosaminoglycan degradation. Characteristic brain imaging abnormalities are seen in MPS patients. This study aims to determine the effects of hematopoietic stem cell transplantation (HSCT) and/or intravenous enzyme replacement therapy (ERT) on these abnormalities. METHODS: A retrospective chart and brain imaging study review was conducted of MPS types I and II patients with brain magnetic resonance imaging (MRI) performed at, and following, initiation of treatment. White matter abnormalities, dilated perivascular spaces, corpus callosal abnormalities, and ventriculomegaly were scored by three independent neuroradiologists blinded to cognitive status, date of treatment initiation, and type(s) of treatment. RESULTS: Five patients were identified: three patients with MPS I and two with MPS II. Duration of follow-up ranged from 13 to 51 months. One patient had severe MPS I (genotype W402X/35del12) and received ERT followed by HSCT. The remaining patients had ERT only. The other two MPS I patients were cognitively normal siblings (genotype P533R/P533R) with an intermediate phenotype. One MPS II patient had moderate cognitive impairment without regression (genotype 979insAGCA); the other (genotype R8X) had normal cognition. There was very little inter-observer variation in MRI scoring. The greatest abnormalities for each patient were found at initial MRI. All patients, including the ERT-only patients, demonstrated improved or unchanged MRI scores following treatment. Severity of white matter abnormalities or dilated perivascular spaces did not correlate with cognitive impairment; as such, extensive pre-treatment MRI abnormalities were noted in the older, cognitively normal MPS I sibling. In comparison, his younger sibling had only mild MRI abnormalities at the same age, after receiving 4 years of ERT. CONCLUSIONS: This study represents one of the first to document the CNS effects of ERT in MPS patients utilizing serial brain MR imaging studies, and raises several important observations. Brain MRI abnormalities typically become more pronounced with age; initiation of ERT or HSCT reversed or stabilized this trend in the MPS patients studied. In addition, earlier initiation of treatment resulted in decreased severity of imaging abnormalities. Possible mechanisms for these observations include improved cerebrospinal fluid dynamics, reduced central nervous system glycosaminoglycan storage via efflux through the blood-brain barrier (BBB), repair of damaged BBB, reduction in CNS inflammation, or ERT permeability through the BBB.


Assuntos
Mapeamento Encefálico , Mucopolissacaridose II/patologia , Mucopolissacaridose II/terapia , Mucopolissacaridose I/patologia , Mucopolissacaridose I/terapia , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Imageamento por Ressonância Magnética , Masculino
11.
Stem Cells ; 27(1): 7-17, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18832593

RESUMO

Hypoxia commonly occurs in solid tumors of the central nervous system (CNS) and often interferes with therapies designed to stop their growth. We found that pediatric high-grade glioma (HGG)-derived precursors showed greater expansion under lower oxygen tension, typical of solid tumors, than normal CNS precursors. Hypoxia inhibited p53 activation and subsequent astroglial differentiation of HGG precursors. Surprisingly, although HGG precursors generated endogenous bone morphogenetic protein (BMP) signaling that promoted mitotic arrest under high oxygen tension, this signaling was actively repressed by hypoxia. An acute increase in oxygen tension led to Smad activation within 30 minutes, even in the absence of exogenous BMP treatment. Treatment with BMPs further promoted astroglial differentiation or death of HGG precursors under high oxygen tension, but this effect was inhibited under hypoxic conditions. Silencing of hypoxia-inducible factor 1alpha (HIF1alpha) led to Smad activation even under hypoxic conditions, indicating that HIF1alpha is required for BMP repression. Conversely, BMP activation at high oxygen tension led to reciprocal degradation of HIF1alpha; this BMP-induced degradation was inhibited in low oxygen. These results show a novel, mutually antagonistic interaction of hypoxia-response and neural differentiation signals in HGG proliferation, and suggest differences between normal and HGG precursors that may be exploited for pediatric brain cancer therapy.


Assuntos
Proteína Morfogenética Óssea 2/farmacologia , Diferenciação Celular/efeitos dos fármacos , Glioma/patologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Astrócitos/citologia , Astrócitos/efeitos dos fármacos , Hipóxia Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Criança , Regulação para Baixo/efeitos dos fármacos , Inativação Gênica/efeitos dos fármacos , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Mitose/efeitos dos fármacos , Células-Tronco Neoplásicas/patologia , Oxigênio/farmacologia , Transdução de Sinais/efeitos dos fármacos , Proteínas Smad/metabolismo , Proteína Supressora de Tumor p53/metabolismo
12.
Nature ; 455(7211): 401-5, 2008 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-18724358

RESUMO

Stem cells are defined as self-renewing cell populations that can differentiate into multiple distinct cell types. However, hundreds of different human cell lines from embryonic, fetal and adult sources have been called stem cells, even though they range from pluripotent cells-typified by embryonic stem cells, which are capable of virtually unlimited proliferation and differentiation-to adult stem cell lines, which can generate a far more limited repertoire of differentiated cell types. The rapid increase in reports of new sources of stem cells and their anticipated value to regenerative medicine has highlighted the need for a general, reproducible method for classification of these cells. We report here the creation and analysis of a database of global gene expression profiles (which we call the 'stem cell matrix') that enables the classification of cultured human stem cells in the context of a wide variety of pluripotent, multipotent and differentiated cell types. Using an unsupervised clustering method to categorize a collection of approximately 150 cell samples, we discovered that pluripotent stem cell lines group together, whereas other cell types, including brain-derived neural stem cell lines, are very diverse. Using further bioinformatic analysis we uncovered a protein-protein network (PluriNet) that is shared by the pluripotent cells (embryonic stem cells, embryonal carcinomas and induced pluripotent cells). Analysis of published data showed that the PluriNet seems to be a common characteristic of pluripotent cells, including mouse embryonic stem and induced pluripotent cells and human oocytes. Our results offer a new strategy for classifying stem cells and support the idea that pluripotency and self-renewal are under tight control by specific molecular networks.


Assuntos
Perfilação da Expressão Gênica , Células-Tronco/classificação , Células-Tronco/metabolismo , Algoritmos , Animais , Inteligência Artificial , Diferenciação Celular , Linhagem Celular , Biologia Computacional , Bases de Dados Factuais , Células-Tronco Embrionárias/classificação , Células-Tronco Embrionárias/metabolismo , Humanos , Camundongos , Células-Tronco Multipotentes/classificação , Células-Tronco Multipotentes/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Oócitos/classificação , Oócitos/metabolismo , Fenótipo , Células-Tronco Pluripotentes/classificação , Células-Tronco Pluripotentes/metabolismo , Ligação Proteica
13.
Cloning Stem Cells ; 10(3): 391-402, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18729769

RESUMO

Work in rodents has demonstrated that progenitor transplantation can achieve limited photoreceptor replacement in the mammalian retina; however, replication of these findings on a clinically relevant scale requires a large animal model. To evaluate the ability of porcine retinal progenitor cells to survival as allografts and integrate into the host retinal architecture, we isolated donor cells from fetal green fluorescent protein (GFP)-transgenic pigs. Cultures were propagated from the brain, retina, and corneo-scleral limbus. GFP expression rapidly increased with time in culture, although lower in conjunction with photoreceptor markers and glial fibrillary acid protein (GFAP), thus suggesting downregulation of GFP during differentiation. Following transplantation, GFP expression allowed histological visualization of integrated cells and extension of fine processes to adjacent plexiform layers. GFP expression in subretinal grafts was high in cells expressing vimentin and lower in cells expressing photoreceptor markers, again consistent with possible downregulation during differentiation. Cells survived transplantation to the injured retina of allorecipients at all time points examined (up to 10 weeks) in the absence of exogenous immune suppression without indications of rejection. These findings demonstrate the feasibility of allogeneic progenitor transplantation in a large mammal and the utility of the pig in ocular regeneration studies.


Assuntos
Animais Geneticamente Modificados , Proteínas de Fluorescência Verde/metabolismo , Retina/citologia , Transplante de Células-Tronco , Células-Tronco/citologia , Transplante Homólogo , Animais , Biomarcadores/metabolismo , Células Cultivadas , Feminino , Genes Reporter , Proteínas de Fluorescência Verde/genética , Gravidez , Retina/fisiologia , Células-Tronco/fisiologia , Suínos
14.
Lab Invest ; 88(9): 910-25, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18607344

RESUMO

DNA hypermethylation-mediated gene silencing is a frequent and early contributor to aberrant cell growth and invasion in cancer. Malignant gliomas are the most common primary brain tumors in adults and the second most common tumor in children. Morbidity and mortality are high in glioma patients because tumors are resistant to treatment and are highly invasive into surrounding brain tissue rendering complete surgical resection impossible. Invasiveness is regulated by the interplay between secreted proteases (eg, cathepsins) and their endogenous inhibitors (cystatins). In our previous studies we identified cystatin E/M (CST6) as a frequent target of epigenetic silencing in glioma. Cystatin E/M is a potent inhibitor of cathepsin B, which is frequently overexpressed in glioma. Here, we study the expression of cystatin E/M in normal brain and show that it is highly and moderately expressed in oligodendrocytes and astrocytes, respectively, but not in neurons. Consistent with this, the CST6 promoter is hypomethylated in all normal samples using methylation-specific PCR, bisulfite genomic sequencing, and pyrosequencing. In contrast, 78% of 28 primary brain tumors demonstrated reduced/absent cystatin E/M expression using a tissue microarray and this reduced expression correlated with CST6 promoter hypermethylation. Interestingly, CST6 was expressed in neural stem cells (NSC) and markedly induced upon differentiation, whereas a glioma tumor initiating cell (TIC) line was completely blocked for CST6 expression by promoter methylation. Analysis of primary pediatric brain tumor-derived lines also showed CST6 downregulation and methylation in nearly 100% of 12 cases. Finally, ectopic expression of cystatin E/M in glioma lines reduced cell motility and invasion. These results demonstrate that epigenetic silencing of CST6 is frequent in adult and pediatric brain tumors and occurs in TICs, which are thought to give rise to the tumor. CST6 methylation may therefore represent a novel prognostic marker and therapeutic target specifically altered in TICs.


Assuntos
Neoplasias Encefálicas/metabolismo , Encéfalo/metabolismo , Cistatinas/metabolismo , Epigênese Genética , Inativação Gênica , Glioma/genética , Sequência de Bases , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Cistatina M , Cistatinas/genética , Metilação de DNA , Primers do DNA , Glioma/patologia , Humanos , Hibridização in Situ Fluorescente , Invasividade Neoplásica , Regiões Promotoras Genéticas , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise Serial de Tecidos
15.
Methods ; 45(2): 142-58, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18593611

RESUMO

Human pluripotent stem cells have the unique properties of being able to proliferate indefinitely in their undifferentiated state and to differentiate into any somatic cell type. These cells are thus posited to be extremely useful for furthering our understanding of both normal and abnormal human development, providing a human cell preparation that can be used to screen for new reagents or therapeutic agents, and generating large numbers of differentiated cells that can be used for transplantation purposes. Critical among the applications for the latter are diseases and injuries of the nervous system, medical approaches to which have been, to date, primarily palliative in nature. Differentiation of human pluripotent stem cells into cells of the neural lineage, therefore, has become a central focus of a number of laboratories. This has resulted in the description in the literature of several dozen methods for neural cell differentiation from human pluripotent stem cells. Among these are methods for the generation of such divergent neural cells as dopaminergic neurons, retinal neurons, ventral motoneurons, and oligodendroglial progenitors. In this review, we attempt to fully describe most of these methods, breaking them down into five basic subdivisions: (1) starting material, (2) induction of loss of pluripotency, (3) neural induction, (4) neural maintenance and expansion, and (5) neuronal/glial differentiation. We also show data supporting the concept that undifferentiated human pluripotent stem cells appear to have an innate neural differentiation potential. In addition, we evaluate data comparing and contrasting neural stem cells differentiated from human pluripotent stem cells with those derived directly from the human brain.


Assuntos
Diferenciação Celular/fisiologia , Neurônios/citologia , Células-Tronco Pluripotentes/citologia , Técnicas de Cultura de Células , Linhagem da Célula/fisiologia , Proliferação de Células , Separação Celular/métodos , Meios de Cultura , Embrião de Mamíferos/metabolismo , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Humanos , Neurônios/metabolismo , Células-Tronco Pluripotentes/metabolismo
16.
Vet Ophthalmol ; 10(4): 245-53, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17565557

RESUMO

The cat has served as an important nonrodent research model for neurophysiology and retinal degenerative disease processes, yet very little is known about feline neural precursor cells. To culture these cells and evaluate marker expression, brains were dissected from 45-day-old fetuses, enzymatically dissociated, and grown in the presence of EGF, bFGF and PDGF. Expanded cells widely expressed nestin, Sox2, Ki-67, fusin (CXCR4) and vimentin, while subpopulations expressed A2B5, GFAP, or beta-III tubulin. Precursors prelabeled with BrdU and/or transduced with a recombinant lentivirus that expresses GFP were transplanted subretinally in five dystrophic Abyssinian cats. Two to 4 weeks following surgery, histology showed survival of grafted cells in three of the animals. Labeled cells were found in the neuroretina and RPE layer, as well as in the vitreous and the vicinity of Bruch's membrane. There was no evidence of an immunologic response in any of the eyes. Neural precursor cells can therefore be cultured from the developing cat brain and survive as allografts for up to 4 weeks without immune suppression. The feasibility of deriving and transplanting feline neural precursor cells, combined with the availability of the dystrophic Abyssinian cat, provide a new feline model system for the study of retinal repair.


Assuntos
Doenças do Gato/cirurgia , Neurônios/citologia , Retina/citologia , Degeneração Retiniana/veterinária , Células Ganglionares da Retina/citologia , Animais , Gatos , Células Cultivadas , Feminino , Masculino , Degeneração Retiniana/cirurgia , Transplante de Células-Tronco/veterinária
17.
Reprod Biomed Online ; 12(6): 771-5, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16792856

RESUMO

This paper considers embryo grading within a given infertility treatment and suggests an ethical approach to embryo donation for embryonic stem cell harvest. It is concluded that ethical considerations regarding human embryos do not necessarily preclude the use of certain embryos for biomedical research or transplantation. The argument is based on the following rationale: all embryos are not physiologically equal, some low-grade embryos will never be chosen for implantation, cells from low-grade embryos may be donated for transplantation or research, and embryonic stem cells can be harvested from low-grade embryos. This argument bears special importance at this time as embryos created by IVF are still the only source of embryonic stem cells, given the current controversy surrounding published studies of human somatic cell nuclear transfer.


Assuntos
Pesquisas com Embriões/ética , Embrião de Mamíferos/citologia , Células-Tronco , Coleta de Tecidos e Órgãos/ética , Transplante de Tecido Fetal , Humanos , Técnicas de Reprodução Assistida
18.
Pediatr Res ; 59(4 Pt 2): 13R-20R, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16549543

RESUMO

Stem cells are quickly coming into focus of much biomedical research eventually aiming at the therapeutic applications for various disorders and trauma. It is important, however, to keep in mind the difference between the embryonic stem cells, somatic stem cells and somatic precursor cells when considering potential clinical applications. Here we provide the review of the current status of stem cell field and discuss the potential of therapeutic applications for blood and Immune system disorders, multiple sclerosis, hypoxic-ischemic brain injury and brain tumors. For the complimentary information about various stem cells and their properties we recommend consulting the National Institutes of Health stem cell resources (http://stemcells.nih.gov/info/basics).


Assuntos
Células-Tronco , Adulto , Animais , Embrião de Mamíferos/citologia , Rejeição de Enxerto , Humanos , Neoplasias/patologia , Células-Tronco/citologia , Células-Tronco/metabolismo
19.
J Neurosci Res ; 83(5): 845-56, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16477652

RESUMO

Deciphering the factors that regulate human neural stem cells will greatly aid in their use as models of development and as therapeutic agents. The extracellular matrix (ECM) is a component of stem cell niches in vivo and regulates multiple functions in diverse cell types, yet little is known about its effects on human neural stem/precursor cells (NSPCs). We therefore plated human NSPCs on four different substrates (poly-L-ornithine, fibronectin, laminin, and matrigel) and compared their responses with those of mouse NSPCs. Compared with the other substrates, laminin matrices enhanced NSPC migration, expansion, differentiation into neurons and astrocytes, and elongation of neurites from NSPC-derived neurons. Laminin had a similar spectrum of effects on both human and mouse cells, highlighting the evolutionary conservation of NSPC regulation by this component of the ECM. Flow cytometry revealed that human NSPCs express on their cell surfaces the laminin-binding integrins alpha3, alpha6, alpha7, beta1, and beta4, and function-blocking antibodies to the alpha6 subunit confirmed a role for integrins in laminin-dependent migration of human NSPCs. These results define laminin and its integrin receptors as key regulators of human NSPCs.


Assuntos
Diferenciação Celular/fisiologia , Matriz Extracelular/química , Integrinas/metabolismo , Laminina/metabolismo , Neurônios/citologia , Células-Tronco/citologia , Animais , Astrócitos/citologia , Movimento Celular/fisiologia , Proliferação de Células , Células Cultivadas , Colágeno/metabolismo , Combinação de Medicamentos , Matriz Extracelular/metabolismo , Citometria de Fluxo , Humanos , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica , Camundongos , Peptídeos/metabolismo , Proteoglicanas/metabolismo
20.
Expert Rev Neurother ; 6(2): 153-61, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16466295

RESUMO

As a novel neurotherapeutic strategy, stem cell transplantation has received considerable attention, yet little of this attention has been devoted to the probabilities of success of stem cell therapies for specific neurological disorders. Given the complexities of the cellular organization of the nervous system and the manner in which it is assembled during development, it is unlikely that a cellular replacement strategy will succeed for any but the simplest of neurological disorders in the near future. A general strategy for stem cell transplantation to prevent or minimize neurological disorders is much more likely to succeed. Two broad categories of neurological disease, inherited metabolic disorders and invasive brain tumors, are among the most likely candidates.


Assuntos
Doenças do Sistema Nervoso/terapia , Transplante de Células-Tronco/métodos , Células-Tronco/fisiologia , Animais , Prova Pericial , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA