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

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 159(7): 1603-14, 2014 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-25525878

RESUMEN

Glioblastomas and brain metastases are highly proliferative brain tumors with short survival times. Previously, using (13)C-NMR analysis of brain tumors resected from patients during infusion of (13)C-glucose, we demonstrated that there is robust oxidation of glucose in the citric acid cycle, yet glucose contributes less than 50% of the carbons to the acetyl-CoA pool. Here, we show that primary and metastatic mouse orthotopic brain tumors have the capacity to oxidize [1,2-(13)C]acetate and can do so while simultaneously oxidizing [1,6-(13)C]glucose. The tumors do not oxidize [U-(13)C]glutamine. In vivo oxidation of [1,2-(13)C]acetate was validated in brain tumor patients and was correlated with expression of acetyl-CoA synthetase enzyme 2, ACSS2. Together, the data demonstrate a strikingly common metabolic phenotype in diverse brain tumors that includes the ability to oxidize acetate in the citric acid cycle. This adaptation may be important for meeting the high biosynthetic and bioenergetic demands of malignant growth.


Asunto(s)
Acetato CoA Ligasa/metabolismo , Acetatos/metabolismo , Neoplasias Encefálicas/metabolismo , Ciclo del Ácido Cítrico , Glioblastoma/metabolismo , Acetato CoA Ligasa/genética , Animales , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/secundario , Modelos Animales de Enfermedad , Glioblastoma/patología , Ácido Glutámico/metabolismo , Humanos , Ratones , Metástasis de la Neoplasia/patología
2.
J Neuroinflammation ; 21(1): 161, 2024 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-38915059

RESUMEN

BACKGROUND: Pediatric acute transverse myelitis (ATM) accounts for 20-30% of children presenting with a first acquired demyelinating syndrome (ADS) and may be the first clinical presentation of a relapsing ADS such as multiple sclerosis (MS). B cells have been strongly implicated in the pathogenesis of adult MS. However, little is known about B cells in pediatric MS, and even less so in pediatric ATM. Our lab previously showed that plasmablasts (PB), the earliest B cell subtype producing antibody, are expanded in adult ATM, and that these PBs produce self-reactive antibodies that target neurons. The goal of this study was to examine PB frequency and phenotype, immunoglobulin selection, and B cell receptor reactivity in pediatric patients presenting with ATM to gain insight to B cell involvement in disease. METHODS: We compared the PB frequency and phenotype of 5 pediatric ATM patients and 10 pediatric healthy controls (HC) and compared them to previously reported adult ATM patients using cytometric data. We purified bulk IgG from the plasma samples and cloned 20 recombinant human antibodies (rhAbs) from individual PBs isolated from the blood. Plasma-derived IgG and rhAb autoreactivity was measured by mean fluorescence intensity (MFI) in neurons and astrocytes of murine brain or spinal cord and primary human astrocytes. We determined the potential impact of these rhAbs on astrocyte health by measuring stress and apoptotic response. RESULTS: We found that pediatric ATM patients had a reduced frequency of peripheral blood PB. Serum IgG autoreactivity to neurons in EAE spinal cord was similar in the pediatric ATM patients and HC. However, serum IgG autoreactivity to astrocytes in EAE spinal cord was reduced in pediatric ATM patients compared to pediatric HC. Astrocyte-binding strength of rhAbs cloned from PBs was dependent on somatic hypermutation accumulation in the pediatric ATM cohort, but not HC. A similar observation in predilection for astrocyte binding over neuron binding of individual antibodies cloned from PBs was made in EAE brain tissue. Finally, exposure of human primary astrocytes to these astrocyte-binding antibodies increased astrocytic stress but did not lead to apoptosis. CONCLUSIONS: Discordance in humoral immune responses to astrocytes may distinguish pediatric ATM from HC.


Asunto(s)
Astrocitos , Mielitis Transversa , Humanos , Mielitis Transversa/inmunología , Animales , Femenino , Astrocitos/metabolismo , Astrocitos/inmunología , Niño , Ratones , Masculino , Adolescente , Células Plasmáticas/inmunología , Células Plasmáticas/metabolismo , Autoanticuerpos/inmunología , Autoanticuerpos/sangre , Ratones Endogámicos C57BL , Células Cultivadas , Preescolar , Inmunoglobulina G/inmunología , Inmunoglobulina G/sangre , Médula Espinal/metabolismo , Médula Espinal/inmunología , Médula Espinal/patología
3.
Bioinformatics ; 39(9)2023 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-37624931

RESUMEN

MOTIVATION: As an important player in transcriptome regulation, microRNAs may effectively diffuse somatic mutation impacts to broad cellular processes and ultimately manifest disease and dictate prognosis. Previous studies that tried to correlate mutation with gene expression dysregulation neglected to adjust for the disparate multitudes of false positives associated with unequal sample sizes and uneven class balancing scenarios. RESULTS: To properly address this issue, we developed a statistical framework to rigorously assess the extent of mutation impact on microRNAs in relation to a permutation-based null distribution of a matching sample structure. Carrying out the framework in a pan-cancer study, we ascertained 9008 protein-coding genes with statistically significant mutation impacts on miRNAs. Of these, the collective miRNA expression for 83 genes showed significant prognostic power in nine cancer types. For example, in lower-grade glioma, 10 genes' mutations broadly impacted miRNAs, all of which showed prognostic value with the corresponding miRNA expression. Our framework was further validated with functional analysis and augmented with rich features including the ability to analyze miRNA isoforms; aggregative prognostic analysis; advanced annotations such as mutation type, regulator alteration, somatic motif, and disease association; and instructive visualization such as mutation OncoPrint, Ideogram, and interactive mRNA-miRNA network. AVAILABILITY AND IMPLEMENTATION: The data underlying this article are available in MutMix, at http://innovebioinfo.com/Database/TmiEx/MutMix.php.


Asunto(s)
Glioma , MicroARNs , Humanos , Difusión , MicroARNs/genética , Mutación , ARN Mensajero
4.
Proc Natl Acad Sci U S A ; 110(10): 4009-14, 2013 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-23412337

RESUMEN

Glioblastoma (GB) is the most common and aggressive primary brain malignancy, with poor prognosis and a lack of effective therapeutic options. Accumulating evidence suggests that intratumor heterogeneity likely is the key to understanding treatment failure. However, the extent of intratumor heterogeneity as a result of tumor evolution is still poorly understood. To address this, we developed a unique surgical multisampling scheme to collect spatially distinct tumor fragments from 11 GB patients. We present an integrated genomic analysis that uncovers extensive intratumor heterogeneity, with most patients displaying different GB subtypes within the same tumor. Moreover, we reconstructed the phylogeny of the fragments for each patient, identifying copy number alterations in EGFR and CDKN2A/B/p14ARF as early events, and aberrations in PDGFRA and PTEN as later events during cancer progression. We also characterized the clonal organization of each tumor fragment at the single-molecule level, detecting multiple coexisting cell lineages. Our results reveal the genome-wide architecture of intratumor variability in GB across multiple spatial scales and patient-specific patterns of cancer evolution, with consequences for treatment design.


Asunto(s)
Neoplasias Encefálicas/genética , Glioblastoma/genética , Secuencia de Bases , Aberraciones Cromosómicas , Variaciones en el Número de Copia de ADN , ADN de Neoplasias/genética , Progresión de la Enfermedad , Evolución Molecular , Genes erbB-1 , Genes p16 , Humanos , Filogenia , Transcriptoma
5.
Future Oncol ; 11(12): 1703-6, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26075436

RESUMEN

"Serial tumor sampling, single-cell genomics and quantitative imaging are all available technologies, but their integration into current pathways of care will require a paradigm shift in the clinical management of patients with glioblastoma."


Asunto(s)
Neoplasias Encefálicas/patología , Glioblastoma/patología , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/terapia , Progresión de la Enfermedad , Glioblastoma/genética , Glioblastoma/terapia , Humanos , Terapia Molecular Dirigida , Mutación , Transducción de Señal
6.
Mol Cancer Res ; 21(8): 755-767, 2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-37255362

RESUMEN

The treatment of the most aggressive primary brain tumor in adults, glioblastoma (GBM), is challenging due to its heterogeneous nature, invasive potential, and poor response to chemo- and radiotherapy. As a result, GBM inevitably recurs and only a few patients survive 5 years post-diagnosis. GBM is characterized by extensive phenotypic and genetic heterogeneity, creating a diversified genetic landscape and a network of biological interactions between subclones, ultimately promoting tumor growth and therapeutic resistance. This includes spatial and temporal changes in the tumor microenvironment, which influence cellular and molecular programs in GBM and therapeutic responses. However, dissecting phenotypic and genetic heterogeneity at spatial and temporal levels is extremely challenging, and the dynamics of the GBM microenvironment cannot be captured by analysis of a single tumor sample. In this review, we discuss the current research on GBM heterogeneity, in particular, the utility and potential applications of fluorescence-guided multiple sampling to dissect phenotypic and genetic intra-tumor heterogeneity in the GBM microenvironment, identify tumor and non-tumor cell interactions and novel therapeutic targets in areas that are key for tumor growth and recurrence, and improve the molecular classification of GBM.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Adulto , Humanos , Glioblastoma/patología , Fluorescencia , Neoplasias Encefálicas/patología , Microambiente Tumoral/genética
7.
Mol Cancer Res ; 21(8): 755-767, 2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-37159022

RESUMEN

The treatment of the most aggressive primary brain tumor in adults, glioblastoma (GBM), is challenging due to its heterogeneous nature, invasive potential, and poor response to chemo- and radiotherapy. As a result, GBM inevitably recurs and only a few patients survive 5 years post-diagnosis. GBM is characterized by extensive phenotypic and genetic heterogeneity, creating a diversified genetic landscape and a network of biological interactions between subclones, ultimately promoting tumor growth and therapeutic resistance. This includes spatial and temporal changes in the tumor microenvironment, which influence cellular and molecular programs in GBM and therapeutic responses. However, dissecting phenotypic and genetic heterogeneity at spatial and temporal levels is extremely challenging, and the dynamics of the GBM microenvironment cannot be captured by analysis of a single tumor sample. In this review, we discuss the current research on GBM heterogeneity, in particular, the utility and potential applications of fluorescence-guided multiple sampling to dissect phenotypic and genetic intra-tumor heterogeneity in the GBM microenvironment, identify tumor and non-tumor cell interactions and novel therapeutic targets in areas that are key for tumor growth and the recurrence, and improve the molecular classification of GBM.

8.
Acta Neurochir (Wien) ; 154(6): 951-69; discussion 969, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22527576

RESUMEN

The neurosurgical management of patients with intrinsic glial cancers is one of the most rapidly evolving areas of practice. This has been fuelled by advances in surgical technique not only in cytoreduction but also in drug delivery. Further innovation will depend on a deeper understanding of the biology of the disease and an appreciation of the limitations of current knowledge. Here we review the controversial topic of cancer stem cells applied to glioma to provide neurosurgeons with a working overview. It is now recognised that the adult human brain contains regionally specified cell populations capable of self-renewal that may contribute to tumour growth and maintenance following accumulated mutational change. Tumour cells adapted to maintain growth demonstrate some stem-like characteristics and as such constitute a legitimate therapeutic target. Cellular reprogramming technologies raise the potential of developing stem cells as novel surgical tools to target disease and possibly ameliorate some of the consequences of treatment. Achieving these goals remains a significant challenge to neurosurgical oncologists, not least in challenging how we think about treating brain cancer. This review will briefly examine our understanding of adult stem cells within the brain, the evidence that they contribute to the development of brain tumours as tumour-initiating cells, and the potential implications for therapy. It will also look at the role stem cells may play in the future management of glioma.


Asunto(s)
Neoplasias Encefálicas/etiología , Neoplasias Encefálicas/patología , Glioma/terapia , Trasplante de Células Madre/tendencias , Animales , Neoplasias Encefálicas/terapia , Diferenciación Celular/fisiología , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Glioma/etiología , Glioma/fisiopatología , Humanos , Células Madre Neoplásicas/patología , Trasplante de Células Madre/métodos
9.
Curr Med Chem ; 28(4): 687-711, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-31830881

RESUMEN

Melanoma is the most life-threatening and aggressive class of skin malignancies. The incidence of melanoma has steadily increased. Metastatic melanoma is greatly resistant to standard antimelanoma treatments such as chemotherapy, and the 5-year survival rate of cases with melanoma who have a metastatic form of the disease is less than 10%. The contributing role of apoptosis, angiogenesis and autophagy in the pathophysiology of melanoma has been previously demonstrated. Thus, it is extremely urgent to search for complementary therapeutic approaches that could enhance the quality of life of subjects and reduce treatment resistance and adverse effects. Resveratrol, known as a polyphenol component present in grapes and some plants, has anti-cancer properties due to its function as an apoptosis inducer in tumor cells, and anti-angiogenic agent to prevent metastasis. However, more clinical trials should be conducted to prove resveratrol efficacy. Herein, for the first time, we summarize the current knowledge of anti-cancerous activities of resveratrol in melanoma.


Asunto(s)
Melanoma , Neoplasias Cutáneas , Estilbenos , Apoptosis , Humanos , Melanoma/tratamiento farmacológico , Calidad de Vida , Resveratrol/farmacología , Resveratrol/uso terapéutico , Neoplasias Cutáneas/tratamiento farmacológico , Estilbenos/farmacología , Estilbenos/uso terapéutico
10.
Future Oncol ; 11(20): 2851, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26358184
11.
Cell Rep ; 30(8): 2489-2500.e5, 2020 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-32101730

RESUMEN

Emerging evidence suggests that crosstalk between glioma cells and the brain microenvironment may influence brain tumor growth. To date, known reciprocal interactions among these cells have been limited to the release of paracrine factors. Combining a genetic strategy with longitudinal live imaging, we find that individual gliomas communicate with distinct sets of non-glioma cells, including glial cells, neurons, and vascular cells. Transfer of genetic material is achieved mainly through extracellular vesicles (EVs), although cell fusion also plays a minor role. We further demonstrate that EV-mediated communication leads to the increase of synaptic activity in neurons. Blocking EV release causes a reduction of glioma growth in vivo. Our findings indicate that EV-mediated interaction between glioma cells and non-glioma brain cells alters the tumor microenvironment and contributes to glioma development.


Asunto(s)
Neoplasias Encefálicas/patología , Encéfalo/patología , Comunicación Celular , Vesículas Extracelulares/metabolismo , Glioma/patología , Animales , Astrocitos/patología , Encéfalo/fisiopatología , Neoplasias Encefálicas/fisiopatología , Fusión Celular , Línea Celular Tumoral , ADN de Neoplasias/genética , Fenómenos Electrofisiológicos , Vesículas Extracelulares/ultraestructura , Glioma/fisiopatología , Humanos , Ratones Endogámicos C57BL , Ratones Desnudos , Neuronas/patología , Imagen de Lapso de Tiempo
12.
Neuro Oncol ; 21(6): 719-729, 2019 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-30590711

RESUMEN

BACKGROUND: Expression of neuron-glial antigen 2 (NG2) identifies an aggressive malignant phenotype in glioblastoma (GBM). Mouse models have implicated NG2 in the genesis, evolution, and maintenance of glial cancers and have highlighted potential interactions between NG2 and epidermal growth factor receptor (EGFR). However, it is unknown whether the lineage relationship of NG2+ and NG2- cells follows a hierarchical or stochastic mode of growth. Furthermore, the interaction between NG2 and EGFR signaling in human GBM is also unclear. METHODS: Single GBM NG2+ and NG2- cells were studied longitudinally to assess lineage relationships. Short hairpin RNA knockdown of NG2 was used to assess the mechanistic role of NG2 in human GBM cells. NG2+ and NG2- cells and NG2 knockdown (NG2-KD) and wild type (NG2-WT) cells were analyzed for differential effects on EGFR signaling. RESULTS: Expression of NG2 endows an aggressive phenotype both at single cell and population levels. Progeny derived from single GBM NG2- or GBM NG2+ cells consistently establish phenotypic equilibrium, indicating the absence of a cellular hierarchy. NG2 knockdown reduces proliferation, and mice grafted with NG2-KD survive longer than controls. Finally, NG2 promotes EGFR signaling and is associated with EGFR expression. CONCLUSIONS: These data support a dynamic evolution in which a bidirectional relationship exists between GBM NG2+ and GBM NG2- cells. Such findings have implications for understanding phenotypic heterogeneity, the emergence of resistant disease, and developing novel therapeutics.


Asunto(s)
Antígenos/metabolismo , Biomarcadores de Tumor/metabolismo , Neoplasias Encefálicas/patología , Regulación Neoplásica de la Expresión Génica , Glioblastoma/patología , Proteoglicanos/metabolismo , Animales , Antígenos/genética , Apoptosis , Biomarcadores de Tumor/genética , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Carcinogénesis , Proliferación Celular , Receptores ErbB/genética , Receptores ErbB/metabolismo , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Ratones , Proteoglicanos/genética , Transducción de Señal , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
13.
J Neuropathol Exp Neurol ; 78(12): 1081-1088, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31589317

RESUMEN

Clear cell, microcytic, and angiomatous meningiomas are 3 vasculature-rich variants with overlapping morphological features but different prognostic and treatment implications. Distinction between them is not always straightforward. We compared the expression patterns of the hypoxia marker carbonic anhydrase IX (CA-IX) in meningiomas with predominant clear cell (n = 15), microcystic (n = 9), or angiomatous (n = 11) morphologies, as well as 117 cases of other World Health Organization recognized histological meningioma variants. Immunostaining for SMARCE1 protein, whose loss-of-function has been associated with clear cell meningiomas, was performed on all clear cell meningiomas, and selected variants of meningiomas as controls. All clear cell meningiomas showed absence of CA-IX expression and loss of nuclear SMARCE1 expression. All microcystic and angiomatous meningiomas showed diffuse CA-IX immunoreactivity and retained nuclear SMARCE1 expression. In other meningioma variants, CA-IX was expressed in a hypoxia-restricted pattern and was highly associated with atypical features such as necrosis, small cell change, and focal clear cell change. In conclusion, CA-IX may serve as a useful diagnostic marker in differentiating clear cell, microcystic, and angiomatous meningiomas.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Anhidrasa Carbónica IX/metabolismo , Neoplasias Meníngeas/enzimología , Meningioma/enzimología , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/metabolismo , Encéfalo/patología , Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Unión al ADN/metabolismo , Femenino , Humanos , Masculino , Neoplasias Meníngeas/diagnóstico , Neoplasias Meníngeas/patología , Meningioma/diagnóstico , Meningioma/patología , Persona de Mediana Edad , Supervivencia sin Progresión
14.
Neuro Oncol ; 18(12): 1644-1655, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27286795

RESUMEN

BACKGROUND: Amplification of the epidermal growth factor receptor (EGFR) and its mutant EGFRvIII are among the most common genetic alterations in glioblastoma (GBM), the most frequent and most aggressive primary brain tumor. METHODS: In the present work, we analyzed the clonal evolution of these major EGFR aberrations in a small cohort of GBM patients using a unique surgical multisampling technique. Furthermore, we overexpressed both receptors separately and together in 2 patient-derived GBM stem cell lines (GSCs) to analyze their functions in vivo in orthotopic xenograft models. RESULTS: In human GBM biopsies, we identified EGFR amplification as an early event because EGFRvIII mutations emerge from intratumoral heterogeneity later in tumor development. To investigate the biological relevance of this distinct developmental pattern, we established experimental model systems. In these models, EGFR+ tumor cells showed activation of classical downstream signaling pathways upon EGF stimulation and displayed enhanced invasive growth without evidence of angiogenesis in vivo. In contrast, EGFRvIII+ tumors were driven by activation of the prototypical Src family kinase c-Src that promoted VEGF secretion leading to angiogenic tumor growth. CONCLUSIONS: The presented work shows that sequential EGFR amplification and EGFRvIII mutations might represent concerted evolutionary events that drive the aggressive nature of GBM by promoting invasion and angiogenesis via distinct signaling pathways. In particular, c-SRC may be an attractive therapeutic target for tumors harboring EGFRvIII as we identified this protein specifically mediating angiogenic tumor growth downstream of EGFRvIII.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Receptores ErbB/genética , Receptores ErbB/metabolismo , Glioblastoma/metabolismo , Neovascularización Patológica/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Encéfalo/diagnóstico por imagen , Encéfalo/patología , Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Evolución Molecular , Glioblastoma/diagnóstico por imagen , Glioblastoma/genética , Glioblastoma/patología , Humanos , Imagen Multimodal , Mutación , Invasividad Neoplásica , Análisis de Supervivencia , Regulación hacia Arriba
15.
CNS Oncol ; 4(6): 399-410, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26497327

RESUMEN

Glioblastoma demonstrates imaging features of intratumor heterogeneity that result from underlying heterogeneous biological properties. This stems from variations in cellular behavior that result from genetic mutations that either drive, or are driven by, heterogeneous microenvironment conditions. Among all imaging methods available, only T1-weighted contrast-enhancing and T2-weighted fluid-attenuated inversion recovery are used in standard clinical glioblastoma assessment and monitoring. Advanced imaging modalities are still considered emerging techniques as appropriate end points and robust methodologies are missing from clinical trials. Discovering how these images specifically relate to the underlying tumor biology may aid in improving quality of clinical trials and understanding the factors involved in regional responses to treatment, including variable drug uptake and effect of radiotherapy. Upon validation and standardization of emerging MR techniques, providing information based on the underlying tumor biology, these images may allow for clinical decision-making that is tailored to an individual's response to treatment.


Asunto(s)
Neoplasias Encefálicas/patología , Glioblastoma/patología , Imagen Multimodal/métodos , Animales , Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Humanos
16.
Stem Cells Transl Med ; 4(7): 822-31, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26019225

RESUMEN

UNLABELLED: : Recent research has focused on the hypothesis that the growth and regeneration of glioblastoma (GB) is sustained by a subpopulation of self-renewing stem-like cells. This has led to the prediction that molecular markers for cancer stem cells in GB may provide a treatment target. One candidate marker is CD15: we wanted to determine if CD15 represented a credible stem cell marker in GB. We first demonstrated that CD15-positive (CD15+) cells were less proliferative than their CD15-negative (CD15-) counterparts in 10 patient GB tumors. Next we compared the proliferative activity of CD15+ and CD15- cells in vitro using tumor-initiating primary GB cell lines (TICs) and found no difference in proliferative behavior. Furthermore, TICs sorted for CD15+ and CD15- were not significantly different cytogenetically or in terms of gene expression profile. Sorted single CD15+ and CD15- cells were equally capable of reconstituting a heterogeneous population containing both CD15+ and CD15- cells over time, and both CD15+ and CD15- cells were able to generate tumors in vivo. No difference was found in the phenotypic or genomic behavior of CD15+ cells compared with CD15- cells from the same patient. Moreover, we found that in vitro, cells were able to interconvert between the CD15+ and CD15- states. Our data challenge the utility of CD15 as a cancer stem cell marker. SIGNIFICANCE: The data from this study contribute to the ongoing debate about the role of cancer stem cells in gliomagenesis. Results showed that CD15, a marker previously thought to be a cancer stem-like marker in glioblastoma, could not isolate a phenotypically or genetically distinct population. Moreover, isolated CD15-positive and -negative cells were able to generate mixed populations of glioblastoma cells in vitro.

17.
Stem Cell Reports ; 4(1): 7-15, 2015 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-25533637

RESUMEN

Glioblastoma (GBM) is a lethal malignancy whose clinical intransigence has been linked to extensive intraclonal genetic and phenotypic diversity and the common emergence of therapeutic resistance. This interpretation embodies the implicit assumption that cancer stem cells or tumor-propagating cells are themselves genetically and functionally diverse. To test this, we screened primary GBM tumors by SNP array to identify copy number alterations (a minimum of three) that could be visualized in single cells by multicolor fluorescence in situ hybridization. Interrogation of neurosphere-derived cells (from four patients) and cells derived from secondary transplants of these same cells in NOD-SCID mice allowed us to infer the clonal and phylogenetic architectures. Whole-exome sequencing and single-cell genetic analysis in one case revealed a more complex clonal structure. This proof-of-principle experiment revealed that subclones in each GBM had variable regenerative or stem cell activity, and highlighted genetic alterations associated with more competitive propagating activity in vivo.


Asunto(s)
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Variación Genética , Glioblastoma/genética , Glioblastoma/metabolismo , Fenotipo , Animales , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Variaciones en el Número de Copia de ADN , Progresión de la Enfermedad , Estudio de Asociación del Genoma Completo , Genómica , Glioblastoma/patología , Xenoinjertos , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Hibridación Fluorescente in Situ , Ratones , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Polimorfismo de Nucleótido Simple , Análisis de la Célula Individual
18.
Nanoscale ; 6(18): 10865-73, 2014 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-25117686

RESUMEN

Glioblastoma multiforme (GBM) remains the most aggressive and challenging brain tumour to treat. We report the first successful chemo-radiotherapy on patient derived treatment resistant GBM cells using a cisplatin-tethered gold nanosphere. After intracellular uptake, the nanosphere effects DNA damage which initiates caspase-mediated apoptosis in those cells. In the presence of radiation, both gold and platinum of cisplatin, serve as high atomic number radiosensitizers leading to the emission of ionizing photoelectrons and Auger electrons. This resulted in enhanced synergy between cisplatin and radiotherapy mediated cytotoxicity, and photo/Auger electron mediated radiosensitisation leading to complete ablation of the tumour cells in an in vitro model system. This study demonstrates the potential of designed nanoparticles to target aggressive cancers in the patient derived cell lines providing a platform to move towards treatment strategies.


Asunto(s)
Cisplatino/química , Oro/química , Nanosferas/química , Fármacos Sensibilizantes a Radiaciones/química , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Caspasas/metabolismo , Línea Celular Tumoral , Cisplatino/toxicidad , Daño del ADN/efectos de los fármacos , Daño del ADN/efectos de la radiación , Rayos gamma , Glioblastoma/metabolismo , Glioblastoma/patología , Histonas/metabolismo , Humanos , Polietileneimina/química , Fármacos Sensibilizantes a Radiaciones/toxicidad
20.
PLoS One ; 8(10): e75945, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24146797

RESUMEN

Glioblastoma multiforme (GBM) is the most common primary malignant adult brain tumor and is associated with poor survival. Recently, stem-like cell populations have been identified in numerous malignancies including GBM. To identify genes whose expression is changed with differentiation, we compared transcript profiles from a GBM oncosphere line before and after differentiation. Bioinformatic analysis of the gene expression profiles identified podocalyxin-like protein (PODXL), a protein highly expressed in human embryonic stem cells, as a potential marker of undifferentiated GBM stem-like cells. The loss of PODXL expression upon differentiation of GBM stem-like cell lines was confirmed by quantitative real-time PCR and flow cytometry. Analytical flow cytometry of numerous GBM oncosphere lines demonstrated PODXL expression in all lines examined. Knockdown studies and flow cytometric cell sorting experiments demonstrated that PODXL is involved in GBM stem-like cell proliferation and oncosphere formation. Compared to PODXL-negative cells, PODXL-positive cells had increased expression of the progenitor/stem cell markers Musashi1, SOX2, and BMI1. Finally, PODXL expression directly correlated with increasing glioma grade and was a marker for poor outcome in patients with GBM. In summary, we have demonstrated that PODXL is expressed in GBM stem-like cells and is involved in cell proliferation and oncosphere formation. Moreover, high PODXL expression correlates with increasing glioma grade and decreased overall survival in patients with GBM.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias Encefálicas/genética , Regulación Neoplásica de la Expresión Génica , Glioblastoma/genética , Células Madre Neoplásicas/metabolismo , Sialoglicoproteínas/genética , Adulto , Biomarcadores de Tumor/metabolismo , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/mortalidad , Neoplasias Encefálicas/patología , Diferenciación Celular , Línea Celular Tumoral , Proliferación Celular , Citometría de Flujo , Técnicas de Silenciamiento del Gen , Glioblastoma/metabolismo , Glioblastoma/mortalidad , Glioblastoma/patología , Humanos , Clasificación del Tumor , Células Madre Neoplásicas/patología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Complejo Represivo Polycomb 1/genética , Complejo Represivo Polycomb 1/metabolismo , Pronóstico , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Sialoglicoproteínas/metabolismo , Esferoides Celulares/metabolismo , Esferoides Celulares/patología , Análisis de Supervivencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA