Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Front Neurosci ; 17: 1217596, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37588515

RESUMEN

Neural precursors generate neurons in the embryonic brain and in restricted niches of the adult brain in a process called neurogenesis. The precise control of cell proliferation and differentiation in time and space required for neurogenesis depends on sophisticated orchestration of gene transcription in neural precursor cells. Much progress has been made in understanding the transcriptional regulation of neurogenesis, which relies on dose- and context-dependent expression of specific transcription factors that regulate the maintenance and proliferation of neural progenitors, followed by their differentiation into lineage-specified cells. Here, we review some of the most widely studied neurogenic transcription factors in the embryonic cortex and neurogenic niches in the adult brain. We compare functions of these transcription factors in embryonic and adult neurogenesis, highlighting biochemical, developmental, and cell biological properties. Our goal is to present an overview of transcriptional regulation underlying neurogenesis in the developing cerebral cortex and in the adult brain.

2.
Cancer Lett ; 571: 216349, 2023 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-37579831

RESUMEN

Glioblastoma is the most lethal brain cancer in adults. These incurable tumors are characterized by profound heterogeneity, therapy resistance, and diffuse infiltration. These traits have been linked to cancer stem cells, which are important for glioblastoma tumor progression and recurrence. The fibroblast growth factor receptor 1 (FGFR1) signaling pathway is a known regulator of therapy resistance and cancer stemness in glioblastoma. FGFR1 expression shows intertumoral heterogeneity and higher FGFR1 expression is associated with a significantly poorer survival in glioblastoma patients. The role of FGFR1 in tumor invasion has been studied in many cancers, but whether and how FGFR1 mediates glioblastoma invasion remains to be determined. Here, we investigated the distribution and functional relevance of FGFR1 and FGFR2 in human glioblastoma xenograft models. We found FGFR1, but not FGFR2, expressed in invasive glioblastoma cells. Loss of FGFR1, but not FGFR2, significantly reduced cell migration in vitro and tumor invasion in human glioblastoma xenografts. Comparative analysis of RNA-sequencing data of FGFR1 and FGFR2 knockdown glioblastoma cells revealed a FGFR1-specific gene regulatory network associated with tumor invasion. Our study reveals new gene candidates linked to FGFR1-mediated glioblastoma invasion.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos , Humanos , Neoplasias Encefálicas/genética , Glioblastoma/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , ARN , Transducción de Señal , Animales
3.
Viruses ; 15(5)2023 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-37243172

RESUMEN

Glioblastoma (GBM) is the most common and aggressive adult brain cancer with an average survival rate of around 15 months in patients receiving standard treatment. Oncolytic adenovirus expressing therapeutic transgenes represent a promising alternative treatment for GBM. Of the many human adenoviral serotypes described to date, adenovirus 5 (HAdV-C5) has been the most utilised clinically and experimentally. However, the use of Ad5 as an anti-cancer agent may be hampered by naturally high seroprevalence rates to HAdV-C5 coupled with the infection of healthy cells via native receptors. To explore whether alternative natural adenoviral tropisms are better suited to GBM therapeutics, we pseudotyped an HAdV-C5-based platform using the fibre knob protein from alternative serotypes. We demonstrate that the adenoviral entry receptor coxsackie, adenovirus receptor (CAR) and CD46 are highly expressed by both GBM and healthy brain tissue, whereas Desmoglein 2 (DSG2) is expressed at a low level in GBM. We demonstrate that adenoviral pseudotypes, engaging CAR, CD46 and DSG2, effectively transduce GBM cells. However, the presence of these receptors on non-transformed cells presents the possibility of off-target effects and therapeutic transgene expression in healthy cells. To enhance the specificity of transgene expression to GBM, we assessed the potential for tumour-specific promoters hTERT and survivin to drive reporter gene expression selectively in GBM cell lines. We demonstrate tight GBM-specific transgene expression using these constructs, indicating that the combination of pseudotyping and tumour-specific promoter approaches may enable the development of efficacious therapies better suited to GBM.


Asunto(s)
Infecciones por Adenoviridae , Neoplasias Encefálicas , Glioblastoma , Humanos , Glioblastoma/genética , Glioblastoma/terapia , Estudios Seroepidemiológicos , Línea Celular Tumoral , Receptores Virales/genética , Adenoviridae/genética , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/terapia , Vectores Genéticos/genética
4.
Cells ; 11(4)2022 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-35203370

RESUMEN

Adult neural stem cells (aNSCs) are the source for the continuous production of new neurons throughout life. This so-called adult neurogenesis has been extensively studied; the intermediate cellular stages are well documented. Recent discoveries have raised new controversies in the field, such as the notion that progenitor cells hold similar self-renewal potential as stem cells, or whether different types of aNSCs exist. Here, we discuss evidence for heterogeneity of aNSCs, including short-term and long-term self-renewing aNSCs, regional and temporal differences in aNSC function, and single cell transcriptomics. Reviewing various genetic mouse models used for targeting aNSCs and lineage tracing, we consider potential lineage relationships between Ascl1-, Gli1-, and Nestin-targeted aNSCs. We present a multidimensional model of adult neurogenesis that incorporates recent findings and conclude that stemness is a phenotype, a state of properties that can change with time, rather than a cell property, which is static and immutable. We argue that singular aNSCs do not exist.


Asunto(s)
Células Madre Adultas , Células-Madre Neurales , Animales , Ratones , Neurogénesis/genética , Neuronas
5.
Methods Mol Biol ; 2389: 95-102, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34558005

RESUMEN

Adult neural precursor cells are an integral part of the brain and have been a focus of intense research for half a century. Even though adult neural stem/progenitor cells in the human brain have been identified over 20 years ago, progress in this area has been slow, and the existence of lifelong neurogenesis in humans is still debated. Remarkable species differences exist between humans and rodents in astrocyte development and diversity, suggesting similar differences may exist in neural stem/progenitor cells and underscoring the need for further research in human tissue.This chapter provides a guideline for dissociation of adult human brain tissue from biopsy or autopsy specimens. While protocols for subsequent culturing of neural precursors are included, the main focus is the preparation of a suspension of viable single cells that may also be useful in other experimental paradigms, such as genomic profiling.


Asunto(s)
Células Madre Adultas , Células-Madre Neurales , Adulto , Encéfalo , Separación Celular , Células Cultivadas , Humanos , Neurogénesis , Neuronas
6.
Cell Rep ; 36(8): 109588, 2021 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-34433050

RESUMEN

Radial glia-like (RGL) stem cells persist in the adult mammalian hippocampus, where they generate new neurons and astrocytes throughout life. The process of adult neurogenesis is well documented, but cell-autonomous factors regulating neuronal and astroglial differentiation are incompletely understood. Here, we evaluate the functions of the transcription factor zinc-finger E-box binding homeobox 1 (ZEB1) in adult hippocampal RGL cells using a conditional-inducible mouse model. We find that ZEB1 is necessary for self-renewal of active RGL cells. Genetic deletion of Zeb1 causes a shift toward symmetric cell division that consumes the RGL cell and generates pro-neuronal progenies, resulting in an increase of newborn neurons and a decrease of newly generated astrocytes. We identify ZEB1 as positive regulator of the ets-domain transcription factor ETV5 that is critical for asymmetric division.


Asunto(s)
Autorrenovación de las Células/fisiología , Hipocampo/metabolismo , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo , Animales , Diferenciación Celular/genética , Células Ependimogliales/metabolismo , Transición Epitelial-Mesenquimal/fisiología , Hipocampo/efectos de los fármacos , Humanos , Ratones , Neurogénesis/fisiología , Neuronas/metabolismo
7.
Analyst ; 146(7): 2277-2291, 2021 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-33617612

RESUMEN

Quantitative hyperspectral coherent Raman scattering microscopy merges imaging with spectroscopy and utilises quantitative data analysis algorithms to extract physically meaningful chemical components, spectrally and spatially-resolved, with sub-cellular resolution. This label-free non-invasive method has the potential to significantly advance our understanding of the complexity of living multicellular systems. Here, we have applied an in-house developed hyperspectral coherent anti-Stokes Raman scattering (CARS) microscope, combined with a quantitative data analysis pipeline, to imaging living mouse liver organoids as well as fixed mouse brain tissue sections xenografted with glioblastoma cells. We show that the method is capable of discriminating different cellular sub-populations, on the basis of their chemical content which is obtained from an unsupervised analysis, i.e. without prior knowledge. Specifically, in the organoids, we identify sub-populations of cells at different phases in the cell cycle, while in the brain tissue, we distinguish normal tissue from cancer cells, and, notably, tumours derived from transplanted cancer stem cells versus non-stem glioblastoma cells. The ability of the method to identify different sub-populations was validated by correlative fluorescence microscopy using fluorescent protein markers. These examples expand the application portfolio of quantitative chemical imaging by hyperspectral CARS microscopy to multicellular systems of significant biomedical relevance, pointing the way to new opportunities in non-invasive disease diagnostics.


Asunto(s)
Glioblastoma , Espectrometría Raman , Algoritmos , Animales , Glioblastoma/diagnóstico por imagen , Ratones , Microscopía Fluorescente , Proteínas
8.
J Clin Invest ; 131(1)2021 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-33108352

RESUMEN

Zeb1, a zinc finger E-box binding homeobox epithelial-mesenchymal transition (EMT) transcription factor, confers properties of "stemness," such as self-renewal, in cancer. Yet little is known about the function of Zeb1 in adult stem cells. Here, we used the hematopoietic system as a well-established paradigm of stem cell biology to evaluate Zeb1-mediated regulation of adult stem cells. We employed a conditional genetic approach using the Mx1-Cre system to specifically knock out (KO) Zeb1 in adult hematopoietic stem cells (HSCs) and their downstream progeny. Acute genetic deletion of Zeb1 led to rapid-onset thymic atrophy and apoptosis-driven loss of thymocytes and T cells. A profound cell-autonomous self-renewal defect and multilineage differentiation block were observed in Zeb1-KO HSCs. Loss of Zeb1 in HSCs activated transcriptional programs of deregulated HSC maintenance and multilineage differentiation genes and of cell polarity consisting of cytoskeleton-, lipid metabolism/lipid membrane-, and cell adhesion-related genes. Notably, epithelial cell adhesion molecule (EpCAM) expression was prodigiously upregulated in Zeb1-KO HSCs, which correlated with enhanced cell survival, diminished mitochondrial metabolism, ribosome biogenesis, and differentiation capacity and an activated transcriptomic signature associated with acute myeloid leukemia (AML) signaling. ZEB1 expression was downregulated in AML patients, and Zeb1 KO in the malignant counterparts of HSCs - leukemic stem cells (LSCs) - accelerated MLL-AF9- and Meis1a/Hoxa9-driven AML progression, implicating Zeb1 as a tumor suppressor in AML LSCs. Thus, Zeb1 acts as a transcriptional regulator in hematopoiesis, critically coordinating HSC self-renewal, apoptotic, and multilineage differentiation fates required to suppress leukemic potential in AML.


Asunto(s)
Células Madre Hematopoyéticas/metabolismo , Leucemia Mieloide Aguda/metabolismo , Células Madre Neoplásicas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo , Animales , Eliminación de Gen , Células Madre Hematopoyéticas/patología , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Ratones , Ratones Noqueados , Células Madre Neoplásicas/patología , Proteínas Supresoras de Tumor/genética , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/genética
9.
Front Oncol ; 10: 574011, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33324551

RESUMEN

Glioblastoma is the most common primary brain tumor in adults with poor overall outcome and 5-year survival of less than 5%. Treatment has not changed much in the last decade or so, with surgical resection and radio/chemotherapy being the main options. Glioblastoma is highly heterogeneous and frequently becomes treatment-resistant due to the ability of glioblastoma cells to adopt stem cell states facilitating tumor recurrence. Therefore, there is an urgent need for novel therapeutic strategies. The ubiquitin system, in particular E3 ubiquitin ligases and deubiquitinating enzymes, have emerged as a promising source of novel drug targets. In addition to conventional small molecule drug discovery approaches aimed at modulating enzyme activity, several new and exciting strategies are also being explored. Among these, PROteolysis TArgeting Chimeras (PROTACs) aim to harness the endogenous protein turnover machinery to direct therapeutically relevant targets, including previously considered "undruggable" ones, for proteasomal degradation. PROTAC and other strategies targeting the ubiquitin proteasome system offer new therapeutic avenues which will expand the drug development toolboxes for glioblastoma. This review will provide a comprehensive overview of E3 ubiquitin ligases and deubiquitinating enzymes in the context of glioblastoma and their involvement in core signaling pathways including EGFR, TGF-ß, p53 and stemness-related pathways. Finally, we offer new insights into how these ubiquitin-dependent mechanisms could be exploited therapeutically for glioblastoma.

10.
Cell Mol Life Sci ; 77(24): 5101-5119, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32506168

RESUMEN

The metabolic complexity and flexibility commonly observed in brain tumors, especially glioblastoma, is fundamental for their development and progression. The ability of tumor cells to modify their genetic landscape and adapt metabolically, subverts therapeutic efficacy, and inevitably instigates therapeutic resistance. To overcome these challenges and develop effective therapeutic strategies targeting essential metabolic processes, it is necessary to identify the mechanisms underlying heterogeneity and define metabolic preferences and liabilities of malignant cells. In this review, we will discuss metabolic diversity in brain cancer and highlight the role of cancer stem cells in regulating metabolic heterogeneity. We will also highlight potential therapeutic modalities targeting metabolic vulnerabilities and examine how intercellular metabolic signaling can shape the tumor microenvironment.


Asunto(s)
Neoplasias Encefálicas/genética , Heterogeneidad Genética , Glioblastoma/genética , Metabolismo/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Glioblastoma/metabolismo , Glioblastoma/patología , Glucólisis/genética , Humanos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Transducción de Señal/genética , Microambiente Tumoral
11.
Expert Opin Ther Targets ; 24(4): 311-318, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32174197

RESUMEN

Introduction: Fibroblast growth factors (FGFs) are key mitogens in tissue homeostasis and cancer. FGF2 regulates self-renewal of multiple stem-cell types, is widely used in stem cell culture paradigms and has been adopted for cultivating the growth of cancer stem cells ex vivo. Research has shed light on the functions of FGF2 in brain tumors, particularly malignant glioma, and this has demonstrated that FGF2 increases self-renewal of glioblastoma stem cells.Areas covered: This review examines the potential targeting of FGF2 signaling as a possible treatment avenue for glioblastoma. The expression of FGF ligands and the FGFR family of receptor tyrosine kinases in the normal brain and in glioblastoma is described. Moreover, the paper sheds light on FGF/FGFR signaling, including the function of heparin/heparan sulfate proteoglycans in facilitating FGF signaling. We speculate on potential avenues for the therapeutic targeting of the FGF2-FGF receptor signaling axis in glioblastoma and the associated challenges envisioned with these approaches.Expert opinion: Precision targeting of FGF/FGFR signaling could improve prospective glioblastoma therapeutics and moderate adverse effects. Shrewd development of experimental models and FGF2 inhibitors could provide a 'pharmacological toolbox' for targeting diverse ligand/receptor combinations.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Factor 2 de Crecimiento de Fibroblastos/genética , Glioblastoma/tratamiento farmacológico , Animales , Antineoplásicos/farmacología , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Glioblastoma/genética , Glioblastoma/patología , Humanos , Terapia Molecular Dirigida , Células Madre Neoplásicas , Transducción de Señal/efectos de los fármacos
12.
Mol Cell Oncol ; 7(1): 1684787, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31993498

RESUMEN

Identification of targetable mechanisms that maintain glioblastoma cancer stem cells (CSCs) remain a priority. Our study reveals a new mechanism by which a disintegrin and metalloproteinase domain-like protein decysin 1 promotes CSC maintenance through the activation of a fibroblast growth factor autocrine signaling loop, which can be blocked pharmacologically.

13.
Cancer Discov ; 9(11): 1574-1589, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31434712

RESUMEN

Glioblastomas (GBM) are lethal brain tumors where poor outcome is attributed to cellular heterogeneity, therapeutic resistance, and a highly infiltrative nature. These characteristics are preferentially linked to GBM cancer stem cells (GSC), but how GSCs maintain their stemness is incompletely understood and the subject of intense investigation. Here, we identify a novel signaling loop that induces and maintains GSCs consisting of an atypical metalloproteinase, ADAMDEC1, secreted by GSCs. ADAMDEC1 rapidly solubilizes FGF2 to stimulate FGFR1 expressed on GSCs. FGFR1 signaling induces upregulation of ZEB1 via ERK1/2 that regulates ADAMDEC1 expression through miR-203, creating a positive feedback loop. Genetic or pharmacologic targeting of components of this axis attenuates self-renewal and tumor growth. These findings reveal a new signaling axis for GSC maintenance and highlight ADAMDEC1 and FGFR1 as potential therapeutic targets in GBM. SIGNIFICANCE: Cancer stem cells (CSC) drive tumor growth in many cancers including GBM. We identified a novel sheddase, ADAMDEC1, which initiates an FGF autocrine loop to promote stemness in CSCs. This loop can be targeted to reduce GBM growth.This article is highlighted in the In This Issue feature, p. 1469.


Asunto(s)
Proteínas ADAM/metabolismo , Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Células Madre Neoplásicas/metabolismo , Transducción de Señal , Animales , Neoplasias Encefálicas/genética , Línea Celular Tumoral , Retroalimentación Fisiológica , Femenino , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Glioblastoma/genética , Humanos , MicroARNs/genética , Trasplante de Neoplasias , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo
14.
Cells ; 8(7)2019 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-31337028

RESUMEN

Glioblastoma is the most lethal brain cancer in adults, with no known cure. This cancer is characterized by a pronounced genetic heterogeneity, but aberrant activation of receptor tyrosine kinase signaling is among the most frequent molecular alterations in glioblastoma. Somatic mutations of fibroblast growth factor receptors (FGFRs) are rare in these cancers, but many studies have documented that signaling through FGFRs impacts glioblastoma progression and patient survival. Small-molecule inhibitors of FGFR tyrosine kinases are currently being trialed, underlining the therapeutic potential of blocking this signaling pathway. Nevertheless, a comprehensive overview of the state of the art of the literature on FGFRs in glioblastoma is lacking. Here, we review the evidence for the biological functions of FGFRs in glioblastoma, as well as pharmacological approaches to targeting these receptors.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/fisiología , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/fisiología , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/fisiología , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/fisiología , Progresión de la Enfermedad , Humanos , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/química , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/química , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/química , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/química
15.
EMBO J ; 37(23)2018 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-30322894

RESUMEN

Metabolic reprogramming has been described in rapidly growing tumors, which are thought to mostly contain fast-cycling cells (FCCs) that have impaired mitochondrial function and rely on aerobic glycolysis. Here, we characterize the metabolic landscape of glioblastoma (GBM) and explore metabolic specificities as targetable vulnerabilities. Our studies highlight the metabolic heterogeneity in GBM, in which FCCs harness aerobic glycolysis, and slow-cycling cells (SCCs) preferentially utilize mitochondrial oxidative phosphorylation for their functions. SCCs display enhanced invasion and chemoresistance, suggesting their important role in tumor recurrence. SCCs also demonstrate increased lipid contents that are specifically metabolized under glucose-deprived conditions. Fatty acid transport in SCCs is targetable by pharmacological inhibition or genomic deletion of FABP7, both of which sensitize SCCs to metabolic stress. Furthermore, FABP7 inhibition, whether alone or in combination with glycolysis inhibition, leads to overall increased survival. Our studies reveal the existence of GBM cell subpopulations with distinct metabolic requirements and suggest that FABP7 is central to lipid metabolism in SCCs and that targeting FABP7-related metabolic pathways is a viable therapeutic strategy.


Asunto(s)
Resistencia a Antineoplásicos , Ácidos Grasos/metabolismo , Glioblastoma/metabolismo , Glucólisis , Mitocondrias/metabolismo , Fosforilación Oxidativa , Animales , Línea Celular Tumoral , Proteína de Unión a los Ácidos Grasos 7/metabolismo , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Mitocondrias/patología , Proteínas de Neoplasias/metabolismo , Proteínas Supresoras de Tumor/metabolismo
16.
Proc Natl Acad Sci U S A ; 115(37): E8765-E8774, 2018 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-30150378

RESUMEN

Huntington disease (HD) is an autosomal dominant neurodegenerative disorder caused by expanded CAG repeats in the huntingtin gene (HTT). Although mutant HTT is expressed during embryonic development and throughout life, clinical HD usually manifests later in adulthood. A number of studies document neurodevelopmental changes associated with mutant HTT, but whether these are reversible under therapy remains unclear. Here, we identify very early behavioral, molecular, and cellular changes in preweaning transgenic HD rats and mice. Reduced ultrasonic vocalization, loss of prepulse inhibition, and increased risk taking are accompanied by disturbances of dopaminergic regulation in vivo, reduced neuronal differentiation capacity in subventricular zone stem/progenitor cells, and impaired neuronal and oligodendrocyte differentiation of mouse embryo-derived neural stem cells in vitro. Interventional treatment of this early phenotype with the histone deacetylase inhibitor (HDACi) LBH589 led to significant improvement in behavioral changes and markers of dopaminergic neurotransmission and complete reversal of aberrant neuronal differentiation in vitro and in vivo. Our data support the notion that neurodevelopmental changes contribute to the prodromal phase of HD and that early, presymptomatic intervention using HDACi may represent a promising novel treatment approach for HD.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Enfermedad de Huntington/fisiopatología , Ácidos Hidroxámicos/farmacología , Indoles/farmacología , Neuronas/efectos de los fármacos , Animales , Animales Modificados Genéticamente , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Modelos Animales de Enfermedad , Femenino , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Proteína Huntingtina/genética , Enfermedad de Huntington/genética , Ventrículos Laterales/patología , Masculino , Ratones Transgénicos , Mutación , Neuronas/metabolismo , Neuronas/fisiología , Panobinostat , Ratas
17.
Oncotarget ; 8(15): 24815-24827, 2017 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-28445977

RESUMEN

Serglycin is an intracellular proteoglycan with a unique ability to adopt highly divergent structures by glycosylation with variable types of glycosaminoglycans (GAGs) when expressed by different cell types. Serglycin is overexpressed in aggressive cancers suggesting its protumorigenic role. In this study, we explored the expression of serglycin in human glioma and its correlation with survival and immune cell infiltration. We demonstrate that serglycin is expressed in glioma and that increased expression predicts poor survival of patients. Analysis of serglycin expression in a large cohort of low- and high-grade human glioma samples reveals that its expression is grade dependent and is positively correlated with mast cell (MC) infiltration. Moreover, serglycin expression in patient-derived glioma cells is significantly increased upon MC co-culture. This is also accompanied by increased expression of CXCL12, CXCL10, as well as markers of cancer progression, including CD44, ZEB1 and vimentin.In conclusion, these findings indicate the importance of infiltrating MCs in glioma by modulating signaling cascades involving serglycin, CD44 and ZEB1. The present investigation reveals serglycin as a potential prognostic marker for glioma and demonstrates an association with the extent of MC recruitment and glioma progression, uncovering potential future therapeutic opportunities for patients.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Glioblastoma/patología , Mastocitos/metabolismo , Mastocitos/patología , Proteoglicanos/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Línea Celular Tumoral , Técnicas de Cocultivo , Progresión de la Enfermedad , Humanos
18.
Biotechniques ; 59(3): 137-48, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26345506

RESUMEN

Diverse cell types have unique transcriptional signatures that are best interrogated at single-cell resolution. Here we describe a novel RNA amplification approach that allows for high fidelity gene profiling of individual cells. This technique significantly diminishes the problem of 3' bias, enabling detection of all regions of transcripts, including the recognition of mRNA with short or completely absent poly(A) tails, identification of noncoding RNAs, and discovery of the full array of splice isoforms from any given gene product. We assess this technique using statistical and bioinformatics analyses of microarray data to establish the limitations of the method. To demonstrate applicability, we profiled individual cells isolated from the mouse subventricular zone (SVZ)-a well-characterized, discrete yet highly heterogeneous neural structure involved in persistent neurogenesis. Importantly, this method revealed multiple splice variants of key germinal zone gene products within individual cells, as well as an unexpected coexpression of several mRNAs considered markers of distinct and separate SVZ cell types. These findings were independently confirmed using RNA-fluorescence in situ hybridization (RNA-FISH), contributing to the utility of this new technology that offers genomic and transcriptomic analysis of small numbers of dynamic and clinically relevant cells.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Técnicas de Amplificación de Ácido Nucleico/métodos , ARN/genética , Análisis de la Célula Individual/métodos , Antígeno AC133 , Animales , Antígenos CD/genética , Línea Celular Tumoral , Proteínas de Unión al ADN , Receptores ErbB/genética , Proteínas del Ojo/genética , Proteína Ácida Fibrilar de la Glía/genética , Glicoproteínas/genética , Proteínas Fluorescentes Verdes/genética , Proteínas de Homeodominio/genética , Humanos , Proteína 1 Inhibidora de la Diferenciación/genética , Ventrículos Laterales/citología , Proteínas de la Membrana/genética , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Factor de Transcripción PAX6 , Factores de Transcripción Paired Box/genética , Péptidos/genética , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Proteínas Represoras/genética
19.
Brain Pathol ; 25(6): 724-32, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25521330

RESUMEN

Diffuse spread through brain parenchyma and the presence of hypoxic foci rimmed by neoplastic cells are two cardinal features of glioblastoma, and low oxygen is thought to drive movement of malignant gliomas in the core of the lesions. Transcription factors associated with epithelial-to-mesenchymal transition (EMT) have been linked to this invasion, and we found that hypoxia increased in vitro invasion up to fourfold in glioblastoma neurosphere lines and induced the expression of ZEB1. Immunohistochemical assessment of 295 surgical specimens consisting of various types of pediatric and adult brain cancers showed that ZEB1 expression was significantly higher in infiltrative lesions than less invasive tumors such as pilocytic astrocytoma and ependymoma. ZEB1 protein was also present in human fetal periventricular stem and progenitor cells and ZEB1 inhibition impaired migration of in vitro propagated human neural stem cells. The induction of ZEB1 protein in hypoxic glioblastoma neurospheres could be partially blocked by the HIF1alpha inhibitor digoxin. Targeting ZEB1 blocked hypoxia-augmented invasion of glioblastoma cells in addition to slowing them in normoxia. These data support the role for ZEB1 in invasive and high-grade brain tumors and suggest its key role in promoting invasion in the hypoxic tumor core as well as in the periphery.


Asunto(s)
Neoplasias Encefálicas/fisiopatología , Hipoxia de la Célula/fisiología , Movimiento Celular/fisiología , Glioma/fisiopatología , Proteínas de Homeodominio/metabolismo , Células-Madre Neurales/fisiología , Factores de Transcripción/metabolismo , Neoplasias Encefálicas/patología , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Técnicas de Silenciamiento del Gen , Glioma/patología , Proteínas de Homeodominio/genética , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Inmunohistoquímica , Clasificación del Tumor , ARN Mensajero/metabolismo , Análisis de Matrices Tisulares , Factores de Transcripción/genética , Homeobox 1 de Unión a la E-Box con Dedos de Zinc
20.
PLoS One ; 9(3): e88770, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24594681

RESUMEN

OBJECTIVE: Deep brain stimulation (DBS) has been used for more than a decade to treat Parkinson's disease (PD); however, its mechanism of action remains unknown. Given the close proximity of the electrode trajectory to areas of the brain known as the "germinal niches," we sought to explore the possibility that DBS influences neural stem cell proliferation locally, as well as more distantly. METHODS: We studied the brains of a total of 12 idiopathic Parkinson's disease patients that were treated with DBS (the electrode placement occurred 0.5-6 years before death), and who subsequently died of unrelated illnesses. These were compared to the brains of 10 control individuals without CNS disease, and those of 5 PD patients with no DBS. RESULTS: Immunohistochemical analyses of the subventricular zone (SVZ) of the lateral ventricles, the third ventricle lining, and the tissue surrounding the DBS lead revealed significantly greater numbers of proliferating cells expressing markers of the cell cycle, plasticity, and neural precursor cells in PD-DBS tissue compared with both normal brain tissue and tissue from PD patients not treated with DBS. The level of cell proliferation in the SVZ in PD-DBS brains was 2-6 fold greater than that in normal and untreated PD brains. CONCLUSIONS: Our data suggest that DBS is capable of increasing cellular plasticity in the brain, and we hypothesize that it may have more widespread effects beyond the electrode location. It is unclear whether these effects of DBS have any symptomatic or other beneficial influences on PD.


Asunto(s)
Proliferación Celular , Estimulación Encefálica Profunda , Enfermedad de Parkinson/terapia , Humanos , Inmunohistoquímica , Enfermedad de Parkinson/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA