Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cancers (Basel) ; 15(20)2023 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-37894405

RESUMEN

Glioblastomas (GBs) are incurable brain tumors. The persistence of aggressive stem-like tumor cells after cytotoxic treatments compromises therapeutic efficacy, leading to GBM recurrence. Forcing the GBM cells to irreversibly abandon their aggressive stem-like phenotype may offer an alternative to conventional cytotoxic treatments. Here, we show that the RNA binding protein CELF2 is strongly expressed in mitotic and OLIG2-positive GBM cells, while it is downregulated in differentiated and non-mitotic cells by miR-199a-3p, exemplifying GBM intra-tumor heterogeneity. Using patient-derived cells and human GBM samples, we demonstrate that CELF2 plays a key role in maintaining the proliferative/OLIG2 cell phenotype with clonal and tumorigenic properties. Indeed, we show that CELF2 deficiency in patient-derived GSCs drastically reduced tumor growth in the brains of nude mice. We further show that CELF2 promotes TRIM28 and G9a expression, which drive a H3K9me3 epigenetic profile responsible for the silencing of the SOX3 gene. Thus, CELF2, which is positively correlated with OLIG2 and Ki67 expression in human GBM samples, is inversely correlated with SOX3 and miR-199a-3p. Accordingly, the invalidation of SOX3 in CELF2-deficient patient-derived cells rescued proliferation and OLIG2 expression. Finally, patients expressing SOX3 above the median level of expression tend to have a longer life expectancy. CELF2 is therefore a crucial target for the malignant potential of GBM and warrants attention when developing novel anticancer strategies.

2.
Cancer Res Commun ; 3(6): 1041-1056, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37377608

RESUMEN

Glioblastomas (GBM) are heterogeneous tumors with high metabolic plasticity. Their poor prognosis is linked to the presence of glioblastoma stem cells (GSC), which support resistance to therapy, notably to temozolomide (TMZ). Mesenchymal stem cells (MSC) recruitment to GBM contributes to GSC chemoresistance, by mechanisms still poorly understood. Here, we provide evidence that MSCs transfer mitochondria to GSCs through tunneling nanotubes, which enhances GSCs resistance to TMZ. More precisely, our metabolomics analyses reveal that MSC mitochondria induce GSCs metabolic reprograming, with a nutrient shift from glucose to glutamine, a rewiring of the tricarboxylic acid cycle from glutaminolysis to reductive carboxylation and increase in orotate turnover as well as in pyrimidine and purine synthesis. Metabolomics analysis of GBM patient tissues at relapse after TMZ treatment documents increased concentrations of AMP, CMP, GMP, and UMP nucleotides and thus corroborate our in vitro analyses. Finally, we provide a mechanism whereby mitochondrial transfer from MSCs to GSCs contributes to GBM resistance to TMZ therapy, by demonstrating that inhibition of orotate production by Brequinar (BRQ) restores TMZ sensitivity in GSCs with acquired mitochondria. Altogether, these results identify a mechanism for GBM resistance to TMZ and reveal a metabolic dependency of chemoresistant GBM following the acquisition of exogenous mitochondria, which opens therapeutic perspectives based on synthetic lethality between TMZ and BRQ. Significance: Mitochondria acquired from MSCs enhance the chemoresistance of GBMs. The discovery that they also generate metabolic vulnerability in GSCs paves the way for novel therapeutic approaches.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Células Madre Mesenquimatosas , Humanos , Glioblastoma/tratamiento farmacológico , Resistencia a Antineoplásicos , Neoplasias Encefálicas/tratamiento farmacológico , Línea Celular Tumoral , Temozolomida/farmacología , Mitocondrias , Células Madre Neoplásicas
4.
Trends Cancer ; 9(1): 9-27, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36400694

RESUMEN

Glioblastoma (GBM) is the most deadly type of malignant brain tumor, despite extensive molecular analyses of GBM cells. In recent years, the tumor microenvironment (TME) has been recognized as an important player and therapeutic target in GBM. However, there is a need for a full and integrated understanding of the different cellular and molecular components involved in the GBM TME and their interactions for the development of more efficient therapies. In this review, we provide a comprehensive report of the GBM TME, which assembles the contributions of physicians and translational researchers working on brain tumor pathology and therapy in France. We propose a holistic view of the subject by delineating the specific features of the GBM TME at the cellular, molecular, and therapeutic levels.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Humanos , Glioblastoma/terapia , Glioblastoma/tratamiento farmacológico , Microambiente Tumoral/genética , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/terapia , Neoplasias Encefálicas/patología
5.
Cell Death Dis ; 13(10): 913, 2022 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-36310164

RESUMEN

Cell motility is critical for tumor malignancy. Metabolism being an obligatory step in shaping cell behavior, we looked for metabolic weaknesses shared by motile cells across the diverse genetic contexts of patients' glioblastoma. Computational analyses of single-cell transcriptomes from thirty patients' tumors isolated cells with high motile potential and highlighted their metabolic specificities. These cells were characterized by enhanced mitochondrial load and oxidative stress coupled with mobilization of the cysteine metabolism enzyme 3-Mercaptopyruvate sulfurtransferase (MPST). Functional assays with patients' tumor-derived cells and -tissue organoids, and genetic and pharmacological manipulations confirmed that the cells depend on enhanced ROS production and MPST activity for their motility. MPST action involved protection of protein cysteine residues from damaging hyperoxidation. Its knockdown translated in reduced tumor burden, and a robust increase in mice survival. Starting from cell-by-cell analyses of the patients' tumors, our work unravels metabolic dependencies of cell malignancy maintained across heterogeneous genomic landscapes.


Asunto(s)
Glioblastoma , Ratones , Animales , Glioblastoma/genética , Cisteína/metabolismo , Sulfurtransferasas/genética , Sulfurtransferasas/metabolismo , Estrés Oxidativo , Movimiento Celular/genética
6.
Proc Natl Acad Sci U S A ; 118(30)2021 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-34301885

RESUMEN

Germ cells form the basis for sexual reproduction by producing gametes. In ovaries, primordial germ cells exit the cell cycle and the pluripotency-associated state, differentiate into oogonia, and initiate meiosis. Despite the importance of germ cell differentiation for sexual reproduction, signaling pathways regulating their fate remain largely unknown. Here, we show in mouse embryonic ovaries that germ cell-intrinsic ß-catenin activity maintains pluripotency and that its repression is essential to allow differentiation and meiosis entry in a timely manner. Accordingly, in ß-catenin loss-of-function and gain-of-function mouse models, the germ cells precociously enter meiosis or remain in the pluripotent state, respectively. We further show that interaction of ß-catenin and the pluripotent-associated factor POU5F1 in the nucleus is associated with germ cell pluripotency. The exit of this complex from the nucleus correlates with germ cell differentiation, a process promoted by the up-regulation of Znrf3, a negative regulator of WNT/ß-catenin signaling. Together, these data identify the molecular basis of the transition from primordial germ cells to oogonia and demonstrate that ß-catenin is a central gatekeeper in ovarian differentiation and gametogenesis.


Asunto(s)
Diferenciación Celular , Células Germinativas/citología , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Células Madre Pluripotentes/citología , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Animales , Femenino , Células Germinativas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Factor 3 de Transcripción de Unión a Octámeros/genética , Células Madre Pluripotentes/metabolismo , Proteínas Wnt/genética , beta Catenina/genética
7.
Cancer Res ; 80(16): 3236-3250, 2020 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-32366479

RESUMEN

There is great interest in understanding how the cancer stem cell population may be maintained in solid tumors. Here, we show that tumor cells exhibiting stem-like properties and expression of pluripotency markers NANOG and OCT4 can arise from original differentiated tumor cells freshly isolated from human glioblastomas (GBM) and that have never known any serum culture conditions. Induction of EGR1 by EGFR/ERK signaling promoted cell conversion from a less aggressive, more differentiated cellular state to a self-renewing and strongly tumorigenic state, expressing NANOG and OCT4. Expression of these pluripotency markers occurred before the cells re-entered the cell cycle, demonstrating their capacity to change and dedifferentiate without any cell divisions. In differentiated GBM cells, ERK-mediated repression of miR-199a-3p induced EGR1 protein expression and triggered dedifferentiation. Overall, this signaling pathway constitutes an ERK-mediated "toggle switch" that promotes pluripotency marker expression and stem-like features in GBM cells. SIGNIFICANCE: This study defines an ERK-mediated molecular mechanism of dedifferentiation of GBM cells into a stem-like state, expressing markers of pluripotency.See related commentary by Koncar and Agnihotri, p. 3195.


Asunto(s)
Glioblastoma , MicroARNs , Desdiferenciación Celular , Diferenciación Celular , Proteína 1 de la Respuesta de Crecimiento Precoz , Glioblastoma/genética , Humanos , MicroARNs/genética , Proteína Homeótica Nanog/genética , Células Madre Neoplásicas
8.
Obes Surg ; 29(5): 1576-1582, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30712172

RESUMEN

BACKGROUND: The impact of bariatric surgery (BS) on the sexual functioning of patients is poorly studied. Our aim was to analyze the sexual function, depressive symptoms, and self-esteem of morbidly obese women (MOW) undergoing BS. PATIENTS AND METHODS: Quality of sexual life was prospectively evaluated in 43 consecutive MOW (18-50 years) who underwent BS. Female sexual function index (FSFI), Beck depression inventory (BDI), and Rosenberg self-esteem scale (RSES) questionnaires were administered to evaluate sexual satisfaction, depressive symptoms, and self-esteem, respectively. A control group of 36 healthy, non-obese, female patients (HW) was recruited for comparison. Results of questionnaires were compared between three periods (before BS and at 3- and 6-month follow-up) and between MOW and HW. RESULTS: Before BS, the FSFI score was significantly lower in MOW compared to HW (17 ± 12 vs 27 ± 8, p = 0.0001) while at 3- and 6-month post-BS, a significant amelioration (p = 0.01) occurred. In particular, after BS, all components of the FSFI score (sexual desire, excitement, lubrification, orgasm, satisfaction, and pain) were ameliorated. The pre-BS BDI score was higher in MOW than in HW (8 ± 6 vs 5 ± 5, p = 0.004) while at postoperative months 3 and 6, a significant amelioration was found (p = 0.025 and 0.005, respectively). Before BS, no significant differences occurred in the RSES score between MOW and HW (30 ± 7 vs 32 ± 6, p = 0.014), whereas the MOW RSES scores at 6-month post-BS were improved when compared with the HW RSES scores. CONCLUSIONS: BS results in a significant improvement in the quality of sexual life, depressive symptoms, and self-esteem in MOW.


Asunto(s)
Cirugía Bariátrica , Obesidad Mórbida/cirugía , Adulto , Depresión/psicología , Femenino , Humanos , Estudios Longitudinales , Persona de Mediana Edad , Obesidad Mórbida/fisiopatología , Obesidad Mórbida/psicología , Estudios Prospectivos , Escalas de Valoración Psiquiátrica , Calidad de Vida , Autoimagen , Conducta Sexual/fisiología , Conducta Sexual/psicología , Encuestas y Cuestionarios , Adulto Joven
9.
Obes Surg ; 29(5): 1462-1469, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30666544

RESUMEN

BACKGROUND: Recent evidence has indicated an increased risk of Barrett's esophagus (BE) in the long term after sleeve gastrectomy (SG). AIM: The aim of the study is to investigate the spectrum of gastroesophageal reflux disease (GERD) symptoms as well as the prevalence of BE, at minimum 5 years after SG in patients who underwent SG in different bariatric centers of two countries: France and Italy. PATIENTS AND METHODS: Five high volume outpatient centers dedicated to bariatric surgery that routinely perform upper GI endoscopy before any bariatric procedures were invited to participate in the study. From January 2017 to June 2018, each center during scheduled postoperative evaluation after surgery asked a minimum 10 consecutive patients, which had performed SG at least 5 years before and with no evidence of BE preoperatively, to undergo another upper GI endoscopy. RESULTS: Ninety (66 F) consecutive patients were enrolled. The mean follow-up was 78 ± 15 months, and the mean total body weight loss was 25 ± 12%. The prevalence of BE was 18.8% with no significant difference among centers. Weight loss failure was significantly associated with BE (p < 0.01). The prevalence of GERD symptoms, erosive esophagitis, and the usage of PPIs increased from 22%, 10%, and 22% before the SG to 76%, 41%, and 52% at the time of follow-up, respectively (p < 0.05). CONCLUSIONS: This multicenter study show a high rate of BE at least 5 years after SG. Weight loss failure was significantly associated with BE. We suggest to provide systematic endoscopy in these patients to rule out this condition.


Asunto(s)
Cirugía Bariátrica/efectos adversos , Esófago de Barrett/etiología , Gastrectomía/efectos adversos , Adulto , Cirugía Bariátrica/métodos , Esófago de Barrett/epidemiología , Endoscopía del Sistema Digestivo/métodos , Esofagitis/epidemiología , Esofagitis/etiología , Femenino , Estudios de Seguimiento , Francia/epidemiología , Gastrectomía/métodos , Reflujo Gastroesofágico/epidemiología , Reflujo Gastroesofágico/etiología , Humanos , Italia/epidemiología , Masculino , Persona de Mediana Edad , Obesidad Mórbida/epidemiología , Obesidad Mórbida/cirugía , Úlcera Péptica/epidemiología , Úlcera Péptica/etiología , Prevalencia , Inhibidores de la Bomba de Protones/uso terapéutico , Pérdida de Peso
10.
Cancer Lett ; 446: 112-122, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30660649

RESUMEN

Glioblastoma (GBM) is the brain tumor with the worst prognosis composed of a cell subpopulation called Glioblastoma Stem-like Cells (GSCs) responsible for tumor recurrence mediated by cell invasion. GSCs persist in a hypoxic microenvironment which promotes extracellular adenosine production and activation of the A3 Adenosine Receptor (A3AR), therefore, the aim of this study was to determine the role of extracellular adenosine and A3AR on GSCs invasion under hypoxia. GSCs were obtained from a U87MG cell line and primary cultures of GBM patients, and then incubated under normoxia or hypoxia. Gene expression was evaluated by RNAseq, RT-qPCR, and western blot. Cell migration was measured by spreading and transwell boyden chamber assays; cell invasion was evaluated by Matrigel-coated transwell, ex vivo brain slice, and in vivo xenograft assays. The contribution of A3AR on cell migration/invasion was evaluated using the A3AR antagonist, MRS1220. Extracellular adenosine production was higher under hypoxia than normoxia, mainly by the catalytic action of the prostatic acid phosphatase (PAP), promoting cell migration/invasion in a HIF-2-dependent process. A3AR blockade decreased cell migration/invasion and the expression of Epithelial-Mesenchymal Transition markers. In conclusion, high levels of extracellular adenosine production enhance cell migration/invasion of GSCs, through HIF-2/PAP-dependent activation of A3AR under hypoxia.


Asunto(s)
Adenosina/metabolismo , Neoplasias Encefálicas/metabolismo , Movimiento Celular , Glioblastoma/metabolismo , Células Madre Neoplásicas/metabolismo , Receptor de Adenosina A3/metabolismo , Fosfatasa Ácida/genética , Fosfatasa Ácida/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Glioblastoma/genética , Glioblastoma/patología , Humanos , Ratones Endogámicos NOD , Ratones SCID , Invasividad Neoplásica , Células Madre Neoplásicas/patología , Receptor de Adenosina A3/genética , Transducción de Señal , Células Tumorales Cultivadas , Hipoxia Tumoral , Microambiente Tumoral
11.
Obes Surg ; 29(2): 609-616, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30448982

RESUMEN

BACKGROUND: Obesity is a well-known risk factor for female pelvic floor disorders (PFD). This study assessed the effects of bariatric surgery (BS) on pelvic organ prolapse symptoms (POPs) and urinary (UI) and anal incontinence (AI) in morbidly obese women undergoing either sleeve gastrectomy (SG) or Roux-en-Y gastric bypass (RYGB). METHODS: Morbidly obese women undergoing BS from June 2016 to May 2017 were prospectively included. POPs, UI, and AI were compared at baseline and at 1 year after surgery using validated questionnaires. RESULTS: Seventy-two consecutive women were enrolled, 54 (75%) (30 (56%) RYBP and 24 (44%) SG) completed the study at 1 year and were considered for the final analysis. The mean age and mean preoperative BMI were 43 ± 11.8 years (range, 20-65) and 41 ± 5.4 kg/m2 (range, 35-56), respectively. At baseline, 30 (56%), 32 (59%), and 27 (50%) patients, respectively, had AI (flatus only 72%), UI, and POPs. The mean TBWL% at 1 year was 33%. In the whole study population, weight loss was associated with a significant improvement in UI (p < 0.001) but there was no significant difference in terms of AI and POPs. In the subgroups analysis, AI increased significantly 1 year after the RYGB (p = 0.02) due to an increase in flatus incontinence (p = 0.04). No significant difference in AI was found 1 year after the SG. CONCLUSION: BS is associated with a significant improvement in UI but not in POPs. RYBP seems to increase AI, mainly flatus incontinence, compared to SG.


Asunto(s)
Incontinencia Fecal/cirugía , Gastrectomía , Derivación Gástrica , Prolapso de Órgano Pélvico/cirugía , Incontinencia Urinaria/cirugía , Adulto , Anciano , Incontinencia Fecal/etiología , Femenino , Flatulencia , Humanos , Laparoscopía , Persona de Mediana Edad , Obesidad Mórbida/complicaciones , Obesidad Mórbida/cirugía , Prolapso de Órgano Pélvico/etiología , Proyectos Piloto , Estudios Prospectivos , Incontinencia Urinaria/etiología , Adulto Joven
12.
Cell Death Dis ; 8(3): e2713, 2017 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-28358371

RESUMEN

Glioblastomas are incurable primary brain tumors that affect patients of all ages. The aggressiveness of this cancer has been attributed in part to the persistence of treatment-resistant glioblastoma stem-like cells. We have previously discovered the tumor-suppressor properties of the microRNA cluster miR-302-367, representing a potential treatment for glioblastoma. Here, we attempted to develop a cell-based therapy by taking advantage of the capability of glioma cells to secrete exosomes that enclose small RNA molecules. We engineered primary glioma cells to stably express the miR-302-367. Remarkably, these cells altered, in a paracrine-dependent manner, the expression of stemness markers, the proliferation and the tumorigenicity of neighboring glioblastoma cells. Further characterization of the secretome derived from miR-302-367 expressing cells showed that a large amount of miR-302-367 was enclosed in exosomes, which were internalized by the neighboring glioblastoma cells. This miR-302-367 cell-to-cell transfer resulted in the inhibition of its targets such as CXCR4/SDF1, SHH, cyclin D, cyclin A and E2F1. Orthotopic xenograft of miR-302-367-expressing cells together with glioblastoma stem-like cells efficiently altered the tumor development in mice brain.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Glioblastoma/metabolismo , MicroARNs/biosíntesis , Familia de Multigenes , Proteínas de Neoplasias/biosíntesis , ARN Neoplásico/biosíntesis , Animales , Tratamiento Basado en Trasplante de Células y Tejidos , Glioblastoma/genética , Glioblastoma/terapia , Humanos , Ratones , MicroARNs/genética , Proteínas de Neoplasias/genética , ARN Neoplásico/genética
13.
Acta Neuropathol ; 133(4): 645-660, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28032215

RESUMEN

Cell populations with differing proliferative, stem-like and tumorigenic states co-exist in most tumors and especially malignant gliomas. Whether metabolic variations can drive this heterogeneity by controlling dynamic changes in cell states is unknown. Metabolite profiling of human adult glioblastoma stem-like cells upon loss of their tumorigenicity revealed a switch in the catabolism of the GABA neurotransmitter toward enhanced production and secretion of its by-product GHB (4-hydroxybutyrate). This switch was driven by succinic semialdehyde dehydrogenase (SSADH) downregulation. Enhancing GHB levels via SSADH downregulation or GHB supplementation triggered cell conversion into a less aggressive phenotypic state. GHB affected adult glioblastoma cells with varying molecular profiles, along with cells from pediatric pontine gliomas. In all cell types, GHB acted by inhibiting α-ketoglutarate-dependent Ten-eleven Translocations (TET) activity, resulting in decreased levels of the 5-hydroxymethylcytosine epigenetic mark. In patients, low SSADH expression was correlated with high GHB/α-ketoglutarate ratios, and distinguished weakly proliferative/differentiated glioblastoma territories from proliferative/non-differentiated territories. Our findings support an active participation of metabolic variations in the genesis of tumor heterogeneity.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Carcinogénesis/metabolismo , Glioma/metabolismo , Hidroxibutiratos/metabolismo , Células Madre Neoplásicas/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Anciano , Animales , Encéfalo/metabolismo , Encéfalo/patología , Encéfalo/cirugía , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/cirugía , Carcinogénesis/patología , Muerte Celular/fisiología , Proliferación Celular/fisiología , Niño , Preescolar , Femenino , Glioma/patología , Glioma/cirugía , Humanos , Masculino , Ratones Desnudos , Persona de Mediana Edad , Trasplante de Neoplasias , Células Madre Neoplásicas/patología , Succionato-Semialdehído Deshidrogenasa/metabolismo
14.
J Biol Chem ; 291(20): 10684-99, 2016 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-27002148

RESUMEN

Glioblastomas are the most common primary brain tumors, highly vascularized, infiltrating, and resistant to current therapies. This cancer leads to a fatal outcome in less than 18 months. The aggressive behavior of glioblastomas, including resistance to current treatments and tumor recurrence, has been attributed to glioma stemlike/progenitor cells. The transcription factor EGR1 (early growth response 1), a member of a zinc finger transcription factor family, has been described as tumor suppressor in gliomas when ectopically overexpressed. Although EGR1 expression in human glioblastomas has been associated with patient survival, its precise location in tumor territories as well as its contribution to glioblastoma progression remain elusive. In the present study, we show that EGR1-expressing cells are more frequent in high grade gliomas where the nuclear expression of EGR1 is restricted to proliferating/progenitor cells. We show in primary cultures of glioma stemlike cells that EGR1 contributes to stemness marker expression and proliferation by orchestrating a PDGFA-dependent growth-stimulatory loop. In addition, we demonstrate that EGR1 acts as a positive regulator of several important genes, including SHH, GLI1, GLI2, and PDGFA, previously linked to the maintenance and proliferation of glioma stemlike cells.


Asunto(s)
Comunicación Autocrina , Neoplasias Encefálicas/metabolismo , Proliferación Celular , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Regulación Neoplásica de la Expresión Génica , Glioblastoma/metabolismo , Proteínas de Neoplasias/metabolismo , Células Madre Neoplásicas/metabolismo , Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Neoplasias Encefálicas/patología , Femenino , Glioblastoma/patología , Humanos , Masculino , Células Madre Neoplásicas/patología , Células Tumorales Cultivadas
15.
Neuro Oncol ; 15(10): 1278-88, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23935154

RESUMEN

BACKGROUND: The differential diagnosis between infiltrative glioma (IG) and benign or curable glial lesions, such as gliosis, pilocytic astrocytoma, dysembryoplastic neuroepithelial tumor, ganglioglioma, or demyelinating disease, may be challenging for the pathologist because specific markers are lacking. Recently, we described a strong EGFR immunolabelling pattern in cells with a high nuclear to cytoplasmic ratio that enables the discrimination of low-grade IG from gliosis. The aim of this study was to extend our observation to high-grade glioma to assess whether EGFR expression pattern is of value in the discrimination of all IG from noninfiltrative glial lesions (NIG), including gliosis, benign tumors, and demyelinating disease. METHODS: One hundred one IG and 58 NIG were compared for immunohistochemical expression of EGFR with use of an antibody that recognizes an epitope in the extracellular domain of both EGFRwt and EGFRvIII. Highly EGFR-positive cells with a high nuclear to cytoplasmic ratio were isolated and further characterized. RESULTS: Cells with intense EGFR staining and a high nuclear to cytoplasmic ratio were significantly associated with the diagnosis of IG (P < .0001). The sensitivity and specificity of this staining pattern for the diagnosis of IG were 95% and 100%, respectively. EGFR expression was independent of IDH1 mutations and EGFR amplification. Finally, we showed that these particular cells displayed the phenotype and properties of glial progenitors and coexpressed CXCR4, a marker of invasiveness. CONCLUSIONS: We demonstrate that cells with intense EGFR staining and a high nuclear to cytoplasmic ratio are specific criteria for the diagnosis of IG, irrespective of grade, histological subtype, and progression pathway, and their identification represents a tool to discriminate IG from benign or curable glial lesions.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias Encefálicas/patología , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Receptores ErbB/metabolismo , Glioma/patología , Adulto , Biomarcadores de Tumor/genética , Western Blotting , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Niño , Receptores ErbB/genética , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Glioma/genética , Glioma/metabolismo , Humanos , Técnicas para Inmunoenzimas , Hibridación Fluorescente in Situ , Masculino , Estudios Retrospectivos , Células Tumorales Cultivadas
16.
Biomaterials ; 34(33): 8279-90, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23899445

RESUMEN

Glioblastoma is an aggressive brain tumor characterized by its high propensity for local invasion, formation of secondary foci within the brain, as well as areas of necrosis. This study aims to (i) provide a technical approach to reproduce features of the disease in vitro and (ii) characterize the tumor/host brain tissue interaction at the molecular level. Human engineered neural tissue (ENT) obtained from pluripotent stem cells was generated and co-cultured with human glioblastoma-initiating cells. Within two weeks, glioblastoma cells invaded the nervous tissue. This invasion displayed features of the disease in vivo: a primary tumor mass, diffuse migration of invading single cells into the nervous tissue, secondary foci, as well as peritumoral cell death. Through comparative molecular analyses, this model allowed the identification of more than 100 genes that are specifically induced and up-regulated by the nervous tissue/tumor interaction. Notably the type I interferon response, extracellular matrix-related genes were most highly represented and showed a significant correlation with patient survival. In conclusion, glioblastoma development within a nervous tissue can be engineered in vitro, providing a relevant model to study the disease and allows the identification of clinically-relevant genes induced by the tumor/host tissue interaction.


Asunto(s)
Neoplasias Encefálicas/patología , Glioblastoma/patología , Animales , Línea Celular , Células Cultivadas , Femenino , Humanos , Inmunohistoquímica , Ratones , Reacción en Cadena de la Polimerasa , Embarazo , Células Madre/citología , Ingeniería de Tejidos
17.
Stem Cells ; 31(7): 1252-65, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23533157

RESUMEN

Stem cell-like properties of glioma initiating cells (GiCs) fuel glioblastoma (GBM) development by providing the different cell types that comprise the tumor. It is therefore likely that the molecular circuitries that regulate their decision to self-renew or commit to a more differentiated state may offer targets for future innovative therapies. In previous micro-RNA profiling studies to search for regulators of stem cell plasticity, we identified miR-18a* as a potential candidate and its expression correlated with the stemness state. Here, using human GiCs we found that miR-18a* expression promotes clonal proliferation in vitro and tumorigenicity in vivo. Mechanistically, ERK-dependent induction of miR-18a* directly represses expression of DLL3, an autocrine inhibitor of NOTCH, thus enhancing the level of activated NOTCH-1. Activated NOTCH-1 in turn is required for sustained ERK activation. This feed-forward loop, driven by miR-18a*, is required to turn on the SHH-GLI-NANOG network, essential for GiC self-renewal. Hence, by tightly regulating expression of DLL3, miR-18a* constitutes an important signaling mediator for fine tuning the level of GiC self-renewal.


Asunto(s)
Glioma/genética , MicroARNs/genética , Receptor Notch1/metabolismo , Anciano , Animales , Diferenciación Celular/fisiología , Procesos de Crecimiento Celular/fisiología , Regulación hacia Abajo , Glioma/metabolismo , Glioma/patología , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Sistema de Señalización de MAP Quinasas/genética , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos NOD , MicroARNs/biosíntesis , MicroARNs/metabolismo , Persona de Mediana Edad , Receptor Notch1/genética , Transfección
18.
Stem Cells ; 27(3): 509-20, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19074418

RESUMEN

Researches on neural differentiation using embryonic stem cells (ESC) require analysis of neurogenesis in conditions mimicking physiological cellular interactions as closely as possible. In this study, we report an air-liquid interface-based culture of human ESC. This culture system allows three-dimensional cell expansion and neural differentiation in the absence of added growth factors. Over a 3-month period, a macroscopically visible, compact tissue developed. Histological coloration revealed a dense neural-like neural tissue including immature tubular structures. Electron microscopy, immunochemistry, and electrophysiological recordings demonstrated a dense network of neurons, astrocytes, and oligodendrocytes able to propagate signals. Within this tissue, tubular structures were niches of cells resembling germinal layers of human fetal brain. Indeed, the tissue contained abundant proliferating cells expressing markers of neural progenitors. Finally, the capacity to generate neural tissues on air-liquid interface differed for different ESC lines, confirming variations of their neurogenic potential. In conclusion, this study demonstrates in vitro engineering of a human neural-like tissue with an organization that bears resemblance to early developing brain. As opposed to previously described methods, this differentiation (a) allows three-dimensional organization, (b) yields dense interconnected neural tissue with structurally and functionally distinct areas, and (c) is spontaneously guided by endogenous developmental cues.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Embrionarias/citología , Sistema Nervioso/citología , Neuronas/citología , Técnicas de Cultivo de Tejidos/métodos , Línea Celular , Electrofisiología , Células Madre Embrionarias/fisiología , Células Madre Embrionarias/ultraestructura , Humanos , Inmunohistoquímica , Microscopía Electrónica de Transmisión , Sistema Nervioso/ultraestructura , Neuronas/ultraestructura , Reacción en Cadena de la Polimerasa
19.
Cell Cycle ; 7(13): 2038-46, 2008 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-18604165

RESUMEN

Tissue homeostasis requires precise control of cell proliferation and arrest in response to environmental cues. In situation such as wound healing, injured cells are stimulated to divide, but as soon as confluence is reached proliferation must be blocked. Such reversible cell cycle exit occurs in G(1), requires pRb family members, and is driven by p27(Kip1)-dependent Cdk inactivation. This implies that, while dividing, cells should simultaneously prepare the exit once mitosis is accomplished. For a long time, the decision to cycle or not was presumed to occur in G(1), prior to the restriction point, beyond which the cells were bound to divide even in the absence of mitogens, before finally arresting after mitosis. However, more recent reports suggested that the commitment to cycle in response to serum occurs already in G(2) phase and requires the Ras-dependent induction of cyclin D1, which promotes following G(1)/S transition. To test whether this hypothesis applies to arrest induced by contact inhibition, we used an in vitro wounding model where quiescent human dermal fibroblasts, stimulated to proliferate by mechanical injury, synchronously exit cell cycle after mitosis due to renewed confluence. We show that this exit is preceded by p27-dependent inhibition of cyclin A-Cdk1/2, cyclin D1 downregulation and reduced pre-mitotic pRb pocket protein phosphorylation. Overexpression of cyclin D1 but not p27 depletion reversed this phenotype and compromised confluence-driven cell cycle exit. Thus, a balance between cyclin D1 and p27 may provide sensitive responses to variations in proliferative cues operating throughout the cell cycle.


Asunto(s)
Ciclo Celular/fisiología , Ciclina D1/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Fibroblastos/metabolismo , Proteínas Quinasas Asociadas a Fase-S/metabolismo , Quinasas Ciclina-Dependientes/metabolismo , Regulación hacia Abajo/fisiología , Fase G2 , Humanos , Mitosis , Fosforilación , Cicatrización de Heridas/fisiología
20.
J Biol Chem ; 282(20): 15090-102, 2007 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-17363378

RESUMEN

Covering denuded dermal surfaces after injury requires migration, proliferation, and differentiation of skin keratinocytes. To clarify the major traits controlling these intermingled biological events, we surveyed the genomic modifications occurring during the course of a scratch wound closure of cultured human keratinocytes. Using a DNA microarray approach, we report the identification of 161 new markers of epidermal repair. Expression data, combined with functional analysis performed with specific inhibitors of ERK, p38(MAPK) and phosphatidylinositol 3-kinase (PI3K), demonstrate that kinase pathways exert very selective functions by precisely controlling the expression of specific genes. Inhibition of the ERK pathway totally blocks the wound closure and inactivates many early transcription factors and EGF-type growth factors. p38(MAPK) inhibition only delays "healing," probably in line with the control of genes involved in the propagation of injury-initiated signaling. In contrast, PI3K inhibition accelerates the scratch closure and potentiates the scratch-dependent stimulation of three genes related to epithelial cell transformation, namely HAS3, HBEGF, and ETS1. Our results define in vitro human keratinocyte wound closure as a repair process resulting from a fine balance between positive signals controlled by ERK and p38(MAPK) and negative ones triggered by PI3K. The perturbation of any of these pathways might lead to dysfunction in the healing process, similar to those observed in pathological wounding phenotypes, such as hypertrophic scars or keloids.


Asunto(s)
Dermis/enzimología , Dermis/lesiones , Queratinocitos/enzimología , Sistema de Señalización de MAP Quinasas , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Biomarcadores/metabolismo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Dermis/patología , Inhibidores Enzimáticos/farmacología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Genoma Humano , Humanos , Queloide/enzimología , Queloide/genética , Queloide/patología , Queratinocitos/patología , Sistema de Señalización de MAP Quinasas/genética , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosfatidilinositol 3-Quinasas/genética , Cicatrización de Heridas/efectos de los fármacos , Cicatrización de Heridas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...