Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Neuro Oncol ; 23(11): 1885-1897, 2021 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-33864086

RESUMEN

BACKGROUND: Glioblastoma cells assemble to a syncytial communicating network based on tumor microtubes (TMs) as ultra-long membrane protrusions. The relationship between network architecture and transcriptional profile remains poorly investigated. Drugs that interfere with this syncytial connectivity such as meclofenamate (MFA) may be highly attractive for glioblastoma therapy. METHODS: In a human neocortical slice model using glioblastoma cell populations of different transcriptional signatures, three-dimensional tumor networks were reconstructed, and TM-based intercellular connectivity was mapped on the basis of two-photon imaging data. MFA was used to modulate morphological and functional connectivity; downstream effects of MFA treatment were investigated by RNA sequencing and fluorescence-activated cell sorting (FACS) analysis. RESULTS: TM-based network morphology strongly differed between the transcriptional cellular subtypes of glioblastoma and was dependent on axon guidance molecule expression. MFA revealed both a functional and morphological demolishment of glioblastoma network architectures which was reflected by a reduction of TM-mediated intercellular cytosolic traffic as well as a breakdown of TM length. RNA sequencing confirmed a downregulation of NCAM and axon guidance molecule signaling upon MFA treatment. Loss of glioblastoma communicating networks was accompanied by a failure in the upregulation of genes that are required for DNA repair in response to temozolomide (TMZ) treatment and culminated in profound treatment response to TMZ-mediated toxicity. CONCLUSION: The capacity of TM formation reflects transcriptional cellular heterogeneity. MFA effectively demolishes functional and morphological TM-based syncytial network architectures. These findings might pave the way to a clinical implementation of MFA as a TM-targeted therapeutic approach.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Ácido Meclofenámico/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Línea Celular Tumoral , Proliferación Celular , Glioblastoma/tratamiento farmacológico , Humanos , Técnicas In Vitro
2.
Glia ; 68(2): 316-327, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31509308

RESUMEN

Glioblastoma (GBM) is a deadly disease with a need for deeper understanding and new therapeutic approaches. The microenvironment of glioblastoma has previously been shown to guide glioblastoma progression. In this study, astrocytes were investigated with regard to their effect on glioblastoma proliferation through correlative analyses of clinical samples and experimental in vitro and in vivo studies. Co-culture techniques were used to investigate the GBM growth enhancing potential of astrocytes. Cell sorting and RNA sequencing were used to generate a GBM-associated astrocyte signature and to investigate astrocyte-induced GBM genes. A NOD scid GBM mouse model was used for in vivo studies. A gene signature reflecting GBM-activated astrocytes was associated with poor prognosis in the TCGA GBM dataset. Two genes, periostin and serglycin, induced in GBM cells upon exposure to astrocytes were expressed at higher levels in cases with high "astrocyte signature score". Astrocytes were shown to enhance glioblastoma cell growth in cell lines and in a patient-derived culture, in a manner dependent on cell-cell contact and involving increased cell proliferation. Furthermore, co-injection of astrocytes with glioblastoma cells reduced survival in an orthotopic GBM model in NOD scid mice. In conclusion, this study suggests that astrocytes contribute to glioblastoma growth and implies this crosstalk as a candidate target for novel therapies.


Asunto(s)
Astrocitos/metabolismo , Neoplasias Encefálicas/metabolismo , Movimiento Celular/fisiología , Glioblastoma/metabolismo , Animales , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Proliferación Celular/fisiología , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Glioblastoma/patología , Glioma/metabolismo , Humanos , Ratones Endogámicos NOD
3.
iScience ; 9: 71-83, 2018 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-30384135

RESUMEN

High-grade gliomas (HGGs) are the most aggressive and invasive primary brain tumors. The platelet-derived growth factor (PDGF) signaling pathway drives HGG progression, and enhanced expression of PDGF receptors (PDGFRs) is a well-established aberration in a subset of glioblastomas (GBMs). PDGFRA is expressed in glioma cells, whereas PDGFRB is mostly restricted to the glioma-associated stroma. Here we show that the spatial location of TAMMs correlates with the expansion of a subset of tumor cells that have acquired expression of PDGFRB in both mouse and human low-grade glioma and HCGs. Furthermore, M2-polarized microglia but not bone marrow (BM)-derived macrophages (BMDMs) induced PDGFRB expression in glioma cells and stimulated their migratory capacity. These findings illustrate a heterotypic cross-talk between microglia and glioma cells that may enhance the migratory and invasive capacity of the latter by inducing PDGFRB.

4.
Oncotarget ; 8(7): 12145-12157, 2017 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-28076333

RESUMEN

Surgical resection is a standard component of treatment in the clinical management of patients with glioblastoma multiforme (GBM). However, experimental therapies are rarely investigated in the context of tumor debulking in preclinical models. Here, a surgical debulking GBM xenograft model was developed in nude rats, and was used in combination with CD47 blocking immunotherapy, a novel treatment strategy that triggers phagocytosis of tumor cells by macrophages in diverse cancer types including GBM. Orthotopic patient-derived xenograft tumors expressing CD47 were resected at 4 weeks after implantation and immediately thereafter treated with anti-CD47 or control antibodies injected into the cavity. Debulking prolonged survival (median survival, 68.5 vs 42.5 days, debulking and non-debulking survival times, respectively; n = 6 animals/group; P = 0.0005). Survival was further improved in animals that underwent combination treatment with anti-CD47 mAbs (median survival, 81.5 days vs 69 days, debulking + anti-CD47 vs debulking + control IgG, respectively; P = 0.0007). Immunohistochemistical staining of tumor sections revealed an increase in recruitment of cells positive for CD68, a marker for macrophages/immune cell types, to the surgical site (50% vs 10%, debulking vs non-debulking, respectively). Finally, analysis of tumor protein lysates on antibody microarrays demonstrated an increase in pro-inflammatory cytokines, such as CXCL10, and a decrease in angiogenic proteins in debulking + anti-CD47 vs non-debulking + IgG tumors. The results indicated that surgical resection combined with anti-CD47 blocking immunotherapy promoted an inflammatory response and prolonged survival in animals, and is therefore an attractive strategy for clinical translation.


Asunto(s)
Antígeno CD47/metabolismo , Procedimientos Quirúrgicos de Citorreducción/métodos , Glioblastoma/terapia , Inmunoterapia/métodos , Macrófagos/metabolismo , Fagocitosis , Animales , Anticuerpos Bloqueadores/inmunología , Anticuerpos Bloqueadores/farmacología , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Antígeno CD47/inmunología , Terapia Combinada , Citocinas/metabolismo , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Inmunohistoquímica , Mediadores de Inflamación/metabolismo , Ratas Desnudas , Análisis de Supervivencia , Factores de Tiempo , Ensayos Antitumor por Modelo de Xenoinjerto
5.
BMC Cancer ; 12: 21, 2012 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-22251838

RESUMEN

BACKGROUND: The tumor microenvironment is pivotal in tumor progression. Thus, we aimed to develop a mammary tumor model to elucidate molecular characteristics in the stroma versus the tumor cell compartment by global gene expression. Secondly, since tumor hypoxia influences several aspects of tumor pathophysiology, we hypothesized that hyperoxia might have an inhibitory effect on tumor growth per se. Finally, we aimed to identify differences in gene expression and key molecular mechanisms, both in the native state and following treatment. METHODS: 4T1 dsRed breast cancer cells were injected into eGFP expressing NOD/SCID mice. Group 1 was exposed to 3 intermittent HBO treatments (Day 1, 4 and 7), Group 2 to 7 daily HBO treatments (both 2.5 bar, 100% O2, à 90 min), whereas the controls were exposed to a normal atmosphere. Tumor growth, histology, vascularisation, cell proliferation, cell death and metastasis were assessed. Fluorescence-activated cell sorting was used to separate tumor cells from stromal cells prior to gene expression analysis. RESULTS: The purity of sorted cells was verified by fluorescence microscopy. Gene expression profiling demonstrated that highly expressed genes in the untreated tumor stroma included constituents of the extracellular matrix and matrix metalloproteinases. Tumor growth was significantly inhibited by HBO, and the MAPK pathway was found to be significantly reduced. Immunohistochemistry indicated a significantly reduced microvessel density after intermittent HBO, whereas daily HBO did not show a similar effect. The anti-angiogenic response was reflected in the expression trends of angiogenic factors. CONCLUSIONS: The present in vivo mammary tumor model enabled us to separate tumor and stromal cells, and demonstrated that the two compartments are characterized by distinct gene expressions, both in the native state and following HBO treatments. Furthermore, hyperoxia induced a significant tumor growth-inhibitory effect, with significant down-regulation of the MAPK pathway. An anti-angiogenic effect after intermittent HBO was observed, and reflected in the gene expression profile.


Asunto(s)
Hipoxia de la Célula/fisiología , Perfilación de la Expresión Génica , Neoplasias Mamarias Experimentales/genética , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Matriz Extracelular/genética , Femenino , Proteínas Fluorescentes Verdes/metabolismo , Oxigenoterapia Hiperbárica , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Metaloproteinasas de la Matriz/genética , Ratones , Ratones Endogámicos NOD , Ratones SCID
6.
Acta Neuropathol ; 122(4): 495-510, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21863242

RESUMEN

Glioblastoma (GBM) is a highly aggressive brain tumour, where patients respond poorly to radiotherapy and exhibit dismal survival outcomes. The mechanisms of radioresistance are not completely understood. However, cancer cells with an immature stem-like phenotype are hypothesised to play a role in radioresistance. Since the progenitor marker neuron-glial-2 (NG2) has been shown to regulate several aspects of GBM progression in experimental systems, we hypothesised that its expression would influence the survival of GBM patients. Quantification of NG2 expression in 74 GBM biopsies from newly diagnosed and untreated patients revealed that 50% express high NG2 levels on tumour cells and associated vessels, being associated with significantly shorter survival. This effect was independent of age at diagnosis, treatment received and hypermethylation of the O(6)-methylguanine methyltransferase (MGMT) DNA repair gene promoter. NG2 was frequently co-expressed with nestin and vimentin but rarely with CD133 and the NG2 positive tumour cells harboured genetic aberrations typical for GBM. 2D proteomics of 11 randomly selected biopsies revealed upregulation of an antioxidant, peroxiredoxin-1 (PRDX-1), in the shortest surviving patients. Expression of PRDX-1 was associated with significantly reduced products of oxidative stress. Furthermore, NG2 expressing GBM cells showed resistance to ionising radiation (IR), rapidly recognised DNA damage and effectuated cell cycle checkpoint signalling. PRDX-1 knockdown transiently slowed tumour growth rates and sensitised them to IR in vivo. Our data establish NG2 as an important prognostic factor for GBM patient survival, by mediating resistance to radiotherapy through induction of ROS scavenging enzymes and preferential DNA damage signalling.


Asunto(s)
Antígenos/biosíntesis , Biomarcadores de Tumor/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/radioterapia , Daño del ADN/genética , Glioblastoma/genética , Glioblastoma/radioterapia , Proteoglicanos/biosíntesis , Células Madre/metabolismo , Anciano , Antígenos/genética , Antígenos/efectos de la radiación , Biomarcadores de Tumor/efectos de la radiación , Neoplasias Encefálicas/patología , Daño del ADN/efectos de la radiación , Femenino , Glioblastoma/patología , Humanos , Masculino , Persona de Mediana Edad , Valor Predictivo de las Pruebas , Pronóstico , Proteoglicanos/genética , Proteoglicanos/efectos de la radiación , Tolerancia a Radiación , Radiación Ionizante , Células Madre/patología , Células Madre/efectos de la radiación , Tasa de Supervivencia/tendencias
7.
Am J Pathol ; 177(4): 1618-28, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20813964

RESUMEN

We have previously established two distinct glioma phenotypes by serial xenotransplantation of human glioblastoma (GBM) biopsies in nude rats. These tumors undergo a gradual transition from a highly invasive nonangiogenic to a less-invasive angiogenic phenotype. In a protein screen to identify molecular markers associated with the infiltrative phenotype, we identified α-basic-crystallin (αBc), a small heat-shock protein with cytoprotective properties. Its increased expression in the infiltrative phenotype was validated by immunohistochemistry and Western blots, confirming its identity to be tumor-derived and not from the host. Stereotactic human GBM biopsies taken from MRI-defined areas verified stronger αBc expression in the infiltrative edge compared to the tumor core. Cell migration assays and immunofluorescence staining showed αBc to be expressed by migrating cells in vitro. To determine αBc function, we altered its expression levels. αBc siRNA depletion caused a loss of migrating tumor cells from biopsy spheroids and delayed monolayer wound closure. In contrast, glioma cell migration in a Boyden chamber assay was unaffected by either αBc knockdown or overexpression, indicating that αBc is not functionally linked to the cell migration machinery. However, after siRNA αBc depletion, a significant sensitization of cells to various apoptotic inducers was observed (actinomycin, tumor necrosis factor α, and TNF-related apoptosis-inducing ligand [TRAIL]). In conclusion, αBc is overexpressed by highly migratory glioma cells where it plays a functional role in apoptosis resistance.


Asunto(s)
Apoptosis , Movimiento Celular , Glioblastoma/metabolismo , Glioblastoma/patología , Cadena B de alfa-Cristalina/metabolismo , Animales , Western Blotting , Encéfalo/metabolismo , Encéfalo/patología , Adhesión Celular , Proliferación Celular , Electroforesis en Gel Bidimensional , Técnica del Anticuerpo Fluorescente , Regulación Neoplásica de la Expresión Génica , Humanos , Técnicas para Inmunoenzimas , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Ratas , Ratas Desnudas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Esferoides Celulares/metabolismo , Esferoides Celulares/patología , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Análisis de Matrices Tisulares , Trasplante Heterólogo , Células Tumorales Cultivadas , Cadena B de alfa-Cristalina/antagonistas & inhibidores , Cadena B de alfa-Cristalina/genética
8.
BMC Cancer ; 9: 465, 2009 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-20040089

RESUMEN

BACKGROUND: Establishing clinically relevant animal models of glioblastoma multiforme (GBM) remains a challenge, and many commonly used cell line-based models do not recapitulate the invasive growth patterns of patient GBMs. Previously, we have reported the formation of highly invasive tumour xenografts in nude rats from human GBMs. However, implementing tumour models based on primary tissue requires that these models can be sufficiently standardised with consistently high take rates. METHODS: In this work, we collected data on growth kinetics from a material of 29 biopsies xenografted in nude rats, and characterised this model with an emphasis on neuropathological and radiological features. RESULTS: The tumour take rate for xenografted GBM biopsies were 96% and remained close to 100% at subsequent passages in vivo, whereas only one of four lower grade tumours engrafted. Average time from transplantation to the onset of symptoms was 125 days +/- 11.5 SEM. Histologically, the primary xenografts recapitulated the invasive features of the parent tumours while endothelial cell proliferations and necrosis were mostly absent. After 4-5 in vivo passages, the tumours became more vascular with necrotic areas, but also appeared more circumscribed. MRI typically revealed changes related to tumour growth, several months prior to the onset of symptoms. CONCLUSIONS: In vivo passaging of patient GBM biopsies produced tumours representative of the patient tumours, with high take rates and a reproducible disease course. The model provides combinations of angiogenic and invasive phenotypes and represents a good alternative to in vitro propagated cell lines for dissecting mechanisms of brain tumour progression.


Asunto(s)
Neoplasias Encefálicas/patología , Modelos Animales de Enfermedad , Glioblastoma/patología , Ratas Desnudas , Adulto , Anciano , Anciano de 80 o más Años , Animales , Biopsia , Femenino , Humanos , Masculino , Persona de Mediana Edad , Trasplante de Neoplasias/patología , Ratas , Reproducibilidad de los Resultados , Trasplante Heterólogo , Células Tumorales Cultivadas , Adulto Joven
9.
Int J Cancer ; 122(4): 761-8, 2008 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-17955491

RESUMEN

CD133 is a cell surface marker expressed on progenitors of haematopoietic and endothelial cell lineages. Moreover, several studies have identified CD133 as a marker of brain tumor-initiating cells. In this study, human glioblastoma multiforme biopsies were engrafted intracerebrally into nude rats. The resulting tumors were serially passaged in vivo, and monitored by magnetic resonance imaging. CD133 expression was analyzed at various passages. Tumors initiated directly from the biopsies expressed little or no CD133, and showed no contrast enhancement suggesting an intact blood-brain barrier. During passaging, the tumors gradually displayed more contrast enhancement, increased angiogenesis and a shorter survival. Real-time qPCR and immunoblots showed that this was accompanied by increased CD133 expression. Primary biopsy spheroids and xenograft tumors were subsequently dissociated and flow sorted into CD133 negative and CD133 positive cell populations. Both populations incorporated BrdU in cell culture, and expressed the neural precursor marker nestin. Notably, CD133 negative cells derived from 6 different patients were tumorgenic when implanted into the rat brains. For 3 of these patients, analysis showed that the resulting tumors contained CD133 positive cells. In conclusion, we show that CD133 negative glioma cells are tumorgenic in nude rats, and that CD133 positive cells can be obtained from these tumors. Upon passaging of the tumors in vivo, CD133 expression is upregulated, coinciding with the onset of angiogenesis and a shorter survival. Thus, our findings do not suggest that CD133 expression is required for brain tumor initiation, but that it may be involved during brain tumor progression.


Asunto(s)
Antígenos CD/metabolismo , Neoplasias Encefálicas/metabolismo , Proliferación Celular , Glioblastoma/metabolismo , Glicoproteínas/metabolismo , Péptidos/metabolismo , Antígeno AC133 , Animales , Antígenos CD/genética , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Progresión de la Enfermedad , Citometría de Flujo , Glioblastoma/genética , Glioblastoma/patología , Glicoproteínas/genética , Humanos , Técnicas para Inmunoenzimas , Imagen por Resonancia Magnética , Neovascularización Patológica , Péptidos/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Desnudas , Células Tumorales Cultivadas
10.
Cancer Res ; 66(20): 9895-902, 2006 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-17047051

RESUMEN

By serial transplantation of human glioblastoma biopsies into the brain of immunodeficient nude rats, two different tumor phenotypes were obtained. Initially, the transplanted xenografts displayed a highly invasive phenotype that showed no signs of angiogenesis. By serial transplantation in animals, the tumors changed to a less invasive, predominantly angiogenic phenotype. To identify novel proteins related to the invasive phenotype, the xenografts were analyzed using a global proteomics approach. One of the identified proteins was protein disulfide isomerase (PDI) A6 precursor. PDI is a chaperone protein that mediates integrin-dependent cell adhesion. It is both present in the cytosol and at the cell surface. We show that PDI is strongly expressed on invasive glioma cells, in both xenografts and at the invasive front of human glioblastomas. Using an in vitro migration assay, we also show that PDI is expressed on migrating glioma cells. To determine the functional significance of PDI in cell migration, we tested the effect of a PDI inhibitor, bacitracin, and a PDI monoclonal antibody on glioma cell migration and invasion in vitro. Both tumor spheroids derived from human glioblastoma xenografts in nude rat brain and cell line spheroids were used. The PDI antibody, as well as bacitracin, inhibited tumor cell migration and invasion. The anti-invasive effect of bacitracin was reversible after withdrawal of the inhibitor, indicating a specific, nontoxic effect. In conclusion, using a global proteomics approach, PDI was identified to play an important role in glioma cell invasion, and its action was effectively inhibited by bacitracin.


Asunto(s)
Glioblastoma/enzimología , Glioblastoma/patología , Proteína Disulfuro Isomerasas/biosíntesis , Animales , Anticuerpos/farmacología , Bacitracina/farmacología , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Sinergismo Farmacológico , Humanos , Integrina beta3/inmunología , Invasividad Neoplásica , Proteína Disulfuro Isomerasas/antagonistas & inhibidores , Proteómica , Ratas , Ratas Desnudas , Esferoides Celulares , Trasplante Heterólogo
11.
Pediatr Neurosurg ; 41(6): 300-4, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16293949

RESUMEN

BACKGROUND: Shunt failure is by far the most frequent problem in children with shunts, and most of them will experience this condition at some point in their lives. In order to identify causes of shunt failure, and to compare multi-component and one-piece shunt systems, we analyzed retrospectively all pediatric shunt procedures in our Department during an 11-year period. The study does not deal with shunt infections. METHODS: We reviewed the records of all pediatric shunting procedures between January 1986 and December 1996. RESULTS: The study included 161 children operated for hydrocephalus with a total of 431 procedures. The procedures included 124 (29%) primary insertions, 10 (2%) reinsertions and 297 (69%) revisions; 206 (69%) of the revisions were due to shunt failures, of which 74 (36%) were caused by the failure of the surgical technique (misplaced ventricular catheters, disconnected shunts, or misplaced peritoneal catheters). CONCLUSIONS: Improvement of the surgical technique may reduce the incidence of shunt failures and revisions. The results obtained in a small department like ours do not seem to differ substantially from those obtained in more specialized departments with a larger patient group. Practical measures that may reduce the risk of shunt failures are suggested.


Asunto(s)
Derivaciones del Líquido Cefalorraquídeo/efectos adversos , Hidrocefalia/cirugía , Complicaciones Posoperatorias/prevención & control , Adolescente , Niño , Falla de Equipo , Femenino , Humanos , Masculino , Complicaciones Posoperatorias/cirugía , Reoperación/estadística & datos numéricos , Factores de Riesgo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA