Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2023 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-37398244

RESUMEN

The acquisition of invasive properties is a prerequisite for tumor progression and metastasis. Molecular subtypes of KRAS-driven lung cancer exhibit distinct modes of invasion that likely contribute to unique growth properties and therapeutic susceptibilities. Despite this, pre-clinical discovery strategies designed to exploit invasive phenotypes are lacking. To address this, we designed an experimental system to screen for targetable signaling pathways linked to active early invasion phenotypes in the two most prominent molecular subtypes, TP53 and LKB1, of KRAS-driven lung adenocarcinoma (LUAD). By combining live-cell imaging of human bronchial epithelial cells in a 3D invasion matrix with RNA transcriptome profiling, we identified the LKB1-specific upregulation of bone morphogenetic protein 6 (BMP6). Examination of early-stage lung cancer patients confirmed upregulation of BMP6 in LKB1-mutant lung tumors. At the molecular level, we find that the canonical iron regulatory hormone Hepcidin is induced via BMP6 signaling upon LKB1 loss, where intact LKB1 kinase activity is necessary to maintain signaling homeostasis. Furthermore, pre-clinical studies in a novel Kras/Lkb1-mutant syngeneic mouse model show that potent growth suppression was achieved by inhibiting the ALK2/BMP6 signaling axis with single agents that are currently in clinical trials. We show that alterations in the iron homeostasis pathway are accompanied by simultaneous upregulation of ferroptosis protection proteins. Thus, LKB1 is sufficient to regulate both the 'gas' and 'breaks' to finely tune iron-regulated tumor progression.

2.
Cell Rep ; 23(6): 1651-1664, 2018 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-29742423

RESUMEN

Cancer stem cells promote neoplastic growth, in part by deregulating asymmetric cell division and enhancing self-renewal. To uncover mechanisms and potential therapeutic targets in glioma stem cell (GSC) self-renewal, we performed a genetic suppressor screen for kinases to reverse the tumor phenotype of our Drosophila brain tumor model and identified dCdk5 as a critical regulator. CDK5, the human ortholog of dCdk5 (79% identity), is aberrantly activated in GBMs and tightly aligned with both chromosome 7 gains and stem cell markers affecting tumor-propagation. Our investigation revealed that pharmaceutical inhibition of CDK5 prevents GSC self-renewal in vitro and in xenografted tumors, at least partially by suppressing CREB1 activation independently of PKA/cAMP. Finally, our TCGA GBM data analysis revealed that CDK5, stem cell, and asymmetric cell division markers segregate within non-mesenchymal patient clusters, which may indicate preferential dependence on CDK5 signaling and sensitivity to its inhibition in this group.


Asunto(s)
Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Quinasa 5 Dependiente de la Ciclina/antagonistas & inhibidores , Proteínas de Drosophila/antagonistas & inhibidores , Glioma/metabolismo , Glioma/patología , Células Madre Neoplásicas/patología , Transducción de Señal , Animales , División Celular Asimétrica/efectos de los fármacos , Biomarcadores de Tumor/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Proliferación Celular/efectos de los fármacos , Autorrenovación de las Células/efectos de los fármacos , Quinasa 5 Dependiente de la Ciclina/genética , Quinasa 5 Dependiente de la Ciclina/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioma/genética , Mesodermo/efectos de los fármacos , Mesodermo/patología , Ratones , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Fosforilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Sci Rep ; 7(1): 15593, 2017 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-29142297

RESUMEN

Glioblastoma (GBM) contains diverse microenvironments with uneven distributions of oncogenic alterations and signaling networks. The diffusely infiltrative properties of GBM result in residual tumor at neurosurgical resection margins, representing the source of relapse in nearly all cases and suggesting that therapeutic efforts should be focused there. To identify signaling networks and potential druggable targets across tumor microenvironments (TMEs), we utilized 5-ALA fluorescence-guided neurosurgical resection and sampling, followed by proteomic analysis of specific TMEs. Reverse phase protein array (RPPA) was performed on 205 proteins isolated from the tumor margin, tumor bulk, and perinecrotic regions of 13 previously untreated, clinically-annotated and genetically-defined high grade gliomas. Differential protein and pathway signatures were established and then validated using western blotting, immunohistochemistry, and comparable TCGA RPPA datasets. We identified 37 proteins differentially expressed across high-grade glioma TMEs. We demonstrate that tumor margins were characterized by pro-survival and anti-apoptotic proteins, whereas perinecrotic regions were enriched for pro-coagulant and DNA damage response proteins. In both our patient cohort and TCGA cases, the data suggest that TMEs possess distinct protein expression profiles that are biologically and therapeutically relevant.


Asunto(s)
Glioblastoma/genética , Recurrencia Local de Neoplasia/genética , Neoplasia Residual/genética , Proteómica , Adulto , Anciano , Ácido Aminolevulínico/administración & dosificación , Receptores ErbB/genética , Femenino , Fluorescencia , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , Glioblastoma/cirugía , Humanos , Masculino , Márgenes de Escisión , Persona de Mediana Edad , Recurrencia Local de Neoplasia/tratamiento farmacológico , Recurrencia Local de Neoplasia/metabolismo , Recurrencia Local de Neoplasia/cirugía , Neoplasia Residual/tratamiento farmacológico , Neoplasia Residual/patología , Neoplasia Residual/cirugía , Fosfohidrolasa PTEN/genética , Análisis por Matrices de Proteínas , Transducción de Señal/efectos de los fármacos , Microambiente Tumoral/genética
4.
Mol Oncol ; 11(3): 280-294, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28100038

RESUMEN

Glioblastoma (GBM) is the most malignant form of primary brain tumor, and GBM stem-like cells (GSCs) contribute to the rapid growth, therapeutic resistance, and clinical recurrence of these fatal tumors. STAT3 signaling supports the maintenance and proliferation of GSCs, yet regulatory mechanisms are not completely understood. Here, we report that tri-partite motif-containing protein 8 (TRIM8) activates STAT3 signaling to maintain stemness and self-renewing capabilities of GSCs. TRIM8 (also known as 'glioblastoma-expressed ring finger protein') is expressed equally in GBM and normal brain tissues, despite its hemizygous deletion in the large majority of GBMs, and its expression is highly correlated with stem cell markers. Experimental knockdown of TRIM8 reduced GSC self-renewal and expression of SOX2, NESTIN, and p-STAT3, and promoted glial differentiation. Overexpression of TRIM8 led to higher expression of p-STAT3, c-MYC, SOX2, NESTIN, and CD133, and enhanced GSC self-renewal. We found that TRIM8 activates STAT3 by suppressing the expression of PIAS3, an inhibitor of STAT3, most likely through E3-mediated ubiquitination and proteasomal degradation. Interestingly, we also found that STAT3 activation upregulates TRIM8, providing a mechanism for normalized TRIM8 expression in the setting of hemizygous gene deletion. These data demonstrate that bidirectional TRIM8-STAT3 signaling regulates stemness in GSC.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Encéfalo/patología , Proteínas Portadoras/metabolismo , Glioblastoma/metabolismo , Chaperonas Moleculares/metabolismo , Células Madre Neoplásicas/patología , Proteínas del Tejido Nervioso/metabolismo , Proteínas Inhibidoras de STAT Activados/metabolismo , Factor de Transcripción STAT3/metabolismo , Encéfalo/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Proteínas Portadoras/genética , Línea Celular , Línea Celular Tumoral , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Glioblastoma/genética , Glioblastoma/patología , Humanos , Chaperonas Moleculares/genética , Células Madre Neoplásicas/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas Inhibidoras de STAT Activados/genética , Interferencia de ARN , ARN Interferente Pequeño/genética , Factor de Transcripción STAT3/genética , Transducción de Señal
5.
FASEB J ; 30(12): 4098-4108, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27572958

RESUMEN

We investigated the intersection of epidermal growth factor receptor (EGFR) and CCAAT enhancer binding protein (C/EBP)-ß signaling in glioblastoma (GBM), given that both gene products strongly influence neoplastic behavior. C/EBP-ß is known to drive the mesenchymal transcriptional signature in GBM, likely through strong microenvironmental influences, whereas the genetic contributions to its up-regulation in this disease are not well described. We demonstrated that stable overexpression and activation of WT EGFR (U87MG-WT) led to elevated C/EBP-ß expression, as well as enhanced nuclear translocation and DNA-binding activity, leading to up-regulation of C/EBP-ß transcription and translation. Deeper investigation identified bidirectional regulation, with C/EBP-ß also causing up-regulation of EGFR that was at least partially dependent on the STAT3. Based on ChIP-based studies, we also found that that the translational isoforms of C/EBP-ß [liver-enriched transcription-activating protein (LAP)-1/2 and liver inhibitory protein (LIP)] have differential occupancy on STAT3 promoter and opposing roles in transcriptional regulation of STAT3 and EGFR. We further demonstrated that the shorter C/EBP-ß isoform, LIP, promoted proliferation and migration of U87MG glioma cells, potentially via induction of cytokine IL-6. Our molecular dissection of EGFR and C/EBP-ß pathway interactions uncovered a complex signaling network in which increased activity of either EGFR or C/EBP-ß leads to the up-regulation of the other, enhancing oncogenic signaling. Disrupting the EGFR-C/EBP-ß signaling axis could attenuate malignant behavior of glioblastoma.-Selagea, L., Mishra, A., Anand, M., Ross, J., Tucker-Burden, C., Kong, J., Brat, D. J. EGFR and C/EBP-ß oncogenic signaling is bidirectional in human glioma and varies with the C/EBP-ß isoform.


Asunto(s)
Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Receptores ErbB/metabolismo , Regulación de la Expresión Génica/fisiología , Glioma/metabolismo , Transducción de Señal , Células Cultivadas , Glioma/genética , Humanos , Regiones Promotoras Genéticas/genética , Isoformas de Proteínas/metabolismo , Transducción de Señal/fisiología , Transcripción Genética/genética , Activación Transcripcional/fisiología
6.
Cancer Res ; 76(8): 2443-52, 2016 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-26893479

RESUMEN

Cancer stem cells exert enormous influence on neoplastic behavior, in part by governing asymmetric cell division and the balance between self-renewal and multipotent differentiation. Growth is favored by deregulated stem cell division, which enhances the self-renewing population and diminishes the differentiation program. Mutation of a single gene in Drosophila, Brain Tumor (Brat), leads to disrupted asymmetric cell division resulting in dramatic neoplastic proliferation of neuroblasts and massive larval brain overgrowth. To uncover the mechanisms relevant to deregulated cell division in human glioma stem cells, we first developed a novel adult Drosophila brain tumor model using brat-RNAi driven by the neuroblast-specific promoter inscuteable Suppressing Brat in this population led to the accumulation of actively proliferating neuroblasts and a lethal brain tumor phenotype. brat-RNAi caused upregulation of Notch signaling, a node critical for self-renewal, by increasing protein expression and enhancing nuclear transport of Notch intracellular domain (NICD). In human glioblastoma, we demonstrated that the human ortholog of Drosophila Brat, tripartite motif-containing protein 3 (TRIM3), similarly suppressed NOTCH1 signaling and markedly attenuated the stem cell component. We also found that TRIM3 suppressed nuclear transport of active NOTCH1 (NICD) in glioblastoma and demonstrated that these effects are mediated by direct binding of TRIM3 to the Importin complex. Together, our results support a novel role for Brat/TRIM3 in maintaining stem cell equilibrium and suppressing tumor growth by regulating NICD nuclear transport. Cancer Res; 76(8); 2443-52. ©2016 AACR.


Asunto(s)
Neoplasias Encefálicas/patología , Proteínas Portadoras/fisiología , Núcleo Celular/metabolismo , Proteínas de Unión al ADN/fisiología , Proteínas de Drosophila/fisiología , Células Madre Neoplásicas/patología , Receptores Notch/metabolismo , Animales , Neoplasias Encefálicas/metabolismo , Carcinogénesis , Drosophila , Humanos , Transporte de Proteínas , Interferencia de ARN
7.
Proc IEEE Int Symp Biomed Imaging ; 2015: 1212-1215, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26405506

RESUMEN

A large number of cell-oriented cancer investigations require an effective and reliable cell segmentation method on three dimensional (3D) fluorescence microscopic images for quantitative analysis of cell biological properties. In this paper, we present a fully automated cell segmentation method that can detect cells from 3D fluorescence microscopic images. Enlightened by fluorescence imaging techniques, we regulated the image gradient field by gradient vector flow (GVF) with interpolated and smoothed data volume, and grouped voxels based on gradient modes identified by tracking GVF field. Adaptive thresholding was then applied to voxels associated with the same gradient mode where voxel intensities were enhanced by a multiscale cell filter. We applied the method to a large volume of 3D fluorescence imaging data of human brain tumor cells with (1) small cell false detection and missing rates for individual cells; and (2) trivial over and under segmentation incidences for clustered cells. Additionally, the concordance of cell morphometry structure between automated and manual segmentation was encouraging. These results suggest a promising 3D cell segmentation method applicable to cancer studies.

8.
Cancer Res ; 74(16): 4536-48, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-24947043

RESUMEN

Cancer stem cells, capable of self-renewal and multipotent differentiation, influence tumor behavior through a complex balance of symmetric and asymmetric cell divisions. Mechanisms regulating the dynamics of stem cells and their progeny in human cancer are poorly understood. In Drosophila, mutation of brain tumor (brat) leads to loss of normal asymmetric cell division by developing neural cells and results in a massively enlarged brain composed of neuroblasts with neoplastic properties. Brat promotes asymmetric cell division and directs neural differentiation at least partially through its suppression on Myc. We identified TRIM3 (11p15.5) as a human ortholog of Drosophila brat and demonstrate its regulation of asymmetric cell division and stem cell properties of glioblastoma (GBM), a highly malignant human brain tumor. TRIM3 gene expression is markedly reduced in human GBM samples, neurosphere cultures, and cell lines and its reconstitution impairs growth properties in vitro and in vivo. TRIM3 expression attenuates stem-like qualities of primary GBM cultures, including neurosphere formation and the expression of stem cell markers CD133, Nestin, and Nanog. In GBM stem cells, TRIM3 expression leads to a greater percentage dividing asymmetrically rather than symmetrically. As with Brat in Drosophila, TRIM3 suppresses c-Myc expression and activity in human glioma cell lines. We also demonstrate a strong regulation of Musashi-Notch signaling by TRIM3 in GBM neurospheres and neural stem cells that may better explain its effect on stem cell dynamics. We conclude that TRIM3 acts as a tumor suppressor in GBM by restoring asymmetric cell division.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Proteínas Portadoras/metabolismo , Glioblastoma/metabolismo , Animales , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Proteínas Portadoras/biosíntesis , Proteínas Portadoras/genética , Procesos de Crecimiento Celular/fisiología , Femenino , Glioblastoma/genética , Glioblastoma/patología , Xenoinjertos , Humanos , Ratones Desnudos , Transducción de Señal , Transfección
9.
Sci Signal ; 6(283): ra55, 2013 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-23838182

RESUMEN

The gene that encodes the epidermal growth factor receptor (EGFR) is frequently overexpressed or mutated in human cancers, including glioblastoma. However, the efficacy of EGFR-targeted small-molecule inhibitors or monoclonal antibodies in glioblastomas that also have mutation or deletion of the gene encoding phosphatase and tensin homolog (PTEN) has been modest. We found that EGFR signaling was blocked by a small molecule (G5-7) that selectively inhibited Janus kinase 2 (JAK2)-mediated phosphorylation and activation of EGFR and STAT3 (signal transducer and activator of transcription 3) by binding to JAK2, thereby decreasing the activity of downstream signaling by mTOR (mammalian target of rapamycin) and inducing cell cycle arrest. G5-7 inhibited the proliferation of PTEN-deficient glioblastoma cell lines harboring a constitutively active variant of EGFR (U87MG/EGFRvIII) and human glioblastoma explant neurosphere cultures, but the drug only weakly inhibited the proliferation of either glioblastoma cell lines that were wild type for EGFR and stably transfected with PTEN (U87MG/PTEN) or normal neural progenitor cells and astrocytes. Additionally, G5-7 reduced vascular endothelial growth factor (VEGF) secretion and endothelial cell migration and induced apoptosis in glioblastoma xenografts, thereby suppressing glioblastoma growth in vivo. Furthermore, G5-7 was more potent than EGFR or JAK2 inhibitors that interfere with either ligand or adenosine 5'-triphosphate (ATP) binding at impeding glioblastoma cell proliferation, demonstrating that this allosteric JAK2 inhibitor may be an effective clinical strategy.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Glioma/tratamiento farmacológico , Janus Quinasa 2/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal/efectos de los fármacos , Regulación Alostérica/efectos de los fármacos , Regulación Alostérica/genética , Animales , Línea Celular Tumoral , Receptores ErbB/genética , Receptores ErbB/metabolismo , Eliminación de Gen , Glioma/enzimología , Glioma/genética , Glioma/patología , Xenoinjertos , Humanos , Janus Quinasa 2/genética , Janus Quinasa 2/metabolismo , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Células-Madre Neurales/enzimología , Células-Madre Neurales/patología , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Fosforilación , Inhibidores de Proteínas Quinasas/química , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/genética
10.
Stem Cells Dev ; 21(13): 2374-86, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22435486

RESUMEN

Glioblastoma (GBM) is a highly aggressive primary brain tumor with a poor prognosis. Despite aggressive therapy with surgery, radiotherapy, and chemotherapy, nearly all patients succumb to disease within 2 years. Several studies have supported the presence of stem-like cells in brain tumor cultures that are CD133-positive, are capable of self-renewal, and give rise to all cell types found within the tumor, potentially perpetuating growth. CD133 is a widely accepted marker for glioma-derived cancer stem cells; however, its reliability has been questioned, creating a need for other identifiers of this biologically important subpopulation. We used a panel of 20 lectins to identify differences in glycan expression found in the glycocalyx of undifferentiated glioma-derived stem cells and differentiated cells that arise from them. Fluorescently labeled lectins that specifically recognize α-N-acetylgalactosamine (GalNAc) and α-N-acetylglucosamine (GlcNAc) differentially bound to the cell surface based on the state of cellular differentiation. GalNAc and GlcNAc were highly expressed on the surface of undifferentiated cells and showed markedly reduced expression over a 12-day duration of differentiation. Additionally, the GalNAc-recognizing lectin Dolichos biflorus agglutinin was capable of specifically selecting and sorting glioma-derived stem cell populations from an unsorted tumor stock and this subpopulation had proliferative properties similar to CD133(+) cells in vitro and also had tumor-forming capability in vivo. Our preliminary results on a single cerebellar GBM suggest that GalNAc and GlcNAc are novel biomarkers for identifying glioma-derived stem cells and can be used to isolate cancer stem cells from unsorted cell populations, thereby creating new cell lines for research or clinical testing.


Asunto(s)
Biomarcadores de Tumor/análisis , Glioblastoma/diagnóstico , Lectinas/metabolismo , Células Madre Neoplásicas/metabolismo , Polisacáridos/análisis , Antígeno AC133 , Acetilgalactosamina/metabolismo , Acetilglucosamina/metabolismo , Antígenos CD/metabolismo , Diferenciación Celular , Línea Celular Tumoral , Proliferación Celular , Citometría de Flujo , Glioblastoma/metabolismo , Glicocálix/metabolismo , Glicoproteínas/metabolismo , Humanos , Inmunohistoquímica , Péptidos/metabolismo , Lectinas de Plantas/metabolismo
12.
Transplantation ; 88(2): 160-9, 2009 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-19623010

RESUMEN

BACKGROUND: The long-term metabolic function of microencapsulated xenogeneic adult porcine islets (API) was assessed in a murine model of type 1 diabetes mellitus. METHODS: API were encapsulated in barium-gelled alginate and transplanted intraperitoneally in diabetic nonobese diabetic (NOD) mice given no immunosuppression or given costimulatory blockade (CoB; CTLA4-Ig+anti-CD154 mAb). Control mice received nonencapsulated API under the kidney capsule. Graft function was monitored by measurement of random blood glucose levels, serum glycosylated hemoglobin (HbA1c), serum porcine C peptide, in vivo glucose tolerance tests, and histologic analyses of host pancreas and graft biopsies. Host immune responses to the islet xenografts were characterized by phenotyping peritoneal cellular infiltrates and by measuring serum antiporcine antibody levels. RESULTS: Without immunosuppression, nonencapsulated API functioned for less than 1 week, and microencapsulated API functioned for 35+/-14 days before rejection, associated with both a cellular and a humoral immune response. With continuous CoB, nonencapsulated API functioned for 27+/-4 days, whereas microencapsulated API functioned for >450 days with measurable levels of serum porcine C peptide, near normal in vivo glucose tolerance tests and HbA1c levels, and intact microcapsules containing viable, insulin-positive porcine islets. CONCLUSIONS: Microencapsulated API restored normoglycemia for more than 1 year in spontaneously diabetic NODs given dual CoB. To our knowledge, this is the first study to document long-term normalized HbA1c, porcine C peptide, and near normal glucose tolerance in immunosuppressed diabetic NOD mice transplanted intraperitoneally with microencapsulated API. Our study suggests that transplantation of microencapsulated porcine islet xenografts may be a future treatment for patients with type 1 diabetes mellitus.


Asunto(s)
Diabetes Mellitus Tipo 1/cirugía , Supervivencia de Injerto/inmunología , Trasplante de Islotes Pancreáticos/inmunología , Tolerancia al Trasplante/inmunología , Trasplante Heterólogo/inmunología , Animales , Péptido C/sangre , Prueba de Tolerancia a la Glucosa , Hemoglobina Glucada/análisis , Rechazo de Injerto/inmunología , Ratones , Ratones Endogámicos NOD , Porcinos
13.
Cancer Res ; 69(6): 2540-9, 2009 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-19276385

RESUMEN

Hypoxia and necrosis are fundamental features of glioblastoma (GBM) and their emergence is critical for the rapid biological progression of this fatal tumor; yet, underlying mechanisms are poorly understood. We have suggested that vaso-occlusion following intravascular thrombosis could initiate or propagate hypoxia and necrosis in GBM. Tissue factor (TF), the main cellular initiator of coagulation, is overexpressed in GBMs and likely favors a thrombotic microenvironment. Epidermal growth factor receptor (EGFR) amplification and PTEN loss are two common genetic alterations seen in GBM but not in lower-grade astrocytomas that could be responsible for TF up-regulation. The most frequent EGFR mutation in GBM involves deletion of exons 2 to 7, resulting in the expression of a constitutively active receptor, EGFRvIII. Here, we show that overexpression of EGFR or EGFRvIII in human glioma cells causes increased basal TF expression and that stimulation of EGFR by its ligand, EGF, leads to a marked dose-dependent up-regulation of TF. In all cases, increased TF expression led to accelerated plasma coagulation in vitro. EGFR-mediated TF expression depended most strongly on activator protein-1 (AP-1) transcriptional activity and was associated with c-Jun NH(2)-terminal kinase (JNK) and JunD activation. Restoration of PTEN expression in PTEN-deficient GBM cells diminished EGFR-induced TF expression by inhibiting JunD/AP-1 transcriptional activity. PTEN mediated this effect by antagonizing phosphatidylinositol 3-kinase activity, which in turn attenuated both Akt and JNK activities. These mechanisms are likely at work in vivo, as EGFR expression was highly correlated with TF expression in human high-grade astrocytoma specimens.


Asunto(s)
Neoplasias Encefálicas/genética , Receptores ErbB/genética , Glioblastoma/genética , Fosfohidrolasa PTEN/genética , Proteínas Proto-Oncogénicas c-jun/genética , Tromboplastina/biosíntesis , Factor de Transcripción AP-1/genética , Neoplasias Encefálicas/enzimología , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/biosíntesis , Receptores ErbB/metabolismo , Glioblastoma/enzimología , Glioblastoma/metabolismo , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Sistema de Señalización de MAP Quinasas , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteína Oncogénica v-akt/metabolismo , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-jun/biosíntesis , Transducción de Señal , Tromboplastina/genética , Factor de Transcripción AP-1/biosíntesis , Transcripción Genética , Regulación hacia Arriba
14.
Cancer Res ; 69(3): 1212-20, 2009 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-19176395

RESUMEN

Angiogenesis is a critical physiologic process that is appropriated during tumorigenesis. Little is known about how this process is specifically regulated in the brain. Brain angiogenesis inhibitor-1 (BAI1) is a brain-predominant seven-transmembrane protein that contains five antiangiogenic thrombospondin type-1 repeats (TSR). We recently showed that BAI1 is cleaved at a conserved proteolytic cleavage site releasing a soluble, 120 kDa antiangiogenic factor called vasculostatin (Vstat120). Vstat120 has been shown to inhibit in vitro angiogenesis and suppress subcutaneous tumor growth. Here, we examine its effect on the intracranial growth of malignant gliomas and further study its antitumor mechanism. First, we show that expression of Vstat120 strongly suppresses the intracranial growth of malignant gliomas, even in the presence of the strong proangiogenic stimulus mediated by the oncoprotein epidermal growth factor receptor variant III (EGFRvIII). This tumor-suppressive effect is accompanied by a decrease in tumor vascular density, suggesting a potent antiangiogenic effect in the brain. Second, and consistent with this interpretation, we find that treatment with Vstat120 reduces the migration of cultured microvascular endothelial cells in vitro and inhibits corneal angiogenesis in vivo. Third, we show that these antivascular effects critically depend on the presence of the cell surface receptor CD36 on endothelial cells in vitro and in vivo, supporting the role of Vstat120 TSRs in mediating these effects. These results advance the understanding of brain-specific angiogenic regulation, and suggest that Vstat120 has therapeutic potential in the treatment of brain tumors and other intracerebral vasculopathies.


Asunto(s)
Proteínas Angiogénicas/biosíntesis , Neoplasias Encefálicas/irrigación sanguínea , Neoplasias Encefálicas/terapia , Antígenos CD36/metabolismo , Glioblastoma/irrigación sanguínea , Glioblastoma/terapia , Fragmentos de Péptidos/biosíntesis , Proteínas Angiogénicas/genética , Proteínas Angiogénicas/metabolismo , Animales , Neoplasias Encefálicas/genética , Línea Celular Tumoral , Movimiento Celular/fisiología , Neovascularización de la Córnea/tratamiento farmacológico , ADN Complementario/administración & dosificación , ADN Complementario/genética , Células Endoteliales/patología , Glioblastoma/genética , Humanos , Ratones , Ratones Desnudos , Neovascularización Patológica/metabolismo , Neovascularización Patológica/terapia , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Ratas , Receptores Acoplados a Proteínas G , Transfección , Ensayos Antitumor por Modelo de Xenoinjerto
15.
J Diabetes Sci Technol ; 2(5): 760-7, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19885258

RESUMEN

BACKGROUND: If alginate microcapsules are to be used clinically for therapeutic cell transplants, capsule formulations must be designed to enhance optimal biocompatibility and immune acceptance. METHODS: Microcapsules were generated using highly purified, endotoxin-free, ultra-low viscosity, high mannuronic acid alginate. The capsules differed with respect to gelling cation (50 mM barium or 100 mM calcium), alginate concentration (2.0% or 3.3%), alginate density (homogeneous or inhomogeneous), and the presence or absence poly-L-lysine (PLL) coating. Four types of empty capsules were implanted intraperitoneally (i.p.) in normal NOD mice, and their biocompatibility was evaluated after various time periods in vivo. Encapsulated adult porcine islets (APIs) were transplanted i.p. in diabetic NOD mice, and immune acceptance was evaluated by graft survival times, host cell adherence to capsule surfaces, and flow cytometric analysis of peritoneal host cells. RESULTS: All empty alginate capsules were biocompatible in vivo, but barium-gelled alginate capsules without PLL were clearly the most biocompatible, since 99% of these empty capsules had no host cell adherence up to 9 months in vivo. In diabetic NOD mice, APIs functioned significantly longer in barium-alginate capsules without PLL than in calcium-alginate capsules with PLL and had strikingly less host cell adherence, although large numbers of host cells (predominantly macrophages and eosinophils) infiltrated the peritoneal cavities of recipients with APIs in both types of capsules. Addition of PLL coatings to barium-alginate capsules dramatically decreased graft survival. CONCLUSIONS: Inhomogeneous barium-gelled alginate capsules without PLL are the optimal candidates for clinical trials, based on their enhanced biocompatibility and immune acceptance in vivo.

16.
Biomaterials ; 28(4): 609-17, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17055571

RESUMEN

The cell membrane establishes an important paradigm for the molecular engineering of coatings for implantable devices because of its intrinsic biocompatibility and ability to act as a template for the assembly of diverse membrane-associated macromolecules. A stabilized membrane-mimetic film was assembled on alginate/Ca(2+) hydrogel microcapsules by in situ polymerization of an acrylate functionalized phospholipid. The phospholipid monomer was prepared as unilamellar vesicles and fused onto octadecyl chains that were components of an amphiphilic terpolymer anchored onto a polyelectrolyte multilayer (PEM) by electrostatic interactions. Microcapsules coated with a membrane-mimetic film were implanted into the peritoneal cavity of C57BL/6 mice, and the short-term biostability and biocompatibility of membrane-mimetic films assembled on two different alginate/poly(l-lysine) PEM cushions were compared. The nature of the underlying PEM support had a profound impact on the biocompatibility of the membrane-mimetic film, as the percentage of retrieved microcapsules completely overgrown with host cells shifted from 66+/-5.9% to less than 1% when modifications to the PEM were made. When assembled on the appropriate PEM support, biocompatibility of membrane-mimetic-coated microspheres was high wherein 87.5+/-5.7% of the implanted microspheres were retrieved 4 weeks after implantation and 92.6+/-6.4% of the retrieved capsules were free of cell adhesion or fibrotic overgrowth. Finally, 4 weeks after implantation, microspheres coated with a Texas red-labeled membrane-mimetic film were imaged with confocal microscopy and exhibited a uniform film around the periphery of the implant, indicating a high degree of film biostability. Hence, membrane-mimetic films provide a new route for generating robust, biocompatible, and biochemically heterogeneous coatings for implantable devices through molecular self-assembly.


Asunto(s)
Materiales Biocompatibles/química , Materiales Biocompatibles/metabolismo , Materiales Biomiméticos/química , Materiales Biomiméticos/metabolismo , Polímeros/química , Alginatos/química , Animales , Materiales Biocompatibles/administración & dosificación , Materiales Biomiméticos/administración & dosificación , Electrólitos/química , Ácido Glucurónico/química , Ácidos Hexurónicos/química , Masculino , Ratones , Ratones Endogámicos C57BL , Estructura Molecular , Polímeros/administración & dosificación , Polímeros/metabolismo , Especificidad por Sustrato
17.
Biochem Biophys Res Commun ; 340(1): 236-43, 2006 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-16375863

RESUMEN

Our goal is to develop effective islet grafts for treating type 1 diabetes. Since human islets are scarce, we evaluated the efficacy of a microencapsulated insulin-secreting conditionally transformed allogeneic beta-cell line (betaTC-tet) in non-obese diabetic mice treated with tetracycline to inhibit cell growth. Relatively low serum levels of tetracycline controlled proliferation of betaTC-tet cells without inhibiting effective control of hyperglycemia in recipients. There was no significant host cellular reaction to the allografts or host cell adherence to microcapsules, and host cytokine levels were similar to those of sham-operated controls. We conclude that encapsulated allogeneic beta-cell lines may be clinically relevant, because they effectively restore euglycemia and do not elicit a strong cellular immune response following transplantation. To our knowledge, this is the first extensive characterization of the kinetics of host cellular and cytokine responses to an encapsulated islet cell line in an animal model of type 1 diabetes.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Citocinas/inmunología , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/cirugía , Reacción Injerto-Huésped/inmunología , Trasplante de Islotes Pancreáticos/inmunología , Trasplante de Islotes Pancreáticos/métodos , Animales , Línea Celular , Diabetes Mellitus Tipo 1/patología , Ratones
18.
Transplantation ; 79(4): 409-18, 2005 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-15729166

RESUMEN

BACKGROUND: Transplantation of human islets has been successful clinically. Since human islets are scarce, we are studying microencapsulated porcine islet xenografts in nonobese diabetic (NOD) mice. We have evaluated the cellular immune response in NOD mice with and without dual costimulatory blockade. METHODS: Alginate-poly-L-lysine-encapsulated adult porcine islets were transplanted i.p. in untreated diabetic NODs and NODs treated with CTLA4-Ig to block CD28/B7 and with anti-CD154 mAb to inhibit CD40/CD40-ligand interactions. Groups of mice were sacrificed on subsequent days; microcapsules were evaluated by histology; peritoneal cells were analyzed by FACS; and peritoneal cytokines were quantified by ELISA. Controls included immunoincompetent NOD-Scids and diabetic NODs given sham surgery or empty microcapsules. RESULTS: Within 20 days, encapsulated porcine islets induced accumulation of large numbers of macrophages, eosinophils, and significant numbers of CD4 and CD8 T cells at the graft site, and all grafts were rejected. During rejection, IFNgamma, IL-12 and IL-5 were significantly elevated over sham-operated controls, whereas IL-2, TNFalpha, IL-4, IL-6, IL-10, IL-1beta and TGFbeta were unchanged. Treatment with CTLA4-Ig and anti-CD154 prevented graft destruction in all animals during the 26 days of the experiment, dramatically inhibited recruitment of host inflammatory cells, and inhibited peritoneal IFNgamma and IL-5 concentrations while delaying IL-12 production. CONCLUSIONS: When two different pathways of T cell costimulation were blocked, T cell-dependent inflammatory responses were inhibited, and survival of encapsulated islet xenografts was significantly prolonged. These findings suggest synergy between encapsulation of donor islets and simultaneous blockade of two host costimulatory pathways in prolonging xenoislet transplant survival.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Antígeno B7-1/fisiología , Antígenos CD28/fisiología , Antígenos CD40/fisiología , Ligando de CD40/fisiología , Inmunoconjugados/farmacología , Terapia de Inmunosupresión , Trasplante de Islotes Pancreáticos/inmunología , Trasplante Heterólogo/inmunología , Abatacept , Animales , Citocinas/biosíntesis , Femenino , Supervivencia de Injerto , Masculino , Ratones , Ratones Endogámicos NOD , Porcinos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...