Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
J Tissue Eng ; 14: 20417314221149208, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36699634

RESUMEN

Microbiome is an integral part of the gut and is essential for its proper function. Imbalances of the microbiota can be devastating and have been linked with several gastrointestinal conditions. Current gastrointestinal models do not fully reflect the in vivo situation. Thus, it is important to establish more advanced in vitro models to study host-microbiome/pathogen interactions. Here, we developed for the first time an apical-out human small intestinal organoid model in hypoxia, where the apical surface is directly accessible and exposed to a hypoxic environment. These organoids mimic the intestinal cell composition, structure and functions and provide easy access to the apical surface. Co-cultures with the anaerobic strains Lactobacillus casei and Bifidobacterium longum showed successful colonization and probiotic benefits on the organoids. These novel hypoxia-tolerant apical-out small intestinal organoids will pave the way for unraveling unknown mechanisms related to host-microbiome interactions and serve as a tool to develop microbiome-related probiotics and therapeutics.

2.
Autophagy ; 14(2): 283-295, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29377763

RESUMEN

Expression of EGFRvIII is frequently observed in glioblastoma and is associated with increased cellular proliferation, enhanced tolerance to metabolic stresses, accelerated tumor growth, therapy resistance and poor prognosis. We observed that expression of EGFRvIII elevates the activation of macroautophagy/autophagy during starvation and hypoxia and explored the underlying mechanism and consequence. Autophagy was inhibited (genetically or pharmacologically) and its consequence for tolerance to metabolic stress and its therapeutic potential in (EGFRvIII+) glioblastoma was assessed in cellular systems, (patient derived) tumor xenopgrafts and glioblastoma patients. Autophagy inhibition abrogated the enhanced proliferation and survival advantage of EGFRvIII+ cells during stress conditions, decreased tumor hypoxia and delayed tumor growth in EGFRvIII+ tumors. These effects can be attributed to the supporting role of autophagy in meeting the high metabolic demand of EGFRvIII+ cells. As hypoxic tumor cells greatly contribute to therapy resistance, autophagy inhibition revokes the radioresistant phenotype of EGFRvIII+ tumors in (patient derived) xenograft tumors. In line with these findings, retrospective analysis of glioblastoma patients indicated that chloroquine treatment improves survival of all glioblastoma patients, but patients with EGFRvIII+ glioblastoma benefited most. Our findings disclose the unique autophagy dependency of EGFRvIII+ glioblastoma as a therapeutic opportunity. Chloroquine treatment may therefore be considered as an additional treatment strategy for glioblastoma patients and can reverse the worse prognosis of patients with EGFRvIII+ glioblastoma.


Asunto(s)
Autofagia/fisiología , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Receptores ErbB/biosíntesis , Glioblastoma/metabolismo , Glioblastoma/patología , Animales , Autofagia/efectos de los fármacos , Autofagia/genética , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/genética , Línea Celular Tumoral , Proliferación Celular , Cloroquina/farmacología , Cloroquina/uso terapéutico , Resistencia a Antineoplásicos , Receptores ErbB/genética , Femenino , Glioblastoma/tratamiento farmacológico , Glioblastoma/genética , Humanos , Masculino , Ratones , Ratones Desnudos , Transducción de Señal , Estrés Fisiológico , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Radiother Oncol ; 116(2): 214-20, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26253949

RESUMEN

PURPOSE: This phase I/II study sought to determine the safety and maximum tolerated dose (MTD) of the combination of rapamycin, an mTOR inhibitor, with short-course radiotherapy in rectal cancer patients. Antitumor activity, changes in metabolic activity and perfusion on imaging, and changes in phosphorylation status of the mTOR pathway were also assessed. MATERIALS AND METHODS: Patients with primary resectable rectal cancer were treated with short-course hypofractionated radiotherapy (5×5 Gy) combined with oral rapamycin 1 week before and during radiotherapy, followed by surgical resection. RESULTS: Thirteen patients were entered in phase I. One patient developed a dose-limiting toxicity, consisting of a grade 4 leak and grade 4 bleeding. Because of an unexpected high rate of grade 3 postoperative toxicity, it was decided to treat patients with delayed surgery in phase II. Primary endpoint for phase II was tumor blood flow (K(trans)) assessed by perfusion CT. Thirty-one patients were treated with the MTD of 6 mg rapamycin daily. One patient (3%) developed a pathological complete response (pCR) and 3 patients (10%) had a ypT1N0 tumor at the time of resection. No change in tumor perfusion was observed on perfusion CT, but a significant decrease of metabolic activity was found on PET-scan. CONCLUSIONS: The combination of short-course radiotherapy and rapamycin turned out to be feasible, provided that the interval between neo-adjuvant treatment and surgical resection is at least 6 weeks. Although from this cohort no clear increase in pCR could be observed, a clear metabolic response after rapamycin run-in was observed, indicating a biological activity of this drug in rectal cancer.


Asunto(s)
Antibióticos Antineoplásicos/administración & dosificación , Quimioradioterapia/métodos , Neoplasias del Recto/terapia , Sirolimus/administración & dosificación , Adulto , Anciano , Anciano de 80 o más Años , Antibióticos Antineoplásicos/efectos adversos , Femenino , Humanos , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Cuidados Preoperatorios , Sirolimus/efectos adversos , Resultado del Tratamiento
4.
Radiother Oncol ; 116(3): 417-22, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26164772

RESUMEN

BACKGROUND AND PURPOSE: The epidermal growth factor receptor (EGFR) is overexpressed, amplified or mutated in various human epithelial tumors and hypoxia is a common feature of solid tumors. Both EGFR and hypoxia are associated with therapy resistance and poor treatment outcome. To survive hypoxia, cells adapt by activation of hypoxia responsive pathways and expression of hypoxia-induced plasma membrane proteins. We observed that GABAA receptor associated protein like1 (GABARAPL1) and plasma membrane expression of EGFR were increased during hypoxia. Here we explored the role of the GABARAPL1 in EGFR membrane expression during hypoxia. MATERIAL AND METHODS: Quantitative qPCR, immunoblot analysis, flow cytometry and cytochemistry were used to assess this interplay. RESULTS: GABARAPL1 mRNA and protein levels are increased during hypoxia in vitro and correlate with tumor hypoxia in a panel of primary HNSCC xenografts. High GABARAPL1 mRNA is associated with poor outcome of HNSCC patients. During hypoxia, EGFR membrane expression is increased and GABARAPL1 and EGFR colocalize at the plasma membrane. GABARAPL1 knockdown inhibits EGFR membrane expression during hypoxia. CONCLUSION: GABARAPL1 is required for increased membrane expression of EGFR during hypoxia, suggesting a role for GABARAPL1 in the trafficking of these membrane proteins.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Carcinoma de Células Escamosas/metabolismo , Receptores ErbB/metabolismo , Neoplasias de Cabeza y Cuello/metabolismo , Hipoxia/fisiopatología , Proteínas Asociadas a Microtúbulos/fisiología , Antimetabolitos/farmacología , Hipoxia de la Célula/fisiología , Membrana Celular/metabolismo , Movimiento Celular/fisiología , Doxiciclina/farmacología , Técnicas de Silenciamiento del Gen , Humanos , ARN Mensajero/metabolismo , Células Tumorales Cultivadas
5.
Radiother Oncol ; 114(3): 406-12, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25779723

RESUMEN

BACKGROUND: (Pre)clinical studies indicate that autophagy inhibition increases response to anti-cancer therapies. Although promising, due to contradicting reports, it remains unclear if radiation therapy changes autophagy activity and if autophagy inhibition changes the cellular intrinsic radiosensitivity. Discrepancies may result from different assays and models through off-target effects and influencing other signaling routes. In this study, we directly compared the effects of genetic and pharmacological inhibition of autophagy after irradiation in human cancer cell lines. MATERIALS AND METHODS: Changes in autophagy activity after ionizing radiation (IR) were assessed by flux analysis in eight cell lines. Clonogenic survival, DNA damage (COMET-assay) and H2AX phosphorylation were assessed after chloroquine or 3-methyladenine pretreatment and after ATG7 or LC3b knockdown. RESULTS: IR failed to induce autophagy and chloroquine failed to change intrinsic radiosensitivity of cells. Interestingly, 3-methyladenine and ATG7- or LC3b-deficiency sensitized cancer cells to irradiation. Surprisingly, the radiosensitizing effect of 3-methyladenine was also observed in ATG7 and LC3b deficient cells and was associated with attenuated γ-H2AX formation and DNA damage repair. CONCLUSION: Our data demonstrate that the anti-tumor effects of chloroquine are independent of changes in intrinsic radioresistance. Furthermore, ATG7 and LC3b support radioresistance independent of canonical autophagy that involves lysosomal degradation.


Asunto(s)
Autofagia , Adenina/análogos & derivados , Adenina/farmacología , Autofagia/efectos de los fármacos , Línea Celular Tumoral , Cloroquina/farmacología , Reparación del ADN/efectos de los fármacos , Humanos , Fosforilación , Tolerancia a Radiación/genética , Radiación Ionizante , Fármacos Sensibilizantes a Radiaciones/farmacología , Transducción de Señal/efectos de los fármacos
6.
Cell Cycle ; 13(1): 42-51, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24335351

RESUMEN

The epidermal growth factor receptor (EGFR) is amplified or mutated in various human epithelial tumors. Its expression and activation leads to cell proliferation, differentiation, and survival. Consistently, EGFR amplification or expression of EGFR variant 3 (EGFRvIII) is associated with resistance to conventional cancer therapy through activation of pro-survival signaling and DNA-repair mechanisms. EGFR targeting has successfully been exploited as strategy to increase treatment efficacy. Nevertheless, these targeting strategies have only been proven effective in a limited percentage of human tumors. Recent knowledge indicates that EGFR deregulated tumors display differences in autophagy and dependence on autophagy for growth and survival and the use of autophagy to increase resistance to EGFR-targeting drugs. In this review the dependency on autophagy and its role in mediating resistance to EGFR-targeting agents will be discussed. Considering the current knowledge, autophagy inhibition could provide a novel strategy to enhance therapy efficacy in treatment of EGFR deregulated tumors.


Asunto(s)
Autofagia/genética , Receptores ErbB/metabolismo , Neoplasias/genética , Transducción de Señal , Antineoplásicos/uso terapéutico , Diferenciación Celular/genética , Proliferación Celular , Supervivencia Celular/genética , Resistencia a Antineoplásicos/genética , Receptores ErbB/genética , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/patología
7.
Radiother Oncol ; 108(3): 479-83, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23891088

RESUMEN

BACKGROUND AND PURPOSE: The epidermal growth factor receptor (EGFR) is overexpressed, amplified or mutated in various human epithelial tumors, and is associated with tumor aggressiveness and therapy resistance. Autophagy activation provides a survival advantage for cells in the tumor microenvironment. In the current study, we assessed the potential of autophagy inhibition (using chloroquine (CQ)) in treatment of EGFR expressing tumors. MATERIAL AND METHODS: Quantitative PCR, immunohistochemistry, clonogenic survival, proliferation assays and in vivo tumor growth were used to assess this potential. RESULTS: We show that EGFR overexpressing xenografts are sensitive to CQ treatment and are sensitized to irradiation by autophagy inhibition. In HNSSC xenografts, a correlation between EGFR and expression of the autophagy marker LC3b is observed, suggesting a role for autophagy in EGFR expressing tumors. This observation was substantiated in cell lines, showing high EGFR expressing cells to be more sensitive to CQ addition as reflected by decreased proliferation and survival. Surprisingly high EGFR expressing cells display a lower autophagic flux. CONCLUSIONS: The EGFR high expressing cells and tumors investigated in this study are highly dependent on autophagy for growth and survival. Inhibition of autophagy may therefore provide a novel treatment opportunity for EGFR overexpressing tumors.


Asunto(s)
Autofagia/fisiología , Proliferación Celular , Receptores ErbB/fisiología , Neoplasias/patología , Animales , Línea Celular Tumoral , Supervivencia Celular , Cloroquina/farmacología , Femenino , Humanos , Ratones , Proteínas Asociadas a Microtúbulos/fisiología
8.
Radiother Oncol ; 108(3): 529-34, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23849170

RESUMEN

BACKGROUND AND PURPOSE: Tumor hypoxia is associated with therapy resistance and malignancy. Previously we demonstrated that activation of autophagy and the unfolded protein response (UPR) promote hypoxia tolerance. Here we explored the importance of ULK1 in hypoxia tolerance, autophagy induction and its prognostic value for recurrence after treatment. MATERIAL AND METHODS: Hypoxic regulation of ULK1 mRNA and protein was assessed in vitro and in primary human head and neck squamous cell carcinoma (HNSCC) xenografts. Its importance in autophagy induction, mitochondrial homeostasis and tolerance to chronic and acute hypoxia was evaluated in ULK1 knockdown cells. The prognostic value of ULK1 mRNA expression was assessed in 82 HNSCC patients. RESULTS: ULK1 enrichment was observed in hypoxic tumor regions. High enrichment was associated with a high hypoxic fraction. In line with these findings, high ULK1 expression in HNSCC patients appeared associated with poor local control. Exposure of cells to hypoxia induced ULK1 mRNA in a UPR and HIF1α dependent manner. ULK1 knockdown decreased autophagy activation, increased mitochondrial mass and ROS exposure and sensitized cells to acute and chronic hypoxia. CONCLUSIONS: We demonstrate that ULK1 is a hypoxia regulated gene and is associated with hypoxia tolerance and a worse clinical outcome.


Asunto(s)
Autofagia , Carcinoma de Células Escamosas/metabolismo , Neoplasias de Cabeza y Cuello/metabolismo , Péptidos y Proteínas de Señalización Intracelular/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Homólogo de la Proteína 1 Relacionada con la Autofagia , Carcinoma de Células Escamosas/patología , Hipoxia de la Célula , Línea Celular Tumoral , Supervivencia Celular , Neoplasias de Cabeza y Cuello/patología , Humanos , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Carcinoma de Células Escamosas de Cabeza y Cuello , Respuesta de Proteína Desplegada
9.
Radiother Oncol ; 99(3): 379-84, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21719133

RESUMEN

BACKGROUND AND PURPOSE: Hypoxia is a common feature of solid tumors that is associated with an aggressive phenotype, resistance to therapy and poor prognosis. Major contributors to these adverse effects are the transcriptional program activated by the HIF family of transcription factors as well as the translational response mediated by PERK-dependent phosphorylation of eIF2α and inhibition of mTORC1 activity. In this study we determined the relative contribution of both transcriptional and translational responses to changes in hypoxia induced gene expression. MATERIAL AND METHODS: Total and efficiently translated (polysomal) mRNA was isolated from DU145 prostate carcinoma cells that were exposed for up to 24 h of hypoxia (<0.02% O(2)). Changes in transcription and translation were assessed using affymetrix microarray technology. RESULTS: Our data reveal an unexpectedly large contribution of translation control on both induced and repressed gene expression at all hypoxic time points, particularly during acute hypoxia (2-4 h). Gene ontology analysis revealed that gene classes like transcription and signal transduction are stimulated by translational control whereas expression of genes involved in cell growth and protein metabolism are repressed during hypoxic conditions by translational control. CONCLUSIONS: Our data indicate that translation influences gene expression during hypoxia on a scale comparable to that of transcription.


Asunto(s)
Hipoxia de la Célula/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias de la Próstata/genética , Línea Celular Tumoral , Perfilación de la Expresión Génica , Humanos , Masculino , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo , Biosíntesis de Proteínas , ARN Mensajero/metabolismo , Transducción de Señal , Células Tumorales Cultivadas
10.
Radiother Oncol ; 99(3): 392-397, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21680037

RESUMEN

BACKGROUND AND PURPOSE: Hypoxia is a hallmark of solid cancers and associated with metastases and treatment failure. During tumor progression epithelial cells often acquire mesenchymal features, a phenomenon known as epithelial-to-mesenchymal transition (EMT). Intratumoral hypoxia has been linked to EMT induction. We hypothesized that signals from the tumor microenvironment such as growth factors and tumor oxygenation collaborate to promote EMT and thereby contribute to radioresistance. MATERIALS AND METHODS: Gene expression changes under hypoxia were analyzed using microarray and validated by qRT-PCR. Conversion of epithelial phenotype upon hypoxic exposure, TGFß addition or oncogene activation was investigated by Western blot and immunofluorescence. Cell survival following ionizing radiation was assayed using clonogenic survival. RESULTS: Upon hypoxia, TGFß addition or EGFRvIII expression, MCF7, A549 and NMuMG epithelial cells acquired a spindle shape and lost cell-cell contacts. Expression of epithelial markers such as E-cadherin decreased, whereas mesenchymal markers such as vimentin and N-cadherin increased. Combining hypoxia with TGFß or EGFRvIII expression, lead to more rapid and pronounced EMT-like phenotype. Interestingly, E-cadherin expression and the mesenchymal appearance were reversible upon reoxygenation. Mesenchymal conversion and E-cadherin loss were associated with radioresistance. CONCLUSIONS: Our findings describe a mechanism by which the tumor microenvironment may contribute to tumor radioresistance via E-cadherin loss and EMT.


Asunto(s)
Cadherinas/metabolismo , Neoplasias/metabolismo , Tolerancia a Radiación , Análisis de Varianza , Western Blotting , Hipoxia de la Célula , Línea Celular Tumoral , Supervivencia Celular/efectos de la radiación , Transformación Celular Neoplásica , Células Epiteliales/metabolismo , Células Epiteliales/patología , Regulación Neoplásica de la Expresión Génica , Humanos , Técnicas para Inmunoenzimas , Mesodermo/metabolismo , Mesodermo/patología , Neoplasias/genética , Neoplasias/patología , Fenotipo , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal , Microambiente Tumoral
11.
Radiother Oncol ; 92(3): 399-404, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19616331

RESUMEN

BACKGROUND AND PURPOSE: The epidermal growth factor receptor (EGFR) is overexpressed or mutated in many tumour types. The truncated, constitutively active EGFRvIII variant has not been detected in normal tissues but is found in many malignancies. In the current study, we have investigated the hypothesis that EGFRvIII contributes to a growth and survival advantage under tumour microenvironment-related stress conditions. MATERIALS AND METHODS: U373MG doxycycline-regulated isogenic cells expressing EGFRwt or EGFRvIII were created and validated using Western blot, FACS and qRT-PCR. In vitro proliferation was evaluated with standard growth assays. Cell survival was assayed using clonogenic survival. Animal experiments were performed using NMRI-nu-xenografted mice. RESULTS: Inducible isogenic cell lines were created and showed high induction of EGFRwt and EGFRvIII upon doxycycline addition. Overexpression of EGFRvIII but not of EGFRwt in this model resulted in a growth and survival advantage upon different tumour microenvironment-related stress conditions in vitro. Induction of EGFRvIII increased tumour growth in vivo, which was reversible upon loss of expression. CONCLUSIONS: Under conditions where nutrients are limited and stress is apparent, as in the tumour microenvironment, expression of EGFRvIII leads to a growth and survival advantage. These data indicate a potential selection of EGFRvIII-expressing tumour cells under such stress conditions.


Asunto(s)
Doxiciclina/farmacología , Ambiente , Receptores ErbB/genética , Eliminación de Gen , Estrés Fisiológico , Animales , Western Blotting , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Receptores ErbB/biosíntesis , Receptores ErbB/efectos de los fármacos , Citometría de Flujo , Regulación Neoplásica de la Expresión Génica , Glioblastoma/tratamiento farmacológico , Glioblastoma/genética , Humanos , Modelos Lineales , Ratones , Ratones Desnudos , Mutación , Probabilidad , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Estadísticas no Paramétricas , Trasplante Heterólogo , Células Tumorales Cultivadas/efectos de los fármacos
12.
Radiother Oncol ; 92(3): 393-8, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19616334

RESUMEN

BACKGROUND AND PURPOSE: Despite the clinical use of cetuximab, a chimeric antibody against EGFR, little is known regarding its interaction with EGFRvIII, a frequently expressed deletion mutant of EGFR. Therefore, we investigated the interaction and the functional consequences of cetuximab treatment on glioma cells stably expressing EGFRvIII. MATERIALS AND METHODS: The human glioma cell line U373 genetically modified to express EGFRvIII was used to measure the binding of cetuximab and its internalization using flow cytometry and confocal microscopy. Proliferation and cell survival were analyzed by cell growth and clonogenic survival assays. RESULTS: Cetuximab is able to bind to EGFRvIII and causes an internalization of the receptor and decreases its expression levels. Furthermore, in contrast to EGF, cetuximab was able to activate EGFRvIII which was evidenced by multiple phosphorylation sites and its downstream signaling targets. Despite this activation, the growth rate and the radiosensitivity of the EGFRvIII-expressing glioma cells were not modulated. CONCLUSIONS: Cetuximab binds to EGFRvIII and leads to the initial activation, internalization and subsequent downregulation of EGFRvIII, but it does not seem to modulate the proliferation or radiosensitivity of EGFRvIII-expressing glioma cells. Thus, approaches to treat EGFRvIII-expressing glioma cells should be evaluated more carefully.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Receptores ErbB/efectos de los fármacos , Receptores ErbB/genética , Eliminación de Gen , Anticuerpos Monoclonales Humanizados , Sitios de Unión/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cetuximab , Citometría de Flujo , Regulación Neoplásica de la Expresión Génica , Glioma/tratamiento farmacológico , Glioma/genética , Humanos , Microscopía Confocal , Probabilidad , Transducción de Señal/efectos de los fármacos , Estadísticas no Paramétricas , Células Tumorales Cultivadas/efectos de los fármacos
13.
J Biol Chem ; 284(36): 24204-12, 2009 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-19564335

RESUMEN

Adaptation to tumor hypoxia is mediated in large part by changes in protein expression. These are driven by multiple pathways, including activation of the hypoxia inducible factor-1 (HIF-1) transcription factor and the PKR-like endoplasmic reticulum kinase PERK, a component of the unfolded protein response. Through gene expression profiling we discovered that induction of the HIF-1 target gene CA9 was defective in mouse embryo fibroblasts derived from mice harboring an eIF2alpha S51A knock-in mutation. This finding was confirmed in two isogenic human cell lines with an engineered defect in eIF2alpha phosphorylation. We show that impaired CA9 expression was not due to changes in HIF activity or CA9 mRNA stability. Using chromatin immunoprecipitation we show that the eIF2alpha-dependent translationally regulated gene ATF4 binds directly to the CA9 promoter and is associated with loss of the transcriptional repressive histone 3 lysine 27 tri-methylation mark. Loss or overexpression of ATF4 confirmed its role in CA9 induction during hypoxia. Our data indicate that expression of CA9 is regulated through both the HIF-1 and unfolded protein response hypoxia response pathways in vitro and in vivo.


Asunto(s)
Antígenos de Neoplasias/biosíntesis , Anhidrasas Carbónicas/biosíntesis , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Proteínas de Neoplasias/metabolismo , Neoplasias/enzimología , Pliegue de Proteína , Factor de Transcripción Activador 4/metabolismo , Animales , Anhidrasa Carbónica IX , Hipoxia de la Célula , Línea Celular Tumoral , Factor 2 Eucariótico de Iniciación/metabolismo , Femenino , Histonas/metabolismo , Humanos , Factor 1 Inducible por Hipoxia/metabolismo , Ratones , Ratones Desnudos , Fosforilación , Regiones Promotoras Genéticas , eIF-2 Quinasa/metabolismo
14.
Radiother Oncol ; 83(3): 333-9, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17512071

RESUMEN

BACKGROUND AND PURPOSE: EGFRvIII has been described to function as an oncoprotein with constitutive activation promoting neoplastic transformation and tumorigenicity. The present study was undertaken to test whether EGFRvIII also contributes to hypoxia tolerance. MATERIAL AND METHODS: The human glioma cell line U373 was genetically modified to stably express EGFRvIII. Western blotting and immunohistochemistry verified the expression of EGFRvIII. Tumour xenografts were produced by injecting U373 control and EGFRvIII positive cells subcutaneously into the lateral flank of recipient mice. Colony formation assays were performed after ionizing radiation at 4Gy and after exposure to anoxia for 1-4 days. RESULTS: EGFRvIII accelerated tumour growth leading to a 3.5-fold increase in tumour size compared to control tumours at 40 days after cell injection. EGFRvIII promoted clonogenic survival by almost 2-fold and 4-fold after 4Gy and 4 days of anoxia, respectively. EGFRvIII was also associated with a substantially bigger colony size after anoxic treatment. CONCLUSIONS: EGFRvIII expression stimulates the growth of tumour xenografts and strongly promotes survival after irradiation and under hypoxic stress.


Asunto(s)
Hipoxia de la Célula , Receptores ErbB/metabolismo , Neoplasias/metabolismo , Tolerancia a Radiación , Animales , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos , Receptores ErbB/genética , Femenino , Variación Genética , Humanos , Ratones , Ratones Desnudos , Neoplasias/radioterapia , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Radiother Oncol ; 83(3): 326-32, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17531336

RESUMEN

BACKGROUND AND PURPOSE: The epidermal growth factor receptor (EGFR) is overexpressed in a significant percentage of human malignancies and its expression is associated with tumour aggressiveness and treatment resistance. The monoclonal antibody cetuximab (IMC-C225) blocks the ligand-binding domain of EGFR with high affinity, preventing downstream signalling resulting in tumour growth inhibition. We developed and characterized a novel imaging probe using Oregon Green 488 labelled cetuximab to evaluate its usage as an imaging agent to target EGFR. MATERIALS AND METHODS: Cells with varying expression levels of EGFR or a mutant form of EGFR, called EGFRvIII, were used for in vitro validation. The in vivo binding of labelled cetuximab to EGFR was also assessed ex vivo on tumour material. RESULTS: The development of Oregon Green 488 labelled cetuximab was successful, demonstrating binding to both EGFR and EGFRvIII in vitro. Accumulation was also found in vivo, which was confirmed by histopathology using anti-EGFR antibodies. However, significant mismatch highlights differences between drug delivery in vivo, and cell expression levels of EGFR. CONCLUSIONS: The monoclonal antibody cetuximab represents a promising probe to evaluate the biologic and pharmacokinetic effects of in vivo cetuximab binding to EGFR. It not only visualizes the presence of the wild type EGFR, but also the presence of the mutant EGFRvIII.


Asunto(s)
Anticuerpos Monoclonales/farmacocinética , Antineoplásicos/farmacocinética , Sistemas de Liberación de Medicamentos , Receptores ErbB/antagonistas & inhibidores , Colorantes Fluorescentes , Neoplasias/diagnóstico , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales Humanizados , Antineoplásicos/química , Ácidos Carboxílicos/química , Línea Celular Tumoral , Cetuximab , Receptores ErbB/análisis , Receptores ErbB/metabolismo , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Colorantes Fluorescentes/química , Colorantes Fluorescentes/farmacocinética , Humanos , Ratones , Ratones Desnudos , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Radiother Oncol ; 83(3): 340-5, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17531340

RESUMEN

BACKGROUND AND PURPOSE: Human tumors are characterized by large variations in oxygen concentration and hypoxic tumors are associated with poor prognosis. In addition, tumors are subjected to periodic changes in oxygenation characterized by hypoxia followed by reoxygenation. Cellular adaptation to hypoxia is well documented, nevertheless little is known about adaptive mechanisms to reoxygenation. Here, we investigate the changes in protein expression during reoxygenation using proteomics. MATERIALS AND METHODS: HeLa cervix carcinoma cells were exposed to 4h of hypoxia (<0.01% O(2)) followed by 1h of reoxygenation. The cellular proteome was examined using 2D gel electrophoresis coupled with mass spectrometry. Validation and investigation of the underlying basis for induced protein expression was investigated using Western blot analysis and quantitative RT-PCR. RESULTS: We identified proteins involved in several cellular processes that are responsible for regulating RNA metabolism, protein synthesis and degradation, including ribosomal protein P0, VCP/p97 and FUSE binding protein 2. CONCLUSIONS: Our results suggest that these newly identified proteins function in pathways that may assist in the recovery of ER stress and protein synthesis during reoxygenation. These proteins may thus be important determinants of the behaviour and survival of tumor cells to transient hypoxic exposures.


Asunto(s)
Hipoxia de la Célula , Retículo Endoplásmico/metabolismo , Regulación Neoplásica de la Expresión Génica , Estrés Oxidativo/genética , Oxígeno/metabolismo , Proteómica , Western Blotting , Retículo Endoplásmico/genética , Femenino , Células HeLa , Humanos , Oxidación-Reducción , Reacción en Cadena de la Polimerasa
17.
Blood ; 102(8): 2803-10, 2003 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-12842993

RESUMEN

Recent suppressive subtractive hybridization analysis on human atherosclerotic plaque-derived RNA revealed genes upregulated in plaques with a thrombus versus stable plaques. Clone SSH6, containing part of a putative open reading frame of an unknown protein, was further investigated. Full-length cDNA, coding for a 473-amino acid (aa) protein, was identified in a vascular smooth muscle cell (SMC) cDNA library. Bioinformatics suggested the presence of multiple SSH6 variants due to alternative splicing of exon 3. Multiple-tissue Northern blot analysis demonstrated a differential expression pattern of these variants, as a ubiquitously expressed SSH6 mRNA missing exon 3, was detected apart from a putative vascular SMC-specific form containing exon 3. Western blot analysis indicated a ubiquitous 35-kDa protein (SSH6-beta), in addition to a 45-kDa protein (vasculin), detected in the vascular wall and in plasma. Analysis of arteries displaying various stages of atherosclerosis indicated that the vasculin/SSH6-beta ratio increases throughout atherogenesis. Immunohistochemical analysis demonstrated cytoplasmic expression of SSH6 gene products in macrophages, endothelial cells, and SMCs. In summary, we identified a novel mRNA/protein, vasculin, in the arterial wall and plasma. The regulated expression of vasculin in plaques suggests a role in atherogenesis. Moreover, its presence in plasma opens perspectives for vasculin as a marker for atherosclerosis.


Asunto(s)
Arteriosclerosis/metabolismo , Proteínas Sanguíneas/biosíntesis , Proteínas Sanguíneas/fisiología , Empalme Alternativo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Northern Blotting , Western Blotting , Línea Celular , Células Cultivadas , ADN Complementario/metabolismo , Proteínas de Unión al ADN , Electroforesis en Gel Bidimensional , Exones , Biblioteca de Genes , Glutatión Transferasa/metabolismo , Humanos , Fragmentos de Inmunoglobulinas/metabolismo , Inmunohistoquímica , Intrones , Datos de Secuencia Molecular , Biblioteca de Péptidos , Plásmidos/metabolismo , Estructura Terciaria de Proteína , ARN Mensajero/metabolismo , Proteínas Recombinantes/metabolismo , Schistosoma japonicum/metabolismo , Distribución Tisular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA