Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Nat Metab ; 5(9): 1563-1577, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37653041

RESUMEN

In the tumor microenvironment, adipocytes function as an alternate fuel source for cancer cells. However, whether adipocytes influence macromolecular biosynthesis in cancer cells is unknown. Here we systematically characterized the bidirectional interaction between primary human adipocytes and ovarian cancer (OvCa) cells using multi-platform metabolomics, imaging mass spectrometry, isotope tracing and gene expression analysis. We report that, in OvCa cells co-cultured with adipocytes and in metastatic tumors, a part of the glucose from glycolysis is utilized for the biosynthesis of glycerol-3-phosphate (G3P). Normoxic HIF1α protein regulates the altered flow of glucose-derived carbons in cancer cells, resulting in increased glycerophospholipids and triacylglycerol synthesis. The knockdown of HIF1α or G3P acyltransferase 3 (a regulatory enzyme of glycerophospholipid synthesis) reduced metastasis in xenograft models of OvCa. In summary, we show that, in an adipose-rich tumor microenvironment, cancer cells generate G3P as a precursor for critical membrane and signaling components, thereby promoting metastasis. Targeting biosynthetic processes specific to adipose-rich tumor microenvironments might be an effective strategy against metastasis.


Asunto(s)
Glicerol , Neoplasias Ováricas , Humanos , Femenino , Adipocitos , Glucosa , Fosfatos , Microambiente Tumoral
2.
FASEB J ; 37(8): e23078, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37405762

RESUMEN

Preeclampsia (PE) is a serious hypertensive complication of pregnancy and is a leading cause of maternal death and major contributor to maternal and perinatal morbidity, including establishment of long-term complications. The continued prevalence of PE stresses the need for identification of novel treatments which can target prohypertensive factors implicated in the disease pathophysiology, such as soluble fms-like tyrosine kinase 1 (sFlt-1). We set out to identify novel compounds to reduce placental sFlt-1 and determine whether this occurs via hypoxia-inducible factor (HIF)-1α inhibition. We utilized a commercially available library of natural compounds to assess their ability to reduce sFlt-1 release from primary human placental cytotrophoblast cells (CTBs). Human placental explants from normotensive (NT) and preeclamptic (PE) pregnancies were treated with varying concentrations of luteolin. Protein and mRNA expression of sFlt-1 and upstream mediators were evaluated using ELISA, western blot, and real-time PCR. Of the natural compounds examined, luteolin showed the most potent inhibition of sFlt-1 release, with >95% reduction compared to vehicle-treated. Luteolin significantly inhibited sFlt-1 in cultured placental explants compared to vehicle-treated in a dose- and time-dependent manner. Additionally, significant decreases in HIF-1α expression were observed in luteolin-treated explants, suggesting a mechanism for sFlt-1 downregulation. The ability of luteolin to inhibit HIF-1α may be mediated through the Akt pathway, as inhibitors to Akt and its upstream regulator phosphatidylinositol-3 kinase (PI3K) resulted in significant HIF-1α reduction. Luteolin reduces anti-angiogenic sFlt-1 through inhibition of HIF-1α, making it a novel candidate for the treatment of PE.


Asunto(s)
Placenta , Preeclampsia , Embarazo , Humanos , Femenino , Placenta/metabolismo , Luteolina/farmacología , Luteolina/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Trofoblastos/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Preeclampsia/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo
3.
Cancer Res ; 80(8): 1748-1761, 2020 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-32054768

RESUMEN

Adipocytes are critical for ovarian cancer cells to home to the omentum, but the metabolic changes initiated by this interaction are unknown. To this end, we carried out unbiased mass spectrometry-based metabolomic and proteomic profiling of cancer cells cocultured with primary human omental adipocytes. Cancer cells underwent significant proteo-metabolomic alteration(s), typified by changes in the lipidome with corresponding upregulation of lipid metabolism proteins. FABP4, a lipid chaperone protein, was identified as the critical regulator of lipid responses in ovarian cancer cells cocultured with adipocytes. Subsequently, knockdown of FABP4 resulted in increased 5-hydroxymethylcytosine levels in the DNA, downregulation of gene signatures associated with ovarian cancer metastasis, and reduced clonogenic cancer cell survival. In addition, clustered regularly interspaced short palindromic repeats (CRISPR)-mediated knockout of FABP4 in high-grade serous ovarian cancer cells reduced metastatic tumor burden in mice. Consequently, a small-molecule inhibitor of FABP4 (BMS309403) not only significantly reduced tumor burden in a syngeneic orthotopic mouse model but also increased the sensitivity of cancer cells toward carboplatin both in vitro and in vivo. Taken together, these results show that targeting FABP4 in ovarian cancer cells can inhibit their ability to adapt and colonize lipid-rich tumor microenvironments, providing an opportunity for specific metabolic targeting of ovarian cancer metastasis. SIGNIFICANCE: Ovarian cancer metastatic progression can be restricted by targeting a critical regulator of lipid responses, FABP4.


Asunto(s)
Adipocitos/metabolismo , Resistencia a Antineoplásicos , Proteínas de Unión a Ácidos Grasos/metabolismo , Neoplasias Ováricas/metabolismo , 5-Metilcitosina/análogos & derivados , 5-Metilcitosina/análisis , Animales , Antineoplásicos/farmacología , Compuestos de Bifenilo/farmacología , Carboplatino/farmacología , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Técnicas de Cocultivo , Metilación de ADN , Regulación hacia Abajo , Proteínas de Unión a Ácidos Grasos/antagonistas & inhibidores , Proteínas de Unión a Ácidos Grasos/genética , Femenino , Técnicas de Silenciamiento del Gen , Xenoinjertos , Humanos , Metabolismo de los Lípidos , Lipidómica , Espectrometría de Masas , Metabolómica/métodos , Ratones , Ratones Desnudos , Metástasis de la Neoplasia/prevención & control , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Epiplón/citología , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Análisis por Matrices de Proteínas , Proteómica/métodos , Pirazoles/farmacología , Carga Tumoral/efectos de los fármacos , Regulación hacia Arriba
4.
Mol Cancer Ther ; 19(1): 52-62, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31562255

RESUMEN

The tumor microenvironment (TME) is a key determinant of metastatic efficiency. We performed a quantitative high-throughput screen (qHTS) of diverse medicinal chemistry tractable scaffolds (44,420 compounds) and pharmacologically active small molecules (386 compounds) using a layered organotypic, robust assay representing the ovarian cancer metastatic TME. This 3D model contains primary human mesothelial cells, fibroblasts, and extracellular matrix, to which fluorescently labeled ovarian cancer cells are added. Initially, 100 compounds inhibiting ovarian cancer adhesion/invasion to the 3D model in a dose-dependent manner were identified. Of those, eight compounds were confirmed active in five high-grade serous ovarian cancer cell lines and were further validated in secondary in vitro and in vivo biological assays. Two tyrosine kinase inhibitors, PP-121 and milciclib, and a previously unreported compound, NCGC00117362, were selected because they had potency at 1 µmol/L in vitro Specifically, NCGC00117362 and PP-121 inhibited ovarian cancer adhesion, invasion, and proliferation, whereas milciclib inhibited ovarian cancer invasion and proliferation. Using in situ kinase profiling and immunoblotting, we found that milciclib targeted Cdk2 and Cdk6, and PP-121 targeted mTOR. In vivo, all three compounds prevented ovarian cancer adhesion/invasion and metastasis, prolonged survival, and reduced omental tumor growth in an intervention study. To evaluate the clinical potential of NCGC00117362, structure-activity relationship studies were performed. Four close analogues of NCGC00117362 efficiently inhibited cancer aggressiveness in vitro and metastasis in vivo Collectively, these data show that a complex 3D culture of the TME is effective in qHTS. The three compounds identified have promise as therapeutics for prevention and treatment of ovarian cancer metastasis.


Asunto(s)
Ensayos Analíticos de Alto Rendimiento/métodos , Metástasis de la Neoplasia/prevención & control , Neoplasias Ováricas/terapia , Microambiente Tumoral/genética , Animales , Femenino , Humanos , Ratones , Ratones Desnudos
5.
Invest Ophthalmol Vis Sci ; 60(6): 2118-2126, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-31099828

RESUMEN

Purpose: Several pieces of epidemiologic evidence have indicated PM2.5 (particulate matter with a diameter of 2.5 µm or less) as a causing factor of allergic conjunctivitis, but without experimental elucidation of mechanism. In the present study, PM2.5 in solution was directly applied to the mouse ocular surface to elucidate whether PM2.5 might cause allergic conjunctivitis, and its underlying mechanisms were analyzed. Methods: ICR mice were challenged for 18 consecutive days with eye drops containing PM2.5 at 3.2, 6.4, and 12.8 mg/mL in 0.9% NaCl saline, along with the controls prepared in parallel without PM2.5 and another control group treated with both PM2.5 at 12.8 mg/mL and artificial tears. On day 19, the whole eyes and meibomian glands were harvested for histopathological analyses and assessment of clinical scoring, tear volume, tear breakup time, and tear ferning. Furthermore, goblet cells by periodic acid Schiff stain and infiltrated eosinophils by Giemsa stain were quantified and compared among study groups. Results: Clinical scoring showed more eyelid edema, tearing, and scratching behaviors, with longer tear breakup time under the influence of increased PM2.5 concentrations. Tear ferning assay showed less tear crystal formation and decreased crystal branches after exposure to PM2.5. In addition, higher goblet cell density in the upper palpebral conjunctiva and extensive eosinophil infiltration in the entire conjunctiva and in the meibomian glands were induced by PM2.5. Conclusions: These results demonstrate that PM2.5 can induce symptoms similar to clinical allergic conjunctivitis and that the murine acute allergic conjunctivitis model can be induced by direct exposure to PM2.5.


Asunto(s)
Conjuntivitis Alérgica/etiología , Material Particulado/efectos adversos , Animales , Modelos Animales de Enfermedad , Edema/patología , Exposición a Riesgos Ambientales/efectos adversos , Párpados/patología , Masculino , Ratones , Ratones Endogámicos ICR , Tamaño de la Partícula , Lágrimas/metabolismo
6.
Cell Rep ; 26(11): 3061-3075.e6, 2019 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-30865894

RESUMEN

Ovarian cancer is typified by the development of chemotherapy resistance. Chemotherapy resistance is associated with high aldehyde dehydrogenase (ALDH) enzymatic activity, increased cancer "stemness," and expression of the stem cell marker CD133. As such, ALDH activity has been proposed as a therapeutic target. Although it remains controversial which of the 19 ALDH family members drive chemotherapy resistance, ALDH1A family members have been primarily linked with chemotherapy resistant and stemness. We identified two ALDH1A family selective inhibitors (ALDH1Ai). ALDH1Ai preferentially kills CD133+ ovarian cancer stem-like cells (CSCs). ALDH1Ai induce necroptotic CSC death, mediated, in part, by the induction of mitochondrial uncoupling proteins and reduction in oxidative phosphorylation. ALDH1Ai is highly synergistic with chemotherapy, reducing tumor initiation capacity and increasing tumor eradication in vivo. These studies link ALDH1A with necroptosis and confirm the family as a critical therapeutic target to overcome chemotherapy resistance and improve patient outcomes.


Asunto(s)
Familia de Aldehído Deshidrogenasa 1/antagonistas & inhibidores , Antineoplásicos/farmacología , Inhibidores Enzimáticos/farmacología , Necroptosis , Células Madre Neoplásicas/metabolismo , Neoplasias Ováricas/metabolismo , Retinal-Deshidrogenasa/antagonistas & inhibidores , Antígeno AC133/genética , Antígeno AC133/metabolismo , Familia de Aldehído Deshidrogenasa 1/metabolismo , Animales , Línea Celular Tumoral , Resistencia a Antineoplásicos , Femenino , Humanos , Ratones , Células Madre Neoplásicas/efectos de los fármacos , Fosforilación Oxidativa , Retinal-Deshidrogenasa/metabolismo
7.
J Biomed Sci ; 26(1): 9, 2019 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-30646920

RESUMEN

Extracellular vesicles (EVs) are cell-derived membrane-bound vesicles that serve a means of cell-cell communication. Studying EVs at a single-particle level is important because EVs are inherently heterogeneous. Novel micro- and nanotechnological tools have open opportunities for realizing single-EV measurements exploiting their biochemical, electrical, mechanical, and/or optical properties. This review summarizes the recent development of technologies toward sorting and analyzing single EVs. Sorting EVs into a more homogeneous subset relaxes the sensitivity and throughput required on the EV detection, and hence related techniques are also included in this review. These exciting technologies are on the rise and will expand our understanding of EVs and their applications in the near future.


Asunto(s)
Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/ultraestructura , Animales , Ensayo de Inmunoadsorción Enzimática/métodos , Citometría de Flujo/métodos , Humanos , Microscopía/métodos , Reacción en Cadena de la Polimerasa/métodos , Espectrometría Raman/métodos
8.
Mol Cancer Res ; 17(4): 870-881, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30655321

RESUMEN

The role of phospholipid signaling in ovarian cancer is poorly understood. Sphingosine-1-phosphate (S1P) is a bioactive metabolite of sphingosine that has been associated with tumor progression through enhanced cell proliferation and motility. Similarly, sphingosine kinases (SPHK), which catalyze the formation of S1P and thus regulate the sphingolipid rheostat, have been reported to promote tumor growth in a variety of cancers. The findings reported here show that exogenous S1P or overexpression of SPHK1 increased proliferation, migration, invasion, and stem-like phenotypes in ovarian cancer cell lines. Likewise, overexpression of SPHK1 markedly enhanced tumor growth in a xenograft model of ovarian cancer, which was associated with elevation of key markers of proliferation and stemness. The diabetes drug, metformin, has been shown to have anticancer effects. Here, we found that ovarian cancer patients taking metformin had significantly reduced serum S1P levels, a finding that was recapitulated when ovarian cancer cells were treated with metformin and analyzed by lipidomics. These findings suggested that in cancer the sphingolipid rheostat may be a novel metabolic target of metformin. In support of this, metformin blocked hypoxia-induced SPHK1, which was associated with inhibited nuclear translocation and transcriptional activity of hypoxia-inducible factors (HIF1α and HIF2α). Further, ovarian cancer cells with high SPHK1 were found to be highly sensitive to the cytotoxic effects of metformin, whereas ovarian cancer cells with low SPHK1 were resistant. Together, the findings reported here show that hypoxia-induced SPHK1 expression and downstream S1P signaling promote ovarian cancer progression and that tumors with high expression of SPHK1 or S1P levels might have increased sensitivity to the cytotoxic effects of metformin. IMPLICATIONS: Metformin targets sphingolipid metabolism through inhibiting SPHK1, thereby impeding ovarian cancer cell migration, proliferation, and self-renewal.


Asunto(s)
Metformina/farmacología , Neoplasias Ováricas/tratamiento farmacológico , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Movimiento Celular/efectos de los fármacos , Femenino , Humanos , Hipoglucemiantes/farmacología , Subunidad alfa del Factor 1 Inducible por Hipoxia , Lisofosfolípidos/metabolismo , Ratones , Ratones Desnudos , Terapia Molecular Dirigida , Neoplasias Ováricas/enzimología , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Nat Commun ; 6: 6220, 2015 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-25653139

RESUMEN

The tumour microenvironment contributes to cancer metastasis and drug resistance. However, most high throughput screening (HTS) assays for drug discovery use cancer cells grown in monolayers. Here we show that a multilayered culture containing primary human fibroblasts, mesothelial cells and extracellular matrix can be adapted into a reliable 384- and 1,536-multi-well HTS assay that reproduces the human ovarian cancer (OvCa) metastatic microenvironment. We validate the identified inhibitors in secondary in vitro and in vivo biological assays using three OvCa cell lines: HeyA8, SKOV3ip1 and Tyk-nu. The active compounds directly inhibit at least two of the three OvCa functions: adhesion, invasion and growth. In vivo, these compounds prevent OvCa adhesion, invasion and metastasis, and improve survival in mouse models. Collectively, these data indicate that a complex three-dimensional culture of the tumour microenvironment can be adapted for quantitative HTS and may improve the disease relevance of assays used for drug screening.


Asunto(s)
Antineoplásicos/farmacología , Matriz Extracelular/efectos de los fármacos , Ensayos Analíticos de Alto Rendimiento/métodos , Neoplasias Ováricas/tratamiento farmacológico , Microambiente Tumoral/efectos de los fármacos , Animales , Antineoplásicos/química , Benzofenantridinas/química , Benzofenantridinas/farmacología , Biguanidas/química , Biguanidas/farmacología , Cantaridina/química , Cantaridina/farmacología , Adhesión Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Técnicas de Cocultivo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/patología , Escina/química , Escina/farmacología , Matriz Extracelular/metabolismo , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/patología , Ensayos Analíticos de Alto Rendimiento/instrumentación , Humanos , Concentración 50 Inhibidora , Isoquinolinas/química , Isoquinolinas/farmacología , Ratones , Ratones Desnudos , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Cultivo Primario de Células , Proclorperazina/química , Proclorperazina/farmacología , Tomatina/química , Tomatina/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
11.
J Clin Invest ; 124(10): 4614-28, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25202979

RESUMEN

Ovarian cancer (OvCa) metastasizes to organs in the abdominal cavity, such as the omentum, which are covered by a single layer of mesothelial cells. Mesothelial cells are generally thought to be "bystanders" to the metastatic process and simply displaced by OvCa cells to access the submesothelial extracellular matrix. Here, using organotypic 3D cultures, we found that primary human mesothelial cells secrete fibronectin in the presence of OvCa cells. Moreover, we evaluated the tumor stroma of 108 human omental metastases and determined that fibronectin was consistently overexpressed in these patients. Blocking fibronectin production in primary mesothelial cells in vitro or in murine models, either genetically (fibronectin 1 floxed mouse model) or via siRNA, decreased adhesion, invasion, proliferation, and metastasis of OvCa cells. Using a coculture model, we determined that OvCa cells secrete TGF-ß1, which in turn activates a TGF-ß receptor/RAC1/SMAD-dependent signaling pathway in the mesothelial cells that promotes a mesenchymal phenotype and transcriptional upregulation of fibronectin. Additionally, blocking α5 or ß1 integrin function with antibodies reduced metastasis in an orthotopic preclinical model of OvCa metastasis. These findings indicate that cancer-associated mesothelial cells promote colonization during the initial steps of OvCa metastasis and suggest that mesothelial cells actively contribute to metastasis.


Asunto(s)
Células Epiteliales/citología , Fibronectinas/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias Ováricas/patología , Animales , Adhesión Celular , Línea Celular Tumoral , Técnicas de Cocultivo , Matriz Extracelular/metabolismo , Femenino , Humanos , Integrinas/metabolismo , Ratones , Invasividad Neoplásica , Metástasis de la Neoplasia , Neoplasias Ováricas/metabolismo , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta1/metabolismo
12.
J Biol Chem ; 288(47): 33861-33872, 2013 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-24129571

RESUMEN

Salt-inducible kinase 2 (SIK2) is an important regulator of cAMP response element-binding protein-mediated gene expression in various cell types and is the only AMP-activated protein kinase family member known to interact with the p97/valosin-containing protein (VCP) ATPase. Previously, we have demonstrated that SIK2 can regulate autophagy when proteasomal function is compromised. Here we report that physical and functional interactions between SIK2 and p97/VCP underlie the regulation of endoplasmic reticulum (ER)-associated protein degradation (ERAD). SIK2 co-localizes with p97/VCP in the ER membrane and stimulates its ATPase activity through direct phosphorylation. Although the expression of wild-type recombinant SIK2 accelerated the degradation and removal of ERAD substrates, the kinase-deficient variant conversely had no effect. Furthermore, down-regulation of endogenous SIK2 or mutation of the SIK2 target site on p97/VCP led to impaired degradation of ERAD substrates and disruption of ER homeostasis. Collectively, these findings highlight a mechanism by which the interplay between SIK2 and p97/VCP contributes to the regulation of ERAD in mammalian cells.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Proteínas de Ciclo Celular/metabolismo , Degradación Asociada con el Retículo Endoplásmico/fisiología , Retículo Endoplásmico/metabolismo , Membranas Intracelulares/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Adenosina Trifosfatasas/genética , Proteínas de Ciclo Celular/genética , Retículo Endoplásmico/genética , Células HEK293 , Células HeLa , Humanos , Mutación , Fosforilación/fisiología , Proteínas Serina-Treonina Quinasas/genética , Proteína que Contiene Valosina
13.
Tissue Eng Part A ; 16(4): 1169-77, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20021271

RESUMEN

Solid cancer tumors are thought to arise from aberrant stem cell populations, called cancer stem cells (CSCs). Hence, the development of effective cancer therapies may rely on developing methods that specifically target these cells. However, the scarcity of CSCs in vivo represents a major impediment to such research, as there is an insufficient supply for basic biochemical and genetic analyses. It is therefore necessary to develop methods to expand reproducibly CSC tissue in vitro in a controlled environment. To date, we have developed bioreactor protocols for the suspension culture of an aggressive and deadly type of brain cancer called glioblastoma multiforme (GBM). Human GBM-derived cells achieved a maximum cell density of 2.4 x 10(6) cells/mL after 24 days under high shear conditions in batch culture conditions. In comparison, fed-batch cultures achieved 4.5 x 10(6) cells/mL after 32 days. Characterization of bioreactor-expanded cells using both flow cytometry and a differentiation assay indicated that bioreactor-generated human GBM-derived cells have similar characteristics to the initial cell population and achieve >90% CD133 expression. Additionally, genomic characterization indicated that a very small number of key genes were differentially expressed in the bioreactor-expanded GBM-derived cells, thereby conserving the basic nature of the brain cancer tissue in the cell expansion process.


Asunto(s)
Reactores Biológicos , Neoplasias Encefálicas/patología , Glioblastoma/patología , Células Madre Neoplásicas/patología , Ingeniería de Tejidos/métodos , Antígeno AC133 , Antígenos CD/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/inmunología , Agregación Celular , Recuento de Células , Diferenciación Celular , Línea Celular Tumoral , Medios de Cultivo , Expresión Génica , Glioblastoma/genética , Glioblastoma/inmunología , Glicoproteínas/metabolismo , Humanos , Células Madre Neoplásicas/inmunología , Células Madre Neoplásicas/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Péptidos/metabolismo , Técnicas de Cultivo de Tejidos/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA