Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Death Discov ; 10(1): 103, 2024 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-38418476

RESUMEN

Glioblastoma multiforme (GBM) is the most fatal form of brain cancer in humans, with a dismal prognosis and a median overall survival rate of less than 15 months upon diagnosis. Glioma stem cells (GSCs), have recently been identified as key contributors in both tumor initiation and therapeutic resistance in GBM. Both public dataset analysis and direct differentiation experiments on GSCs have demonstrated that CREB5 is more highly expressed in undifferentiated GSCs than in differentiated GSCs. Additionally, gene silencing by short hairpin RNA (shRNA) of CREB5 has prevented the proliferation and self-renewal ability of GSCs in vitro and decreased their tumor forming ability in vivo. Meanwhile, RNA-sequencing, luciferase reporter assay, and ChIP assay have all demonstrated the closely association between CREB5 and OLIG2. These findings suggest that targeting CREB5 could be an effective approach to overcoming GSCs.

2.
JCI Insight ; 8(21)2023 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-37788099

RESUMEN

Glioblastoma (GBM) is the most lethal brain cancer with a dismal prognosis. Stem-like GBM cells (GSCs) are a major driver of GBM propagation and recurrence; thus, understanding the molecular mechanisms that promote GSCs may lead to effective therapeutic approaches. Through in vitro clonogenic growth-based assays, we determined mitogenic activities of the ligand molecules that are implicated in neural development. We have identified that semaphorin 3A (Sema3A), originally known as an axon guidance molecule in the CNS, promotes clonogenic growth of GBM cells but not normal neural progenitor cells (NPCs). Mechanistically, Sema3A binds to its receptor neuropilin-1 (NRP1) and facilitates an interaction between NRP1 and TGF-ß receptor 1 (TGF-ßR1), which in turn leads to activation of canonical TGF-ß signaling in both GSCs and NPCs. TGF-ß signaling enhances self-renewal and survival of GBM tumors through induction of key stem cell factors, but it evokes cytostatic responses in NPCs. Blockage of the Sema3A/NRP1 axis via shRNA-mediated knockdown of Sema3A or NRP1 impeded clonogenic growth and TGF-ß pathway activity in GSCs and inhibited tumor growth in vivo. Taken together, these findings suggest that the Sema3A/NRP1/TGF-ßR1 signaling axis is a critical regulator of GSC propagation and a potential therapeutic target for GBM.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Humanos , Semaforina-3A/metabolismo , Semaforina-3A/farmacología , Glioblastoma/patología , Neuropilina-1/genética , Neoplasias Encefálicas/patología , Factor de Crecimiento Transformador beta
3.
Cancer Cell ; 41(8): 1480-1497.e9, 2023 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-37451272

RESUMEN

Radiation therapy (RT) provides therapeutic benefits for patients with glioblastoma (GBM), but inevitably induces poorly understood global changes in GBM and its microenvironment (TME) that promote radio-resistance and recurrence. Through a cell surface marker screen, we identified that CD142 (tissue factor or F3) is robustly induced in the senescence-associated ß-galactosidase (SA-ßGal)-positive GBM cells after irradiation. F3 promotes clonal expansion of irradiated SA-ßGal+ GBM cells and orchestrates oncogenic TME remodeling by activating both tumor-autonomous signaling and extrinsic coagulation pathways. Intratumoral F3 signaling induces a mesenchymal-like cell state transition and elevated chemokine secretion. Simultaneously, F3-mediated focal hypercoagulation states lead to activation of tumor-associated macrophages (TAMs) and extracellular matrix (ECM) remodeling. A newly developed F3-targeting agent potently inhibits the aforementioned oncogenic events and impedes tumor relapse in vivo. These findings support F3 as a critical regulator for therapeutic resistance and oncogenic senescence in GBM, opening potential therapeutic avenues.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Humanos , Glioblastoma/tratamiento farmacológico , Glioblastoma/genética , Glioblastoma/radioterapia , Tromboplastina , Línea Celular Tumoral , Recurrencia Local de Neoplasia , Transducción de Señal , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/radioterapia , Neoplasias Encefálicas/metabolismo , Microambiente Tumoral
4.
Neoplasia ; 39: 100894, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36972629

RESUMEN

Recent studies indicate that signaling molecules traditionally associated with central nervous system function play critical roles in cancer. Dopamine receptor signaling is implicated in various cancers including glioblastoma (GBM) and it is a recognized therapeutic target, as evidenced by recent clinical trials with a selective dopamine receptor D2 (DRD2) inhibitor ONC201. Understanding the molecular mechanism(s) of the dopamine receptor signaling will be critical for development of potent therapeutic options. Using the human GBM patient-derived tumors treated with dopamine receptor agonists and antagonists, we identified the proteins that interact with DRD2. DRD2 signaling promotes glioblastoma (GBM) stem-like cells and GBM growth by activating MET. In contrast, pharmacological inhibition of DRD2 induces DRD2-TRAIL receptor interaction and subsequent cell death. Thus, our findings demonstrate a molecular circuitry of oncogenic DRD2 signaling in which MET and TRAIL receptors, critical factors for tumor cell survival and cell death, respectively, govern GBM survival and death. Finally, tumor-derived dopamine and expression of dopamine biosynthesis enzymes in a subset of GBM may guide patient stratification for DRD2 targeting therapy.


Asunto(s)
Glioblastoma , Humanos , Línea Celular Tumoral , Dopamina , Glioblastoma/patología , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF , Transducción de Señal , Receptores de Dopamina D2/metabolismo
5.
Sci Rep ; 12(1): 19842, 2022 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-36400883

RESUMEN

COVID-19 is a highly contagious respiratory infection caused by the SARS-CoV-2 virus. The infected lung epithelial cells secrete a group of chemokines and cytokines, which triggers harmful cytokine storms and hyper-thrombotic responses. Recent studies have proposed that viral-induced senescence is responsible for cytokine release and inflammation in COVID-19 patients. However, it is unknown whether cellular senescence is commonly triggered after viral infection and how inflammation and thrombosis, hyper-activated in these patients, are functionally connected. To address these questions, we conducted a bioinformatics-based meta-analysis using single-cell and bulk RNA sequencing datasets obtained from human patient studies, animal models, and cell lines infected with SARS-CoV-2 and other respiratory viruses. We found that the senescence phenotype is robustly upregulated in most SARS-CoV-2-infected patients, especially in the infected lung epithelial cells. Notably, the upregulation of Tissue factor (F3), a key initiator of the extrinsic blood coagulation pathway, occurs concurrently with the upregulation of the senescence-associated secretory phenotype (SASP) factors. Furthermore, F3 levels are positively correlated with the senescence and hyper-coagulation gene signatures in COVID-19 patients. Together, these data demonstrate the prevalence of senescence in respiratory viral infection and suggest F3 as a critical link between inflammation, thrombosis, and senescence in these disease states.


Asunto(s)
COVID-19 , Trombosis , Humanos , Animales , Tromboplastina/genética , SARS-CoV-2 , Inflamación , Citocinas/metabolismo
6.
Transl Oncol ; 15(1): 101251, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34700192

RESUMEN

BACKGROUND: Glioblastoma (GBM) is the most common primary brain tumor with a dismal prognosis. The inherent cellular diversity and interactions within tumor microenvironments represent significant challenges to effective treatment. Traditional culture methods such as adherent or sphere cultures may mask such complexities whereas three-dimensional (3D) organoid culture systems derived from patient cancer stem cells (CSCs) can preserve cellular complexity and microenvironments. The objective of this study was to determine if GBM organoids may offer a platform, complimentary to traditional sphere culture methods, to recapitulate patterns of clinical drug resistance arising from 3D growth. METHODS: Adult and pediatric surgical specimens were collected and established as organoids. We created organoid microarrays and visualized bulk and spatial differences in cell proliferation using immunohistochemistry (IHC) staining, and cell cycle analysis by flow cytometry paired with 3D regional labeling. We tested the response of CSCs grown in each culture method to temozolomide, ibrutinib, lomustine, ruxolitinib, and radiotherapy. RESULTS: GBM organoids showed diverse and spatially distinct proliferative cell niches and include heterogeneous populations of CSCs/non-CSCs (marked by SOX2) and cycling/senescent cells. Organoid cultures display a comparatively blunted response to current standard-of-care therapy (combination temozolomide and radiotherapy) that reflects what is seen in practice. Treatment of organoids with clinically relevant drugs showed general therapeutic resistance with drug- and patient-specific antiproliferative, apoptotic, and senescent effects, differing from those of matched sphere cultures. CONCLUSIONS: Therapeutic resistance in organoids appears to be driven by altered biological mechanisms rather than physical limitations of therapeutic access. GBM organoids may therefore offer a key technological approach to discover and understand resistance mechanisms of human cancer cells.

7.
Psychiatr Psychol Law ; 27(5): 912-923, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33841023

RESUMEN

We compared the predictive validity of Psychopathy Checklist: Youth Version (PCL:YV) scores assigned by a licensed clinician to scores assigned by a graduate student across a sample of 82 juvenile offenders. Although both raters completed in-depth training and practice scoring cases, the graduate student had no prior clinical experience. The raters showed a high level of agreement in their scoring for 11 reliability check cases (intraclass correlation coefficient, ICCA,1 = .90 for PCL:YV Total score), but the scores assigned by the licensed clinician were better predictors of post-release recidivism (area under the curve, AUC = .77) than those assigned by the graduate student (AUC = .45). There was more variability in the scores assigned by the licensed clinician than those assigned by the graduate student, suggesting that more experienced clinicians' willingness to assign both high and low scores may help explain rater differences in predictive validity.

8.
Biochem Biophys Res Commun ; 496(4): 1013-1018, 2018 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-28216164

RESUMEN

Glioma stem-like cells (GSCs) contribute to tumor initiation, progression, and therapeutic resistance, but their cellular origin remains largely unknown. Here, using a stem/progenitor cell-fate tracking reporter system in which eGFP is expressed by promoter of OCT4 that is activated in stem/progenitor cells, we demonstrate that eGFP-negative glioma cells (GCs) became eGFP-positive-GCs in both in vitro cultures and in vivo xenografts. These eGFP-positive-GCs exhibited GSC features and primarily localized to the perivascular region in tumor xenografts, similar to the existence of OCT4-expressing GCs in the perivascular region of human glioblastoma specimens. Angiocrine factors, including nitric oxide (NO), converted eGFP-negative-GCs into eGFP-positive-GCs. Mechanistically, NO signaling conferred GSC features to GCs by increasing OCT4 and NOTCH signaling via ID4. NO signaling blockade and a suicide gene induction prevented tumorigenicity with a decrease in eGFP-positive-GCs in the perivascular region. Taken together, our results reveal the molecular mechanism underlying GSCs generation by cancer cell dedifferentiation.


Asunto(s)
Proteínas Angiogénicas/metabolismo , Desdiferenciación Celular , Glioma/metabolismo , Glioma/patología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Animales , Línea Celular Tumoral , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Humanos , Ratones , Ratones Desnudos , Neovascularización Patológica
9.
Biochem Biophys Res Commun ; 486(2): 564-570, 2017 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-28327358

RESUMEN

Most cancer-related signaling pathways sustain their active or inactive status via genetic mutations or various regulatory mechanisms. Previously, we demonstrated that platelet-derived growth factor (PDGF) activates Notch signaling through nitric oxide (NO)-signaling-driven activation of inhibitor of differentiation 4 (ID4) in glioblastoma (GBM) stem cells (GSCs) and endothelial cells in the vascular niche of GBM, leading to maintenance of GSC traits and GBM progression. Here, we determined that the PDGF-NO-ID4-signaling axis is constantly activated through a positive regulatory circuit. ID4 expression significantly increased PDGF subunit B expression in both in vitro cultures and in vivo tumor xenografts and regulated NO synthase 2 (NOS2) expression and NO production by activating PDGF signaling, as well as that of its receptor (PDGFR). Additionally, ectopic expression of PDGFRα, NOS2, or ID4 activated the PDGF-NO-ID4-signaling circuit and enhanced the self-renewal of GBM cell lines. These results suggested that the positive regulatory circuit associated with PDGF-NO-ID4 signaling plays a pivotal role in regulating the self-renewal and tumor-initiating capacity of GSCs and might provide a promising therapeutic target for GBM.


Asunto(s)
Neoplasias Encefálicas/genética , Regulación Neoplásica de la Expresión Génica , Glioblastoma/genética , Proteínas Inhibidoras de la Diferenciación/genética , Proteínas Proto-Oncogénicas c-sis/genética , Transducción de Señal/genética , Animales , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Genes Reporteros , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Proteínas Inhibidoras de la Diferenciación/metabolismo , Luciferasas/genética , Luciferasas/metabolismo , Ratones Endogámicos BALB C , Ratones Desnudos , Trasplante de Neoplasias , Células Madre Neoplásicas , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Proteínas Proto-Oncogénicas c-sis/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo
10.
Ann Transl Med ; 5(1): 5, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28164090

RESUMEN

In a number of cancers, deregulated MET pathway leads to aberrantly activated proliferative and invasive signaling programs that promote malignant transformation, cell motility and migration, angiogenesis, survival in hypoxia, and invasion. A better understanding of oncogenic MET signaling will help us to discover effective therapeutic approaches and to identify which tumors are likely to respond to MET-targeted cancer therapy. In this review, we will summarize the roles of MET signaling in cancer, with particular focus on epithelial-mesenchymal transition (EMT) and cancer stemness. Then, we will provide update on MET targeting agents and discuss the challenges that should be overcome for the development of an effective therapy.

11.
Cell Rep ; 16(6): 1629-1641, 2016 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-27477274

RESUMEN

Inhibitor of differentiation 1 (ID1) is highly expressed in glioblastoma stem cells (GSCs). However, the regulatory mechanism responsible for its role in GSCs is poorly understood. Here, we report that ID1 activates GSC proliferation, self-renewal, and tumorigenicity by suppressing CULLIN3 ubiquitin ligase. ID1 induces cell proliferation through increase of CYCLIN E, a target molecule of CULLIN3. ID1 overexpression or CULLIN3 knockdown confers GSC features and tumorigenicity to murine Ink4a/Arf-deficient astrocytes. Proteomics analysis revealed that CULLIN3 interacts with GLI2 and DVL2 and induces their degradation via ubiquitination. Consistent with ID1 knockdown or CULLIN3 overexpression in human GSCs, pharmacologically combined control of GLI2 and ß-CATENIN effectively diminishes GSC properties. A ID1-high/CULLIN3-low expression signature correlates with a poor patient prognosis, supporting the clinical relevance of this signaling axis. Taken together, a loss of CULLIN3 represents a common signaling node for controlling the activity of intracellular WNT and SHH signaling pathways mediated by ID1.


Asunto(s)
Proteínas Cullin/metabolismo , Glioblastoma/metabolismo , Proteína 1 Inhibidora de la Diferenciación/metabolismo , Células Madre Neoplásicas/metabolismo , Vía de Señalización Wnt/fisiología , Animales , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Proliferación Celular/fisiología , Proteínas Hedgehog/metabolismo , Humanos , Ratones , beta Catenina/metabolismo
12.
Anim Biotechnol ; 27(2): 126-32, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26913554

RESUMEN

Comparing the coding and regulatory sequences of genes in different species provides information on whether proteins translated from genes have conserved functions or gene expressions are regulated by analogical mechanisms. Herein, we compared the coding and regulatory sequences of glial fibrillary acidic protein (GFAP) from humans, mice, and pigs. The GFAP gene encodes a class III intermediate filament protein expressed specifically in astrocytes of the central nervous system. On comparing the mRNA, regulatory region (promoter), and protein sequences of GFAP gene in silico, we found that GFAP mRNA 3'-untranslated region (3'-UTR), promoter, and amino acid sequences showed higher similarities between humans and pigs than between humans and mice. In addition, the promoter-luciferase reporter gene assay revealed that the pig GFAP promoter functioned in human astrocytes. Notably, the 1.8-kb promoter fragment upstream from transcription initiation site showed strongest transcriptional activity compared to 5.2-kb DNA fragment or other regions of GFAP promoter. We also found that pig GFAP mRNA and promoter activity increased in pig fibroblasts by human IL-1ß treatment. Taken together, these results suggest that the regulatory mechanisms and functions of pig genes might be more similar to those of humans than mice, indicating that pigs, particularly miniature pigs, are a useful model for studying human biological and pathological events.


Asunto(s)
Proteína Ácida Fibrilar de la Glía/genética , Secuencias Reguladoras de Ácidos Nucleicos/genética , Transcripción Genética/genética , Animales , Astrocitos , Humanos , Ratones , ARN Mensajero/química , ARN Mensajero/genética , Porcinos , Porcinos Enanos/genética , Regiones no Traducidas/genética
13.
Cancer Lett ; 353(2): 194-200, 2014 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-25079688

RESUMEN

The invasiveness of glioblastoma is a major cause of poor prognosis and relapse. However, the molecular mechanism controlling glioma cell invasion is poorly understood. Here, we report that receptor activator of nuclear factor kappa-B (NFκB) ligand (RANKL) promotes glioma cell invasion in vivo, but not in vitro. Unlike the invasiveness under in vitro culture conditions, in vivo xenograft studies revealed that LN229 cells expressing high endogenous RANKL generated more invasive tumors than U87MG cells expressing relatively low endogenous RANKL. Consistently, RANKL-overexpressing U87MG resulted in invasive tumors, whereas RANKL-depleted LN229 generated rarely invasive tumors. We found that the number of activated astrocytes was markedly increased in the periphery of RANKL-high invasive tumors. RANKL activated astrocytes through NFκB signaling and these astrocytes in turn secreted various factors which regulate glioma cell invasion. Among them, transforming growth factor ß (TGF-ß) signaling was markedly increased in glioblastoma specimens and xenograft tumors expressing high levels of RANKL. These results indicate that RANKL contributes to glioma invasion by modulating the peripheral microenvironment of the tumor, and that targeting RANKL signaling has important implications for the prevention of highly invasive glioblastoma.


Asunto(s)
Astrocitos/fisiología , Neoplasias Encefálicas/metabolismo , Citocinas/fisiología , Glioblastoma/metabolismo , Ligando RANK/fisiología , Animales , Neoplasias Encefálicas/mortalidad , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Movimiento Celular , Glioblastoma/mortalidad , Glioblastoma/patología , Humanos , Estimación de Kaplan-Meier , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , FN-kappa B/metabolismo , Invasividad Neoplásica , Trasplante de Neoplasias , Transducción de Señal , Microambiente Tumoral
14.
Cancer Res ; 74(16): 4482-92, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-24962027

RESUMEN

Glioma-initiating cells (GIC), which reside within the perivascular microenvironment to maintain self-renewal capacity, are responsible for glioblastoma initiation, progression, and recurrence. However, the molecular mechanisms controlling crosstalk between GICs and endothelial cells are poorly understood. Here, we report that, in both GICs and endothelial cells, platelet-derived growth factor (PDGF)-driven activation of nitric oxide (NO) synthase increases NO-dependent inhibitor of differentiation 4 (ID4) expression, which in turn promotes JAGGED1-NOTCH activity through suppression of miR129 that specifically represses JAGGED1 suppression. This signaling axis promotes tumor progression along with increased GIC self-renewal and growth of tumor vasculature in the xenograft tumors, which is dramatically suppressed by NOTCH inhibitor. ID4 levels correlate positively with NOS2 (NO synthase-2), HES1, and HEY1 and negatively with miR129 in primary GICs. Thus, targeting the PDGF-NOS-ID4-miR129 axis and NOTCH activity in the perivascular microenvironment might serve as an efficacious therapeutic modality for glioblastoma.


Asunto(s)
Neoplasias Encefálicas/patología , Comunicación Celular/fisiología , Células Endoteliales/patología , Glioma/patología , Células Madre Neoplásicas/patología , Animales , Neoplasias Encefálicas/metabolismo , Diferenciación Celular/fisiología , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Progresión de la Enfermedad , Células Endoteliales/metabolismo , Glioma/metabolismo , Humanos , Ratones , Células Madre Neoplásicas/metabolismo , Transducción de Señal
15.
Brain ; 135(Pt 4): 1055-69, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22434214

RESUMEN

Inflammatory microenvironment signalling plays a crucial role in tumour progression (i.e. cancer cell proliferation, survival, angiogenesis and metastasis) in many types of human malignancies. However, the role of inflammation in brain tumour pathology remains poorly understood. Here, we report that interferon regulatory factor 7 is a crucial regulator of brain tumour progression and heterogeneity. Ectopic expression of interferon regulatory factor 7 in glioma cells promotes tumorigenicity, angiogenesis, microglia recruitment and cancer stemness in vivo and in vitro through induction of interleukin 6, C-X-C motif chemokine 1 and C-C motif chemokine 2. In particular, interferon regulatory factor 7-driven interleukin 6 plays a pivotal role in maintaining glioma stem cell properties via Janus kinase/signal transducer and activator of transcription-mediated activation of Jagged-Notch signalling in glioma cells and glioma stem cells derived from glioma patients. Accordingly, the short hairpin RNA-mediated depletion of interferon regulatory factor 7 in glioma stem cells markedly suppressed interleukin 6-Janus kinase/signal transducer and activator of transcription-mediated Jagged-Notch-signalling pathway, leading to decreases in glioma stem cell marker expression, tumoursphere-forming ability, and tumorigenicity. Furthermore, in a mouse model of wound healing, depletion of interferon regulatory factor 7 suppressed tumour progression and decreased cellular heterogeneity. Finally, interferon regulatory factor 7 was overexpressed in patients with high-grade gliomas, suggesting its potential as an independent prognostic marker for glioma progression. Taken together, our findings indicate that interferon regulatory factor 7-mediated inflammatory signalling acts as a major driver of brain tumour progression and cellular heterogeneity via induction of glioma stem cell genesis and angiogenesis.


Asunto(s)
Glioma/patología , Factor 7 Regulador del Interferón/metabolismo , Interleucina-6/metabolismo , Células Madre Neoplásicas/fisiología , Receptor Notch1/metabolismo , Transducción de Señal/fisiología , Antígeno AC133 , Antígenos CD/metabolismo , Astrocitos/metabolismo , Encéfalo/citología , Línea Celular Tumoral , Movimiento Celular/fisiología , Proliferación Celular , Células Cultivadas , Quimiocina CCL2/metabolismo , Quimiocina CXCL1/metabolismo , Inmunoprecipitación de Cromatina , Biología Computacional , Células Endoteliales , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/genética , Glicoproteínas/metabolismo , Humanos , Factor 7 Regulador del Interferón/genética , Neovascularización Patológica/inducido químicamente , Neovascularización Patológica/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Células-Madre Neurales/fisiología , Péptidos/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción Genética/métodos , Ensayo de Tumor de Célula Madre
16.
Cancer Res ; 71(9): 3410-21, 2011 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-21531766

RESUMEN

Glioma stem cells (GSC) possess tumor-initiating potential and are relatively resistant to conventional chemotherapy and irradiation. Thus, they are considered to be major drivers for glioma initiation, progression, and recurrence. However, the precise mechanism governing acquisition of their drug resistance remains to be elucidated. Our previous study has shown that inhibitor of differentiation 4 (ID4) dedifferentiates Ink4a/Arf(-/-) mouse astrocytes and human glioma cells to glioma stem-like cells (induced GSCs or iGSCs). In this article, we report that ID4-driven iGSCs exhibit chemoresistant behavior to anticancer drugs through activation of ATP-binding cassette (ABC) transporters. We found that ID4 enhanced SOX2 protein expression by suppressing microRNA-9* (miR-9*), which can repress SOX2 by targeting its 3'-untranslated region. Consequently, ID4-mediated SOX2 induction enhanced ABCC3 and ABCC6 expression through direct transcriptional regulation, indicating that ID4 regulates the chemoresistance of iGSCs by promoting SOX2-mediated induction of ABC transporters. Furthermore, we found that short hairpin RNA-mediated knockdown of SOX2 in ID4-driven iGSCs resulted in loss of cancer stemness. Moreover, ectopic expression of SOX2 could dedifferentiate Ink4a/Arf(-/-) astrocytes and glioma cells to iGSCs, indicating a crucial role of SOX2 in genesis and maintenance of GSCs. Finally, we found that the significance of the ID4-miR-9*-SOX2-ABCC3/ABCC6 regulatory pathway is recapitulated in GSCs derived from patients with glioma. Together, our results reveal a novel regulatory mechanism by which ID4-driven suppression of miR-9* induces SOX2, which imparts stemness potential and chemoresistance to glioma cells and GSCs.


Asunto(s)
Proteínas Inhibidoras de la Diferenciación/metabolismo , MicroARNs/biosíntesis , Factores de Transcripción SOXB1/metabolismo , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Resistencia a Antineoplásicos , Técnicas de Silenciamiento del Gen , Glioma/tratamiento farmacológico , Glioma/genética , Glioma/metabolismo , Glioma/patología , Humanos , Proteínas Inhibidoras de la Diferenciación/genética , MicroARNs/genética , MicroARNs/metabolismo , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/biosíntesis , Células Madre Neoplásicas , Factores de Transcripción SOXB1/biosíntesis , Factores de Transcripción SOXB1/genética , Transducción Genética , Regulación hacia Arriba
17.
Mol Cells ; 30(5): 403-8, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20814749

RESUMEN

A growing body of evidence indicates that deregulation of stem cell fate determinants is a hallmark of many types of malignancies. The neural stem cell fate determinant TLX plays a pivotal role in neurogenesis in the adult brain by maintaining neural stem cells. Here, we report a tumorigenic role of TLX in brain tumor initiation and progression. Increased TLX expression was observed in a number of glioma cells and glioma stem cells, and correlated with poor survival of patients with gliomas. Ectopic expression of TLX in the U87MG glioma cell line and Ink4a/Arf-deficient mouse astrocytes (Ink4a/Arf(-/-) astrocytes) induced cell proliferation with a concomitant increase in cyclin D expression, and accelerated foci formation in soft agar and tumor formation in in vivo transplantation assays. Furthermore, overexpression of TLX in Ink4a/Arf(-/-) astrocytes inhibited cell migration and invasion and promoted neurosphere formation and Nestin expression, which are hallmark characteristics of glioma stem cells, under stem cell culture conditions. Our results indicate that TLX is involved in glioma stem cell genesis and represents a potential therapeutic target for this type of malignancy.


Asunto(s)
Neoplasias Encefálicas/patología , Transformación Celular Neoplásica/patología , Glioma/patología , Células Madre Neoplásicas/patología , Células-Madre Neurales/patología , Receptores Citoplasmáticos y Nucleares/fisiología , Adulto , Animales , Astrocitos/metabolismo , Astrocitos/patología , Astrocitoma/genética , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Movimiento Celular/fisiología , Transformación Celular Neoplásica/genética , Neoplasias del Sistema Nervioso Central , Ciclina D/genética , Glioma/genética , Glioma/metabolismo , Humanos , Proteínas de Filamentos Intermediarios/genética , Ratones , Ratones Desnudos , Invasividad Neoplásica , Metástasis de la Neoplasia , Células Madre Neoplásicas/metabolismo , Proteínas del Tejido Nervioso/genética , Nestina , Células-Madre Neurales/metabolismo , Neurogénesis , Receptores Nucleares Huérfanos , Pronóstico , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Regulación hacia Arriba
18.
Genes Dev ; 22(15): 2028-33, 2008 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-18676808

RESUMEN

Cellular origins and genetic factors governing the genesis and maintenance of glioblastomas (GBM) are not well understood. Here, we report a pathogenetic role of the developmental regulator Id4 (inhibitor of differentiation 4) in GBM. In primary murine Ink4a/Arf(-/-) astrocytes, and human glioma cells, we provide evidence that enforced Id4 can drive malignant transformation by stimulating increased cyclin E to produce a hyperproliferative profile and by increased Jagged1 expression with Notch1 activation to drive astrocytes into a neural stem-like cell state. Thus, Id4 plays an integral role in the transformation of astrocytes via its combined actions on two-key cell cycle and differentiation regulatory molecules.


Asunto(s)
Astrocitos/fisiología , Transformación Celular Neoplásica , Ciclina E/metabolismo , Proteínas Inhibidoras de la Diferenciación/metabolismo , Receptor Notch1/metabolismo , Transducción de Señal/fisiología , Animales , Astrocitos/citología , Proteínas de Unión al Calcio/metabolismo , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Células Cultivadas , Corteza Cerebral/citología , Glioblastoma/patología , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteína Jagged-1 , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , ARN Mensajero/metabolismo , Receptor Notch1/genética , Proteínas Serrate-Jagged , Transducción de Señal/genética
19.
Clin Cancer Res ; 14(13): 4059-66, 2008 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-18593982

RESUMEN

PURPOSE: The potential of metastasis can be predicted from clinical features like tumor size, histologic grade, and gene expression patterns. We examined the whole-genome transcriptomic profile of a xenograft model of breast cancer to understand the characteristics of brain metastasis. EXPERIMENTAL DESIGN: Variants of the MDA-MB-435 cell were established from experimental brain metastases. The LvBr2 variant was isolated from lesions in a mouse injected in the left ventricle of the heart, and these cells were used for two cycles of injection into the internal carotid artery and selection of brain lesions, resulting in the Br4 variant. To characterize the different metastatic variants, we examined the gene expression profile of MDA-MB-435, LvBr2, and Br4 cells using microarrays. RESULTS: We could identify 2,016 differentially expressed genes in Br4 by using the F test. Various metastasis-related genes and a number of genes related to angiogenesis, migration, tumorigenesis, and cell cycle were differentially expressed by the Br4 cells. Notably, the Notch signaling pathway was activated in Br4, with increased Jag2 mRNA, activated Notch intracellular domain, and Notch intracellular domain/CLS promoter-luciferase activity. Br4 cells were more migratory and invasive than MDA-MB-435 cells in collagen and Matrigel Transwell assays, and the migration and invasion of Br4 cells were significantly inhibited by inactivation of Notch signaling using DAPT, a gamma-secretase inhibitor, and RNA interference-mediated knockdown of Jagged 2 and Notch1. CONCLUSIONS: Taken together, these results suggest that we have isolated variants of a human cancer cell line with enhanced brain metastatic properties, and the activation of Notch signaling might play a crucial role in brain metastasis.


Asunto(s)
Encéfalo/patología , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Receptores Notch/metabolismo , Animales , Encéfalo/metabolismo , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Movimiento Celular , Humanos , Ratones , Modelos Biológicos , Invasividad Neoplásica , Metástasis de la Neoplasia , Trasplante de Neoplasias , Estructura Terciaria de Proteína
20.
Biochem Biophys Res Commun ; 365(3): 496-502, 2008 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-18021740

RESUMEN

Here, we show that H-ras(V12) causes the p53-knockout mouse astrocytes (p53-/- astrocytes) to be transformed into brain cancer stem-like cells. H-ras(V12) triggers the p53-/- astrocytes to express a Nestin and a Cd133, which are expressed in normal and cancer neural stem cells. H-ras(V12) also induces the formation of a single cell-derived neurosphere under neural stem cell culture conditions. Furthermore, H-ras(V12)-overexpressing p53-/- astrocytes (p53-/-ast-H-ras(V12)) possess an in vitro self-renewal capacity, and are aberrantly differentiated into Tuj1-positve neurons both in vitro and in vivo. Amongst a variety of Ras-mediated canonical signaling pathways, we demonstrated that the MEK/ERK signaling pathway is responsible for neurosphere formation in p53-deficient astrocytes, whereas the PI3K/AKT signaling pathway is involved in oncogenic transformation in these cells. These findings suggest that the activation of Ras signaling pathways promotes the generation of brain cancer stem-like cells from p53-deficient mouse astrocytes by changing cell fate and transforming cell properties.


Asunto(s)
Astrocitos/patología , Neoplasias Encefálicas/etiología , Neoplasias Encefálicas/patología , Transformación Celular Neoplásica/patología , Células Madre Neoplásicas/patología , Proteína Oncogénica p21(ras)/metabolismo , Animales , Astrocitos/metabolismo , Biomarcadores/análisis , Neoplasias Encefálicas/metabolismo , Transformación Celular Neoplásica/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteínas de Filamentos Intermediarios/análisis , Proteínas de Filamentos Intermediarios/metabolismo , Quinasas Quinasa Quinasa PAM/metabolismo , Ratones , Ratones Noqueados , Células Madre Neoplásicas/química , Células Madre Neoplásicas/metabolismo , Proteínas del Tejido Nervioso/análisis , Proteínas del Tejido Nervioso/metabolismo , Nestina , Neuronas/química , Neuronas/patología , Proteína Oncogénica p21(ras)/análisis , Proteína Oncogénica p21(ras)/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Tubulina (Proteína)/análisis , Proteína p53 Supresora de Tumor/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...