Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
N Engl J Med ; 364(7): 627-37, 2011 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-21175304

RESUMEN

BACKGROUND: Amplification and activating mutations of the epidermal growth factor receptor (EGFR) oncogene are molecular hallmarks of glioblastomas. We hypothesized that deletion of NFKBIA (encoding nuclear factor of κ-light polypeptide gene enhancer in B-cells inhibitor-α), an inhibitor of the EGFR-signaling pathway, promotes tumorigenesis in glioblastomas that do not have alterations of EGFR. METHODS: We analyzed 790 human glioblastomas for deletions, mutations, or expression of NFKBIA and EGFR. We studied the tumor-suppressor activity of NFKBIA in tumor-cell culture. We compared the molecular results with the outcome of glioblastoma in 570 affected persons. RESULTS: NFKBIA is often deleted but not mutated in glioblastomas; most deletions occur in nonclassical subtypes of the disease. Deletion of NFKBIA and amplification of EGFR show a pattern of mutual exclusivity. Restoration of the expression of NFKBIA attenuated the malignant phenotype and increased the vulnerability to chemotherapy of cells cultured from tumors with NFKBIA deletion; it also reduced the viability of cells with EGFR amplification but not of cells with normal gene dosages of both NFKBIA and EGFR. Deletion and low expression of NFKBIA were associated with unfavorable outcomes. Patients who had tumors with NFKBIA deletion had outcomes that were similar to those in patients with tumors harboring EGFR amplification. These outcomes were poor as compared with the outcomes in patients with tumors that had normal gene dosages of NFKBIA and EGFR. A two-gene model that was based on expression of NFKBIA and O(6)-methylguanine DNA methyltransferase was strongly associated with the clinical course of the disease. CONCLUSIONS: Deletion of NFKBIA has an effect that is similar to the effect of EGFR amplification in the pathogenesis of glioblastoma and is associated with comparatively short survival.


Asunto(s)
Eliminación de Gen , Genes erbB-1 , Glioblastoma/genética , Proteínas I-kappa B/genética , Análisis Mutacional de ADN , Amplificación de Genes , Expresión Génica , Glioblastoma/mortalidad , Humanos , Estimación de Kaplan-Meier , Inhibidor NF-kappaB alfa , Pronóstico , Células Tumorales Cultivadas
2.
Histochem Cell Biol ; 141(3): 289-300, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24186058

RESUMEN

The embryonic muscles of the axial skeleton and limbs take their origin from the dermomyotomes of the somites. During embryonic myogenesis, muscle precursors delaminate from the dermomyotome giving rise to the hypaxial and epaxial myotome. Mutant studies for myogenic regulatory factors have shown that the development of the hypaxial myotome differs from the formation of the epaxial myotome and that the development of the hypaxial myotome depends on the latter within the trunk region. The transcriptional networks that regulate the transition of proliferative dermomyotomal cells into the predominantly post-mitotic hypaxial myotome, as well as the eventual patterning of the myotome, are not fully understood. Similar transitions occurring during the development of the neural system have been shown to be controlled by the Atonal family of helix-loop-helix transcription factors. Here, we demonstrate that ATOH8, a member of the Atonal family, is expressed in a subset of embryonic muscle cells in the dermomyotome and myotome. Using the RNAi approach, we show that loss of ATOH8 in the lateral somites at the trunk level results in a blockage of differentiation and thus causes cells to be maintained in a predetermined state. Furthermore, we show that ATOH8 is also expressed in cultured C2C12 mouse myoblasts and becomes dramatically downregulated during their differentiation. We propose that ATOH8 plays a role during the transition of myoblasts from the proliferative phase to the differentiation phase and in the regulation of myogenesis in the hypaxial myotome of the trunk.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Tipificación del Cuerpo/genética , Desarrollo de Músculos/genética , Músculo Esquelético/embriología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Diferenciación Celular/genética , Línea Celular , Linaje de la Célula , Embrión de Pollo , Regulación hacia Abajo , Regulación del Desarrollo de la Expresión Génica , Ratones , Mioblastos/citología , Factor 5 Regulador Miogénico/biosíntesis , Miogenina/biosíntesis , Factor de Transcripción PAX7/biosíntesis , Interferencia de ARN , ARN Interferente Pequeño , Somitos/fisiología
3.
Transl Res ; 253: 80-94, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36223881

RESUMEN

Integration of high-risk human papillomavirus (HPV) into the host genome is a crucial event for the development of cervical cancer, however, the underlying mechanism of HPV integration-driven carcinogenesis remains unknown. Here, we performed long-read RNA sequencing on 12 high-grade squamous intraepithelial lesions (HSIL) and cervical cancer patients, including 3 pairs of cervical cancer and corresponding para-cancerous tissue samples to investigate the full-length landscape of cross-species genome integrations. In addition to massive unannotated isoforms, transcriptional regulatory events, and gene chimerism, more importantly, we found that HPV-human fusion events were prevalent in HPV-associated cervical cancers. Combined with the genome data, we revealed the existence of a universal transcription pattern in these fusion events, whereby structurally similar fusion transcripts were generated by specific splicing in E6 and a canonical splicing donor site in E1 linking to various human splicing acceptors. Highly expressed HPV-human fusion transcripts, eg, HPV16 E6*I-E7-E1SD880-human gene, were the key driver of cervical carcinogenesis, which could trigger overexpression of E6*I and E7, and destroy the transcription of tumor suppressor genes CMAHP, TP63 and P3H2. Finally, evidence from in vitro and in vivo experiments demonstrates that the novel read-through fusion gene mRNA, E1-CMAHP (E1C, formed by the integration of HPV58 E1 with CMAHP), existed in the fusion transcript can promote malignant transformation of cervical epithelial cells via regulating downstream oncogenes to participate in various biological processes. Taken together, we reveal a previously unknown mechanism of HPV integration-driven carcinogenesis and provide a novel target for the diagnosis and treatment of cervical cancer.


Asunto(s)
Infecciones por Papillomavirus , Neoplasias del Cuello Uterino , Femenino , Humanos , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/patología , Infecciones por Papillomavirus/complicaciones , Infecciones por Papillomavirus/genética , Infecciones por Papillomavirus/diagnóstico , Carcinogénesis/genética
4.
Nat Biotechnol ; 2023 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-37679545

RESUMEN

Certified RNA reference materials are indispensable for assessing the reliability of RNA sequencing to detect intrinsically small biological differences in clinical settings, such as molecular subtyping of diseases. As part of the Quartet Project for quality control and data integration of multi-omics profiling, we established four RNA reference materials derived from immortalized B-lymphoblastoid cell lines from four members of a monozygotic twin family. Additionally, we constructed ratio-based transcriptome-wide reference datasets between two samples, providing cross-platform and cross-laboratory 'ground truth'. Investigation of the intrinsically subtle biological differences among the Quartet samples enables sensitive assessment of cross-batch integration of transcriptomic measurements at the ratio level. The Quartet RNA reference materials, combined with the ratio-based reference datasets, can serve as unique resources for assessing and improving the quality of transcriptomic data in clinical and biological settings.

5.
Nat Biotechnol ; 2023 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-37679543

RESUMEN

Characterization and integration of the genome, epigenome, transcriptome, proteome and metabolome of different datasets is difficult owing to a lack of ground truth. Here we develop and characterize suites of publicly available multi-omics reference materials of matched DNA, RNA, protein and metabolites derived from immortalized cell lines from a family quartet of parents and monozygotic twin daughters. These references provide built-in truth defined by relationships among the family members and the information flow from DNA to RNA to protein. We demonstrate how using a ratio-based profiling approach that scales the absolute feature values of a study sample relative to those of a concurrently measured common reference sample produces reproducible and comparable data suitable for integration across batches, labs, platforms and omics types. Our study identifies reference-free 'absolute' feature quantification as the root cause of irreproducibility in multi-omics measurement and data integration and establishes the advantages of ratio-based multi-omics profiling with common reference materials.

6.
Hippocampus ; 22(3): 434-41, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21240919

RESUMEN

Sprouty proteins act as negative feedback inhibitors of fibroblast growth factor (FGF) signaling. FGFs belong to the neurotrophic factors and are involved in axonal growth during development and repair. We investigated the expression of Sprouty isoforms in hippocampal neurons as well as the regulation of Sprouty2 and -4 during development and their role in axon growth. Sprouty2 and -4 were located in the nucleus, the cytoplasm, in dendrites, and axons of hippocampal neurons concentrated in growth cones. During development in vivo and differentiation in vitro, expression of Sprouty2 and -4 was gradually downregulated in hippocampal neurons. Between 5 and 24 days in culture expression of both Sprouty isoforms was reduced by 70%. In vivo expression of Sprouty2 was reduced by 79% and of Sprouty4 by 93% on postnatal day 14 compared to embryonic day 16.5. Downregulation of Sprouty2 and -4 by shRNAs strongly promoted elongative axon growth by cultured hippocampal neurons, which was further increased by FGF-2 treatment. In addition, FGF-2 reduced expression of Sprouty2 by 33% and of Sprouty4 by 44%. Together, our results imply that Sprouty2 and -4 are downregulated in the hippocampus during postnatal brain development and that they can act as regulators of developmental axon growth.


Asunto(s)
Axones/metabolismo , Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores , Conos de Crecimiento/metabolismo , Hipocampo/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Células Cultivadas , Regulación hacia Abajo , Hipocampo/citología , Péptidos y Proteínas de Señalización Intracelular , Proteínas de la Membrana/genética , Ratones , Proteínas del Tejido Nervioso/genética , Neuronas/citología , Proteínas Serina-Treonina Quinasas , Transfección
7.
Mol Cell Neurosci ; 42(4): 328-40, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19683577

RESUMEN

Fibroblast growth factors (FGFs) play a prominent role in axonal growth during development and repair. Treatment with FGF-2 or overexpression of FGF receptors promotes peripheral axon regeneration mainly by activation of extracellular signal-regulated kinase (ERK). The Ras/Raf/ERK pathway is under the control of Sprouty proteins acting as negative feedback inhibitors. We investigated the expression of Sprouty isoforms in adult sensory neurons of dorsal root ganglia (DRG) as well as the effects of Sprouty inhibition on axon growth by small interfering RNAs (siRNAs). Sprouty2 revealed the highest expression level in DRG neurons. Down-regulation of Sprouty2 promoted elongative axon growth by adult sensory neurons accompanied by enhanced FGF-2-induced activation of ERK and Ras, whereas Sprouty2 overexpression inhibited axon growth. Sprouty2 was not regulated in vivo in response to a sciatic nerve lesion. Together, our results imply that Sprouty2 is highly expressed in adult peripheral neurons and its down-regulation strongly promotes elongative axon growth by activation of the Ras/Raf/ERK pathway.


Asunto(s)
Axones/fisiología , Regulación hacia Abajo , Ganglios Espinales/citología , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Isoformas de Proteínas/metabolismo , Células Receptoras Sensoriales , Proteínas Adaptadoras Transductoras de Señales , Animales , Axones/ultraestructura , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Ganglios Espinales/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Proteínas de la Membrana/genética , Ratones , Células 3T3 NIH , Proteínas del Tejido Nervioso/genética , Células PC12 , Isoformas de Proteínas/genética , Proteínas Serina-Treonina Quinasas , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Factores de Crecimiento de Fibroblastos/genética , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Células Receptoras Sensoriales/fisiología , Células Receptoras Sensoriales/ultraestructura , Transducción de Señal/fisiología , Quinasas raf/genética , Quinasas raf/metabolismo , Proteínas ras/genética , Proteínas ras/metabolismo
8.
Cells Tissues Organs ; 190(3): 121-34, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19147985

RESUMEN

Acetylation is one of the key chromatin modifications that control gene transcription during embryonic development and tumorigenesis. The types of genes sensitive to such modifications in vivo are not known to date. We investigated the expression of a number of genes involved in embryonic development after treatment with trichostatin A (TSA), a histone deacetylase (HDAC) inhibitor, in the limbs of chicken embryos. Our results show that TSA affects the expression profiles of some genes that play important roles during limb development. The expression of BMP4, SF/HGF and Twist1 increased, whereas the expression of BMP2, FGF8, Shh, Scleraxis, Myf5 and MyoD was decreased or even inhibited. In contrast, the expression of Pax3, Paraxis, Msx1, CREB, and PCNA was not affected. Our results indicate that the chicken embryo can serve as an effective in vivo model for studying the effect of HDAC inhibitors on gene expression and can be helpful for understanding the role of chromatin remodeling and epigenetic control of gene expression.


Asunto(s)
Extremidades/embriología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Inhibidores de Histona Desacetilasas , Ácidos Hidroxámicos/farmacología , Esbozos de los Miembros/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Proteínas Morfogenéticas Óseas/genética , Proteínas Morfogenéticas Óseas/metabolismo , Embrión de Pollo , Factor 8 de Crecimiento de Fibroblastos/genética , Factor 8 de Crecimiento de Fibroblastos/metabolismo , Factor de Crecimiento de Hepatocito/genética , Factor de Crecimiento de Hepatocito/metabolismo , Esbozos de los Miembros/embriología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
9.
Clin Cancer Res ; 14(14): 4593-602, 2008 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-18628474

RESUMEN

PURPOSE: KML001 (sodium metaarsenite) is an orally bioavailable arsenic compound that has entered phase I/II clinical trials in prostate cancer. In this study, we elucidated the mode of action of KML001 and investigated its effects on telomerase and telomeres. EXPERIMENTAL DESIGN: We compared telomere length to KML001 cytotoxic activity in a panel of human solid tumor cell lines. Duration of exposure and concentrations of KML001 that affect telomerase and telomeres were evaluated in relation to established mechanisms of arsenite action such as reactive oxygen species-related DNA damage induction. Binding of KML001 to telomeres was assessed by matrix-assisted laser desorption/ionization mass spectrometry. RESULTS: We established a significant inverse correlation (r(2) = 0.9) between telomere length and cytotoxicity. KML001 exhibited activity in tumor cells with short telomeres at concentrations that can be achieved in serum of patients. We found that telomerase is not directly inhibited by KML001. Instead, KML001 specifically binds to telomeric sequences at a ratio of one molecule per three TTAGGG repeats leading to translocation of the telomerase catalytic subunit into the cytoplasm. In prostate cancer cells with short telomeres, KML001 caused telomere-associated DNA damage signaling as shown by gamma-H2AX induction and chromatin immunoprecipitation assays as well as a rapid telomere erosion shown by metaphase fluorescence in situ hybridization. These effects were not seen in a lung cancer cell line with long telomeres. Importantly, arsenification of telomeres preceded DNA lesions caused by reactive oxygen species production. CONCLUSIONS: Sodium metaarsenite is a telomere targeting agent and should be explored for the treatment of tumors with short telomeres.


Asunto(s)
Antineoplásicos/farmacología , Arsenitos/farmacología , Daño del ADN/efectos de los fármacos , Neoplasias de la Próstata/tratamiento farmacológico , Compuestos de Sodio/farmacología , Telómero/efectos de los fármacos , Western Blotting , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Línea Celular Tumoral , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Inmunoprecipitación , Hibridación in Situ , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Masculino , Neoplasias de la Próstata/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Telomerasa/efectos de los fármacos
10.
Int J Dev Biol ; 52(1): 87-92, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18033676

RESUMEN

Cell migration plays a fundamental role in a wide variety of biological processes including development, tissue repair and disease. These processes depend on directed cell migration along and through cell layers. Chemokines are small secretory proteins that exert their effects by activating a family of G-protein coupled receptors and have been shown to play numerous fundamental roles in the control of physiological and pathological processes during development and in adult tissues, respectively. Stromal-derived factor-1 (SDF-1/CXCL12), a ligand of the chemokine receptor, CXCR4, is involved in providing cells with directional cues as well as in controlling their proliferation and differentiation. Here we studied the expression pattern of SDF-1 in the developing chick embryo. We could detect a specific expression of SDF-1 in the ectoderm, the sclerotome, the intersomitic spaces and the developing limbs. The expression domains of SDF-1 reflect its role in somitic precursor migration and vessel formation in the limbs.


Asunto(s)
Quimiocina CXCL12/metabolismo , Animales , Diferenciación Celular/fisiología , Movimiento Celular/fisiología , Quimiocina CXCL12/fisiología , Embrión de Pollo , Embrión no Mamífero/embriología , Embrión no Mamífero/metabolismo , Expresión Génica , Hibridación in Situ , Ligandos , Receptores CXCR4/genética
12.
Cancer Res ; 65(4): 1489-96, 2005 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-15735037

RESUMEN

Interference with telomerase and telomere maintenance is emerging as an attractive target for anticancer therapies. Ligand-induced stabilization of G-quadruplex formation by the telomeric DNA single-stranded 3' overhang inhibits telomerase from catalyzing telomeric DNA synthesis and from capping telomeric ends. We report here the effects of a 3,6,9-trisubstituted acridine compound, BRACO-19, on telomerase function in vitro and in vivo. The biological activity of BRACO-19 was evaluated in the human uterus carcinoma cell line UXF1138L, which has very short telomeres (2.7 kb). In vitro, nuclear human telomerase reverse transcriptase (hTERT) expression was drastically decreased after 24 hours, induction of cellular senescence and complete cessation of growth was seen after 15 days, paralleled by telomere shortening of ca. 0.4 kb. In vivo, BRACO-19 was highly active as a single agent against early-stage (68 mm(3)) tumors in a s.c. growing xenograft model established from UXF1138L cells, if given chronically at 2 mg per kg per day i.p. BRACO-19 produced growth inhibition of 96% compared with controls accompanied by partial regressions (P < 0.018). Immunostaining of xenograft tissues showed that this response was paralleled by loss of nuclear hTERT protein expression and an increase in atypical mitoses indicative of telomere dysfunction. Cytoplasmic hTERT expression and its colocalization with ubiquitin was observed suggesting that hTERT is bound to ubiquitin and targeted for enhanced degradation upon BRACO-19 treatment. This is in accord with a model of induced displacement of telomerase from the telomere. The in vitro and in vivo data presented here is consistent with the G-quadruplex binding ligand BRACO-19 producing an anticancer effect by inhibiting the capping and catalytic functions of telomerase.


Asunto(s)
Acridinas/farmacología , ADN/metabolismo , Telomerasa/antagonistas & inhibidores , Telómero/efectos de los fármacos , Neoplasias Uterinas/tratamiento farmacológico , Animales , Línea Celular Tumoral , Núcleo Celular/metabolismo , Citoplasma/metabolismo , ADN/efectos de los fármacos , ADN/genética , ADN de Cadena Simple/efectos de los fármacos , ADN de Cadena Simple/genética , ADN de Cadena Simple/metabolismo , Proteínas de Unión al ADN , Femenino , G-Cuádruplex , Guanina/metabolismo , Humanos , Ratones , Ratones Desnudos , Telomerasa/biosíntesis , Telomerasa/metabolismo , Telómero/genética , Telómero/metabolismo , Ubiquitina/metabolismo , Neoplasias Uterinas/enzimología , Neoplasias Uterinas/genética , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Oncotarget ; 8(4): 6940-6954, 2017 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-28036297

RESUMEN

High-grade gliomas (HGG) are the most common brain tumors, with an average survival time of 14 months. A glioma-CpG island methylator phenotype (G-CIMP), associated with better clinical outcome, has been described in low and high-grade gliomas. Mutation of IDH1 is known to drive the G-CIMP status. In some cases, however, the hypermethylation phenotype is independent of IDH1 mutation, suggesting the involvement of other mechanisms. Here, we demonstrate that DNMT1 expression is higher in low-grade gliomas compared to glioblastomas and correlates with phosphorylated c-Jun. We show that phospho-c-Jun binds to the DNMT1 promoter and causes DNA hypermethylation. Phospho-c-Jun activation by Anisomycin treatment in primary glioblastoma-derived cells attenuates the aggressive features of mesenchymal glioblastomas and leads to promoter methylation and downregulation of key mesenchymal genes (CD44, MMP9 and CHI3L1). Our findings suggest that phospho-c-Jun activates an important regulatory mechanism to control DNMT1 expression and regulate global DNA methylation in Glioblastoma.


Asunto(s)
Neoplasias Encefálicas/metabolismo , ADN (Citosina-5-)-Metiltransferasa 1/genética , Metilación de ADN , Glioma/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Anisomicina/farmacología , Neoplasias Encefálicas/genética , Línea Celular Tumoral , ADN (Citosina-5-)-Metiltransferasa 1/metabolismo , Metilación de ADN/efectos de los fármacos , Epigénesis Genética/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genoma Humano , Glioma/genética , Humanos , Fosforilación , Pronóstico , Regiones Promotoras Genéticas/efectos de los fármacos , Análisis de Supervivencia , Regulación hacia Arriba/efectos de los fármacos
14.
Mol Cancer Res ; 15(8): 998-1011, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28512252

RESUMEN

Glioblastoma (GBM) comprises distinct subtypes characterized by their molecular profile. Mesenchymal identity in GBM has been associated with a comparatively unfavorable prognosis, primarily due to inherent resistance of these tumors to current therapies. The identification of molecular determinants of mesenchymal transformation could potentially allow for the discovery of new therapeutic targets. Zinc Finger and BTB Domain Containing 18 (ZBTB18/ZNF238/RP58) is a zinc finger transcriptional repressor with a crucial role in brain development and neuronal differentiation. Here, ZBTB18 is primarily silenced in the mesenchymal subtype of GBM through aberrant promoter methylation. Loss of ZBTB18 contributes to the aggressive phenotype of glioblastoma through regulation of poor prognosis-associated signatures. Restitution of ZBTB18 expression reverses the phenotype and impairs tumor-forming ability. These results indicate that ZBTB18 functions as a tumor suppressor in GBM through the regulation of genes associated with phenotypically aggressive properties.Implications: This study characterizes the role of the putative tumor suppressor ZBTB18 and its regulation by promoter hypermethylation, which appears to be a common mechanism to silence ZBTB18 in the mesenchymal subtype of GBM and provides a new mechanistic opportunity to specifically target this tumor subclass. Mol Cancer Res; 15(8); 998-1011. ©2017 AACR.


Asunto(s)
Metilación de ADN/genética , Epigénesis Genética/genética , Glioblastoma/genética , Proteínas Represoras/genética , Apoptosis/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Glioblastoma/patología , Humanos , Invasividad Neoplásica/genética , Pronóstico , Regiones Promotoras Genéticas
15.
Anat Embryol (Berl) ; 211(6): 639-48, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16977479

RESUMEN

Congenital myopathies often have an unclear aetiology. Here, we studied a novel case of a severe congenital myopathy with a failure of myotube formation. Polymerase chain reaction-based analysis was performed to characterize the expression patterns of the Desmin, p21, p57, and muscle regulatory factors (MRFs) MyoD, Myf4, Myf5 and Myf6 in differentiating skeletal muscle cells (SkMCs), normal human fibroblasts and patient-derived fibroblasts during trans-differentiation. The temporal and spatial pattern of MRFs was further characterized by immunocyto- and immunohistochemical stainings. In differentiating SkMCs, each MRF showed a characteristic expression pattern. Normal trans-differentiating fibroblasts formed myotubes and expressed all of the MRFs, which were detected. Interestingly, the patient's fibroblasts also showed some fusion events during trans-differentiation with a comparable expression profile for the MRFs, particularly, with increased expression of Myf4 and p21. Immunohistochemical analysis of normal and patient-derived skeletal musculature revealed that Myf4, which is downregulated during normal fetal development, was still present in patient-derived skeletal head muscle, which was also positive for Desmin and sarcomeric actin. The abnormal upregulation of Myf4 and p21 in the patient who suffered from a severe congenital myopathy suggests that the regulation of Myf4 and p21 gene expression during myogenesis might be of interest for further studies.


Asunto(s)
Diferenciación Celular/fisiología , Fibroblastos/citología , Desarrollo de Músculos/fisiología , Fibras Musculares Esqueléticas/citología , Miogenina/metabolismo , Miopatías Estructurales Congénitas/metabolismo , Células Cultivadas , Cromosomas Humanos , Análisis Citogenético , Resultado Fatal , Femenino , Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Humanos , Recién Nacido , Músculos/anomalías , Músculos/citología , Músculos/metabolismo , Miogenina/genética , Polimorfismo Genético , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Regulación hacia Arriba
16.
Anat Embryol (Berl) ; 211(6): 649-57, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17013620

RESUMEN

Genetic information that directs a cell during different phases of embryogenesis is locked up in the genome. Therein is contained the road map for growth, proliferation, differentiation and morphogenesis. The cellular transportation machinery plays a major role to ensure that all the components for transcription and translation are available at the right place at the right time. Nucleolar complex associated protein2 (NOC2) has a highly conserved UPF0120 domain, and is an element involved in ribosome transportation from the nucleoplasm to the cytoplasm. However, its gene expression pattern is still unknown. We chose the developing chick embryo to investigate the possible involvement of avian NOC2 (cNOC2) in developmental processes, particularly neurogenesis and myogenesis. For this purpose, we constructed a fragment of chicken cNOC2, which contains the UPF0120 domain coding sequence, into pDrive vector, and performed in situ hybridization on chicken embryos of different stages with this gene probe. A dynamic expression pattern of cNOC2 transcripts can be seen beginning as early as from stage HH7 until stage HH32. Using in situ hybridization we could detect that cNOC2 transcripts were expressed ubiquitously, but prominent expression could be found in the neural tissue, the somites and in the developing limbs. Comparison of cNOC2 gene expression with the proliferation marker gene cPCNA, muscle specific marker genes cMyf5 and cMyoD in single or double in situ hybridisation show that cNOC2 is expressed in the myotome, similar to cMyf5 and cMyoD, but not like cPCNA, which is hardly detectable in the myotome. Our results suggest that cNOC2 is involved in the development of neural tissue, somites and limbs.


Asunto(s)
Proteínas Portadoras/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas Nucleares/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas Portadoras/genética , Embrión de Pollo , Secuencia Conservada , Embrión no Mamífero , Extremidades/embriología , Datos de Secuencia Molecular , Proteína MioD/metabolismo , Factor 5 Regulador Miogénico/metabolismo , Neuronas/metabolismo , Proteínas Nucleares/genética , Antígeno Nuclear de Célula en Proliferación/metabolismo , Estructura Terciaria de Proteína/genética , Proteínas de Unión al ARN , Proteínas de Saccharomyces cerevisiae/genética , Homología de Secuencia de Aminoácido , Somitos/metabolismo
17.
EBioMedicine ; 12: 72-85, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27667176

RESUMEN

Glioblastomas are characterized by transcriptionally distinct subtypes, but despite possible clinical relevance, their regulation remains poorly understood. The commonly used molecular classification systems for GBM all identify a subtype with high expression of mesenchymal marker transcripts, strongly associated with invasive growth. We used a comprehensive data-driven network modeling technique (augmented sparse inverse covariance selection, aSICS) to define separate genomic, epigenetic, and transcriptional regulators of glioblastoma subtypes. Our model identified Annexin A2 (ANXA2) as a novel methylation-controlled positive regulator of the mesenchymal subtype. Subsequent evaluation in two independent cohorts established ANXA2 expression as a prognostic factor that is dependent on ANXA2 promoter methylation. ANXA2 knockdown in primary glioblastoma stem cell-like cultures suppressed known mesenchymal master regulators, and abrogated cell proliferation and invasion. Our results place ANXA2 at the apex of a regulatory cascade that determines glioblastoma mesenchymal transformation and validate aSICS as a general methodology to uncover regulators of cancer subtypes.


Asunto(s)
Anexina A2/metabolismo , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Glioblastoma/genética , Glioblastoma/metabolismo , Mesenquimoma/genética , Mesenquimoma/metabolismo , Algoritmos , Anexina A2/genética , Biomarcadores de Tumor , Línea Celular Tumoral , Biología Computacional/métodos , Metilación de ADN , Bases de Datos de Ácidos Nucleicos , Transición Epitelial-Mesenquimal , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Glioblastoma/mortalidad , Glioblastoma/patología , Humanos , Mesenquimoma/mortalidad , Mesenquimoma/patología , Anotación de Secuencia Molecular , Clasificación del Tumor , Células Madre Neoplásicas/metabolismo , Pronóstico , Regiones Promotoras Genéticas
18.
Anat Embryol (Berl) ; 210(1): 35-41, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16047188

RESUMEN

The chemokine receptor CXCR4 plays a decisive role in physiological cell migration both in developmental processes and adult tissues; it has also been implicated in metastasis formation of different human cancers (Balkwill 2004) and in HIV pathogenesis (Murdoch 2000). Here we present the expression pattern of this important chemokine receptor CXCR4 in the chick embryo. A dynamic expression pattern can be detected beginning as early as the gastrulation stages until the observed stage of HH28. During gastrulation, expression was observed in the epiblast at the level of the primitive streak and in the endoderm. Later, expression was noticeable in the ventral foregut portal, developing somites, tail bud, neural tube, the intermediate mesoderm, Wolffian duct, the lateral plate mesoderm and the developing blood vessels. Our descriptive data suggest a role for CXCR4 in gastrulation and other morphogenetic events connected with angiogenesis and kidney development.


Asunto(s)
Quimiocinas/metabolismo , Desarrollo Embrionario/fisiología , Neovascularización Fisiológica/fisiología , Organogénesis/fisiología , Receptores CXCR4/metabolismo , Animales , Vasos Sanguíneos/citología , Vasos Sanguíneos/embriología , Vasos Sanguíneos/metabolismo , Diferenciación Celular/fisiología , Movimiento Celular/fisiología , Embrión de Pollo , Endodermo/citología , Endodermo/metabolismo , Gástrula/citología , Gástrula/metabolismo , Riñón/citología , Riñón/embriología , Riñón/metabolismo , Mesodermo/citología , Mesodermo/metabolismo , Somitos/citología , Somitos/metabolismo
19.
Sci Rep ; 5: 10726, 2015 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-26137913

RESUMEN

Gastric cancer is the second leading cause of cancer-related death worldwide. RNA nanotechnology has recently emerged as an important field due to recent finding of its high thermodynamic stability, favorable and distinctive in vivo attributes. Here we reported the use of the thermostable three-way junction (3WJ) of bacteriophage phi29 motor pRNA to escort folic acid, a fluorescent image marker and BRCAA1 siRNA for targeting, imaging, delivery, gene silencing and regression of gastric cancer in animal models. In vitro assay revealed that the RNA nanoparticles specifically bind to gastric cancer cells, and knock-down the BRCAA1 gene. Apoptosis of gastric cancer cells was observed. Animal trials confirmed that these RNA nanoparticles could be used to image gastric cancer in vivo, while showing little accumulation in crucial organs and tissues. The volume of gastric tumors noticeably decreased during the course of treatment. No damage to important organs by RNA nanoparticles was detectible. All the results indicated that this novel RNA nanotechnology can overcome conventional cancer therapeutic limitations and opens new opportunities for specific delivery of therapeutics to stomach cancer without damaging normal cells and tissues, reduce the toxicity and side effect, improve the therapeutic effect, and exhibit great potential in clinical tumor therapy.


Asunto(s)
Antígenos de Neoplasias/genética , Nanopartículas/administración & dosificación , Proteínas de Neoplasias/genética , ARN Interferente Pequeño/administración & dosificación , Neoplasias Gástricas/terapia , Animales , Antígenos de Neoplasias/metabolismo , Apoptosis , Secuencia de Bases , Línea Celular Tumoral , Proliferación Celular , Femenino , Técnicas de Silenciamiento del Gen , Terapia Genética , Humanos , Inyecciones Intravenosas , Secuencias Invertidas Repetidas , Ratones Desnudos , Nanopartículas/efectos adversos , Proteínas de Neoplasias/metabolismo , Especificidad de Órganos , Interferencia de ARN , ARN Interferente Pequeño/efectos adversos , ARN Interferente Pequeño/genética , Neoplasias Gástricas/patología , Carga Tumoral
20.
PLoS One ; 9(3): e92679, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24671096

RESUMEN

The embryonic origin of the dermis in vertebrates can be traced back to the dermomyotome of the somites, the lateral plate mesoderm and the neural crest. The dermal precursors directly overlying the neural tube display a unique dense arrangement and are the first to induce skin appendage formation in vertebrate embryos. These dermal precursor cells have been shown to derive from the dorsomedial lip of the dermomyotome (DML). Based on its expression pattern in the DML, Wnt11 is a candidate regulator of dorsal dermis formation. Using EGFP-based cell labelling and time-lapse imaging, we show that the Wnt11 expressing DML is the source of the dense dorsal dermis. Loss-of-function studies in chicken embryos show that Wnt11 is indeed essential for the formation of dense dermis competent to support cutaneous appendage formation. Our findings show that dermogenic progenitors cannot leave the DML to form dense dorsal dermis following Wnt11 silencing. No alterations were noticeable in the patterning or in the epithelial state of the dermomyotome including the DML. Furthermore, we show that Wnt11 expression is regulated in a manner similar to the previously described early dermal marker cDermo-1. The analysis of Wnt11 mutant mice exhibits an underdeveloped dorsal dermis and strongly supports our gene silencing data in chicken embryos. We conclude that Wnt11 is required for dense dermis and subsequent cutaneous appendage formation, by influencing the cell fate decision of the cells in the DML.


Asunto(s)
Movimiento Celular , Pollos/metabolismo , Dermis/citología , Dermis/embriología , Células Madre/citología , Proteínas Wnt/metabolismo , Animales , Biomarcadores/metabolismo , Proteína Morfogenética Ósea 2/metabolismo , Embrión de Pollo , Regulación del Desarrollo de la Expresión Génica , Folículo Piloso/citología , Ratones Noqueados , Interferencia de ARN , Transducción de Señal , Células Madre/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA