Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Environ Toxicol ; 37(9): 2133-2142, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35506701

RESUMEN

The effects of ISG15 or ISGylation on tumor progression have been widely revealed; however, its roles in glioma progression are largely unknown. This study aims to explore the roles and underlying mechanisms of ISG15 in glioma progression. Here, ISG15 level was found to be upregulated in glioma tissues compared to the paired/unpaired normal tissues, and positively correlated with the level of stemness markers in glioma tissues. Loss of functional experiments indicated that ISG15 positively regulated glioma cell stemness, as evident by the increase of sphere formation ability, ALDH activity, stemness marker expression, and tumor-initiating ability. Further mechanistic studies revealed that ISG15 directly interacted with Oct4 protein, a critical stemness promoter, induced the ISGylation of Oct4 protein, and thus enhanced Oct4 protein stability. Additionally, it was found that Oct4 was ISGylated at lysine 284 (K284), which has been confirmed to be the ubiquitination site of Oct4 protein, and ISG15 knockdown did not degrade K284R mutant Oct4. Furthermore, ISG15 knockdown-induced downregulation of glioma cell stemness was rescued by Oct4 overexpression, but not by K284R mutant Oct4. Altogether, we suggest that ISG15-induced ISGylation of Oct4 protein is essential for glioma cell stemness.


Asunto(s)
Citocinas , Glioma , Factor 3 de Transcripción de Unión a Octámeros , Ubiquitinas , Citocinas/genética , Citocinas/metabolismo , Regulación hacia Abajo , Glioma/genética , Humanos , Células Madre Neoplásicas , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Estabilidad Proteica , Ubiquitinación , Ubiquitinas/genética , Ubiquitinas/metabolismo
2.
J Cell Mol Med ; 24(24): 14171-14183, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33124187

RESUMEN

Sustained hyperglycaemia and hyperlipidaemia incur endoplasmic reticulum stress (ER stress) and reactive oxygen species (ROS) overproduction in pancreatic ß-cells. ER stress or ROS causes c-Jun N-terminal kinase (JNK) activation, and the activated JNK triggers apoptosis in different cells. Nuclear receptor subfamily 4 group A member 1 (NR4A1) is an inducible multi-stress response factor. The aim of this study was to explore the role of NR4A1 in counteracting JNK activation induced by ER stress or ROS and the related mechanism. qPCR, Western blotting, dual-luciferase reporter and ChIP assays were applied to detect gene expression or regulation by NR4A1. Immunofluorescence was used to detect a specific protein expression in ß-cells. Our data showed that NR4A1 reduced the phosphorylated JNK (p-JNK) in MIN6 cells encountering ER stress or ROS and reduced MKK4 protein in a proteasome-dependent manner. We found that NR4A1 increased the expression of cbl-b (an E3 ligase); knocking down cbl-b expression increased MKK4 and p-JNK levels under ER stress or ROS conditions. We elucidated that NR4A1 enhanced the transactivation of cbl-b promoter by physical association. We further confirmed that cbl-b expression in ß-cells was reduced in NR4A1-knockout mice compared with WT mice. NR4A1 down-regulates JNK activation by ER stress or ROS in ß-cells via enhancing cbl-b expression.


Asunto(s)
Estrés del Retículo Endoplásmico , Células Secretoras de Insulina/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Línea Celular , Regulación de la Expresión Génica , Peróxido de Hidrógeno/metabolismo , MAP Quinasa Quinasa 4/metabolismo , Ratones , Ratones Noqueados , Modelos Biológicos , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Fosforilación , Regiones Promotoras Genéticas , Unión Proteica , Ubiquitinación
3.
Cell Physiol Biochem ; 44(4): 1616-1628, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29212066

RESUMEN

BACKGROUND: Gliomas result in the highest morbidity and mortality rates of intracranial primary central nervous system tumors because of their aggressive growth characteristics and high postoperative recurrence. They are characterized by genetic instability, intratumoral histopathological variability and unpredictable clinical behavior in patients. Proliferation is a key aspect of the clinical progression of malignant gliomas, complicating complete surgical resection and enabling tumor regrowth and further proliferation of the surviving tumor cells. METHODS: The expression of Fstl1 was detected by western blotting and qRT-PCR. We used cell proliferation and colony formation assays to measure proliferation. Then, flow cytometry was used to analyze cell cycle progression. The expression of Fstl1, p-Smad1/5/8 and p21 in GBM tissue sections was evaluated using immunohistochemical staining. Furthermore, we used coimmunoprecipitation (Co-IP) and immunoprecipitation to validate the relationship between Fstl1, BMP4 and BMPR2. Finally, we used orthotopic xenograft studies to measure the growth of tumors in vivo. RESULTS: We found that follistatin-like 1 (Fstl1) was upregulated in high-grade glioma specimens and that its levels correlated with poor prognosis. Fstl1 upregulation increased cell proliferation, colony formation and cell cycle progression, while its knockdown inhibited these processes. Moreover, Fstl1 interacted with bone morphogenetic protein (BMP) 4, but not BMP receptor (BMPR) 2, and competitively inhibited their association. Furthermore, Fstl1 overexpression suppressed the activation of the BMP4/Smad1/5/8 signaling pathway, while BMP4 overexpression reversed this effect. CONCLUSION: Our study demonstrated that Fstl1 promoted glioma growth through the BMP4/Smad1/5/8 signaling pathway, and these findings suggest potential new glioblastoma treatment strategies.


Asunto(s)
Proteína Morfogenética Ósea 4/metabolismo , Neoplasias Encefálicas/patología , Proteínas Relacionadas con la Folistatina/metabolismo , Glioma/patología , Proteína Smad1/metabolismo , Proteína Smad5/metabolismo , Proteína Smad8/metabolismo , Animales , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/metabolismo , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/mortalidad , Línea Celular Tumoral , Proteínas Relacionadas con la Folistatina/antagonistas & inhibidores , Proteínas Relacionadas con la Folistatina/genética , Glioma/metabolismo , Glioma/mortalidad , Humanos , Inmunoprecipitación , Estimación de Kaplan-Meier , Ratones , Fosforilación , Unión Proteica , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Puntos de Control de la Fase S del Ciclo Celular , Transducción de Señal , Trasplante Heterólogo
4.
J Neurooncol ; 133(1): 59-68, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28425046

RESUMEN

Glioblastoma is one of the most frequent and aggressive brain tumors. Accumulating evidence indicates that microRNAs are involved in glioma proliferation, invasion and drug resistance. Previous studies showed that miR-198 is downregulated in glioblastoma. However, the function of miR-198 in glioblastoma is still unclear. In this study, we report that miR-198 levels were greatly downregulated in glioblastoma specimens and decreased expression of miR-198 was associated with poor prognosis in patients with glioblastoma. And overexpression of miR-198 increased chemosensitivity to temozolomide in vitro and in vivo. O6-methylguanine-DNA methyltransferase (MGMT) was identified as a direct target of miR-198, and miR-198 overexpression prevented the protein translation of MGMT. Furthermore, overexpression of MGMT restored miR-198-induced chemosensitivity to temozolomide. Moreover, the protein levels of MGMT were upregulated in clinical glioblastoma specimens and inversely correlated with miR-198 levels. In conclusion, our studies revealed that MiR-198 induces chemosensitivity in glioblastoma by targeting MGMT and that miR-198 may be used as a new diagnostic marker and therapeutic target for glioblastoma in the future.


Asunto(s)
Antineoplásicos Alquilantes/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Metilasas de Modificación del ADN/metabolismo , Enzimas Reparadoras del ADN/metabolismo , Dacarbazina/análogos & derivados , Glioblastoma/tratamiento farmacológico , MicroARNs/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Adulto , Anciano , Animales , Antineoplásicos Alquilantes/farmacología , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Metilasas de Modificación del ADN/genética , Enzimas Reparadoras del ADN/genética , Dacarbazina/farmacología , Dacarbazina/uso terapéutico , Resistencia a Antineoplásicos/fisiología , Femenino , Glioblastoma/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Masculino , Ratones Endogámicos NOD , Ratones SCID , Persona de Mediana Edad , Trasplante de Neoplasias , ARN Mensajero/metabolismo , Temozolomida , Proteínas Supresoras de Tumor/genética , Adulto Joven
5.
Oncogene ; 42(6): 461-470, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36517590

RESUMEN

Glioblastoma (GBM) is the most lethal primary brain tumor in adults and harbors a subpopulation of glioma stem cells (GSCs). Enhancer of Zeste Homolog 2 (EZH2), a histone lysine methyltransferase, deeply involves in the stemness maintenance of GSC. However, the precise mechanism and therapeutic potential remain elusive. We postulated that the interactome of EZH2 in GSC is unique. Therefore, we performed proteomic and transcriptomic research to unveil the oncogenic mechanism of EZH2. Immunoprecipitation and mass spectrometry were used to identify proteins that co-precipitate with EZH2. We show that EZH2 binds to heterochromatin protein 1 binding protein 3 (HP1BP3) in GSCs and impairs the methylation of H3K9. Overexpression of HP1BP3 enhances the proliferation, self-renewal and temozolomide (TMZ) resistance of GBM cells. Furthermore, EZH2 and HP1BP3 co-activate WNT7B expression thereby increasing TMZ resistance and stemness of GBM cells. Importantly, inhibition of WNT7B autocrine via LGK974 effectively reverses the TMZ resistance. Our work clarifies a new oncogenic mechanism of EZH2 by which it interacts with HP1BP3 and epigenetically activates WNT7B thereby promoting TMZ resistance in GSCs. Our results provide a rationale for targeting WNT/ß-catenin pathway as a promising strategy to overcome TMZ resistance in GSCs.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Humanos , Temozolomida/farmacología , Temozolomida/uso terapéutico , Glioblastoma/tratamiento farmacológico , Glioblastoma/genética , Glioblastoma/metabolismo , Proteína Potenciadora del Homólogo Zeste 2/genética , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Proteómica , Vía de Señalización Wnt , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Resistencia a Antineoplásicos/genética , Células Madre Neoplásicas/patología , Proteínas Wnt/metabolismo
6.
Front Immunol ; 13: 841404, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35265085

RESUMEN

The glioma immune microenvironment (GIM), consisting of glioma cells, stromal cells, and immune cells, accelerates the initiation, development, immune evasion, chemoresistance, and radioresistance of glioblastoma (GBM), whereas the immunosuppressive mechanisms of GBM have not been thoroughly elucidated to date. The glioma data downloaded from The Cancer Genome Atlas (TCGA) and the Chinese Glioma Genome Atlas (CGGA) databases were used to evaluate the composition of tumor-infiltrating immune cells (TICs) by the CIBERSORT algorithm. RNA-seq datasets from the TCGA and CGGA were used to analyze the relationship between immune scores with patients' characteristics and TICs, which showed higher ratios of tumor-inhibiting/tumor-promoting signatures (M2/M1 macrophages) along with higher immune scores. The distribution of TICs among different glioma patients and the correlation with hazard ratio (HR) analysis suggested that M2 macrophages were abundant in malignant gliomas and indicated an unfavorable prognosis. We further analyzed TCGA cases with available mutation and copy-number alteration information, which showed that the status of PTEN could influence the immune microenvironment of glioma patients. Tissue microarrays of 39 GBM patients were carried out to confirm the clinical significance of PTEN and macrophage markers. We found that the high expression of PTEN was associated with a more extended survival period of glioma patients, positively correlated with M2 macrophages and negatively with M1 macrophages. Transwell and flow cytometry analyses demonstrated that PTEN status could prevent M1 to M2 polarization and M2 macrophage recruitment of gliomas in vitro. The newly discovered immunoregulatory activity of PTEN opens innovative avenues for investigations relevant to counteracting cancer development and progression.


Asunto(s)
Glioblastoma , Glioma , Macrófagos , Fosfohidrolasa PTEN , Microambiente Tumoral , Regulación Neoplásica de la Expresión Génica , Glioblastoma/genética , Glioblastoma/patología , Glioma/genética , Glioma/patología , Humanos , Macrófagos/metabolismo , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Pronóstico , Microambiente Tumoral/genética
7.
Mol Ther Nucleic Acids ; 25: 25-36, 2021 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-34168916

RESUMEN

A number of studies indicate that circular RNAs (circRNAs) play paramount roles in regulating the biological behavior of glioblastoma multiforme (GBM). In this study, we investigated the underlying mechanism of circMELK in GBM. Real-time PCRs were used to examine the expression of circMELK in glioma tissues and normal brain tissues (NBTs). Localization of circMELK in GBM cells was estimated by fluorescence in situ hybridization (FISH). Transwell migration and three-dimensional invasion assays were performed to examine glioma cell migration and invasion in vitro. Spheroid formation, clonogenicity, and cell viability assays were implemented to test the stemness of glioma stem cells (GSCs). The functions of circMELK in vivo were investigated in a xenograft nude-mouse model. We have proved that circMELK functions as a sponge for tumor suppressor microRNA-593 (miR-593) by RNA immunoprecipitation and circRNA precipitation assays, which targets the oncogenic gene Eph receptor B2 (EphB2). Dual-luciferase reporter assays were adopted to estimate the interactions between miR-593 and circMELK or EphB2. We demonstrated that circMELK was upregulated in GBM, acting as an oncogene and regulating GBM mesenchymal transition and GSC maintenance via sponging of miR-593. Furthermore, we found that EphB2 was involved in circMELK/miR-593 axis-induced GBM tumorigenesis. This function opens the opportunity for the development of a novel therapeutic target for the treatment of gliomas.

8.
Neuro Oncol ; 23(3): 435-446, 2021 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-32813021

RESUMEN

BACKGROUND: Our previous studies have indicated that miR-198 reduces cellular methylguanine DNA methyltransferase (MGMT) levels to enhance temozolomide sensitivity. Transforming growth factor beta 1 (TGF-ß1) switches off miR-198 expression by repressing K-homology splicing regulatory protein (KSRP) expression in epidermal keratinocytes. However, the underlying role of TGF-ß1 in temozolomide resistance has remained unknown. METHODS: The distribution of KSRP was detected by western blotting and immunofluorescence. Microarray analysis was used to compare the levels of long noncoding RNAs (lncRNAs) between TGF-ß1-treated and untreated cells. RNA immunoprecipitation was performed to verify the relationship between RNAs and KSRP. Flow cytometry and orthotopic and subcutaneous xenograft tumor models were used to determine the function of TGF-ß1 in temozolomide resistance. RESULTS: Overexpression of TGF-ß1 contributed to temozolomide resistance in MGMT promoter hypomethylated glioblastoma cells in vitro and in vivo. TGF-ß1 treatment reduced cellular MGMT levels through suppressing the expression of miR-198. However, TGF-ß1 upregulation did not affect KSRP expression in glioma cells. We identified and characterized 2 lncRNAs (H19 and HOXD-AS2) that were upregulated by TGF-ß1 through Smad signaling. H19 and HOXD-AS2 exhibited competitive binding to KSRP and prevented KSRP from binding to primary miR-198, thus decreasing miR-198 expression. HOXD-AS2 or H19 upregulation strongly promoted temozolomide resistance and MGMT expression. Moreover, KSRP depletion abrogated the effects of TGF-ß1 and lncRNAs on miR-198 and MGMT. Finally, we found that patients with low levels of TGF-ß1 or lncRNA expression benefited from temozolomide therapy. CONCLUSIONS: Our results reveal an underlying mechanism by which TGF-ß1 confers temozolomide resistance. Furthermore, our findings suggest that a novel combination of temozolomide with a TGF-ß inhibitor may serve as an effective therapy for glioblastomas.


Asunto(s)
Glioblastoma , MicroARNs , Antineoplásicos Alquilantes/farmacología , Antineoplásicos Alquilantes/uso terapéutico , Línea Celular Tumoral , Metilasas de Modificación del ADN/genética , Enzimas Reparadoras del ADN , Resistencia a Antineoplásicos/genética , Glioblastoma/tratamiento farmacológico , Glioblastoma/genética , Humanos , MicroARNs/genética , Temozolomida/farmacología , Temozolomida/uso terapéutico , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/uso terapéutico , Proteínas Supresoras de Tumor/genética
9.
Cell Death Discov ; 7(1): 133, 2021 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-34088892

RESUMEN

Under adverse conditions, such as sustained or chronic hyperglycemia or hyperlipidemia, ROS (reactive oxygen species) or/and ER-stress (endoplasmic reticulum stress) will be induced in pancreatic ß cells. ROS or ER-stress damages ß-cells even leads to apoptosis. Previously we found ROS or ER-stress resulted in JNK activation in ß cells and overexpressing NR4A1 in MIN6 cells reduced JNK activation via modulating cbl-b expression and subsequent degrading the upstream JNK kinase (MKK4). To search other possible mechanisms, we found the mRNA level and protein level of MKP7 (a phosphatase for phospho-JNK) were dramatic reduced in pancreatic ß cells in the islets from NR4A1 KO mice compared with that from wild type mice. To confirm what we found in animals, we applied pancreatic ß cells (MIN6 cells) and found that the expression of MKP7 was increased in NR4A1-overexpression MIN6 cells. We further found that knocking down the expression of MKP7 increased the p-JNK level in pancreatic ß cells upon treatment with TG or H2O2. After that, we figured out that NR4A1 did enhance the transactivation of the MKP7 promoter by physical association with two putative binding sites. In sum, NR4A1 attenuates JNK phosphorylation incurred by ER-stress or ROS partially via enhancing MKP7 expression, potentially decreases pancreatic ß cell apoptosis induced by ROS or ER-stress. Our finding provides a clue for diabetes prevention.

10.
J Exp Clin Cancer Res ; 38(1): 289, 2019 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-31277685

RESUMEN

BACKGROUND: Glioblastoma (GBM) cells with stem cell-like properties are called glioma stem cells (GSCs). GSCs display highly treatment resistance and are responsible for tumor recurrence. Napabucasin (BBI608), a novel small molecule inhibitor of STAT3, has been identified to eliminate stemness-like tumor cells in some cancers. However, the influence of Napabucasin on GBM cells, especially on GSCs, is currently unclear. In this study, we explored the influence and underlying mechanisms of Napabucasin on GBM cells. METHODS: STAT3 expression and its correlation with the glioma grade and patient survival were analyzed using CGGA and TCGA glioma databases. The influence of Napabucasin on proliferation, stemness, the cell cycle, apoptosis, and invasion of human GBM cell lines U87MG and LN229 was tested by CCK8, EdU incorporation, colony formation, Transwell invasion, and three-dimensional spheroid assays as well as flow cytometry, qPCR, and western blot analysis. The ability of Napabucasin to inhibit cell proliferation of U87MG tumor xenografts in mice was assessed using a live animal bioluminescence imaging system and immunohistochemistry. RESULTS: Napabucasin suppressed the proliferation, colony formation, and invasion of U87MG and LN229 cells. Furthermore, Napabucasin induced cell cycle arrest and apoptosis. More importantly, Napabucasin treatment obviously inhibited expression of stemness-associated genes including STAT3 and suppressed the spheroid formation of glioma cells in vitro. Napabucasin also disrupted the NF-κB signaling pathway via downregulation of RelA (p65). Finally, glioma growth was effectively impaired by Napabucasin in nude mice bearing intracranial glioma xenografts. CONCLUSIONS: Napabucasin treatment may be a novel approach for the treatment of GBM, particularly GSCs.


Asunto(s)
Benzofuranos/uso terapéutico , Glioblastoma/tratamiento farmacológico , Naftoquinonas/uso terapéutico , Factor de Transcripción STAT3/antagonistas & inhibidores , Células Madre/metabolismo , Animales , Benzofuranos/farmacología , Proliferación Celular/efectos de los fármacos , Femenino , Glioblastoma/patología , Humanos , Ratones , Ratones Desnudos , Naftoquinonas/farmacología
11.
J Exp Clin Cancer Res ; 38(1): 133, 2019 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-30898167

RESUMEN

BACKGROUND: Glioblastoma multiforme (GBM) is the most common and aggressive form of astrocytoma among adult brain tumors. Multiple studies have shown that long non-coding RNAs (lncRNAs) play important roles in acting as molecular sponge for competing with microRNAs (miRNAs) to regulate downstream molecules in tumor progression. We previously reported that miR155 host gene (miR155HG), an lncRNA, and its derivative miR-155 promote epithelial-to-mesenchymal transition in glioma. However, the other biological functions and mechanisms of miR155HG sponging miRNAs have been unknown. Considering ANXA2 has been generally accepted as oncogene overexpressed in a vast of cancers correlated with tumorigenesis, which might be the target molecule of miR155HG sponging miRNA via bioinformatics analysis. We designed this study to explore the interaction of miR155HG and ANXA2 to reveal the malignancy of them in GBM development. METHODS: The expression of miR155HG was analyzed in three independent databases and clinical GBM specimens. Bioinformatics analysis was performed to assess the potential tumor-related functions of miR155HG. The interaction of miR155HG and miR-185 and the inhibition of ANXA2 by miR-185 were analyzed by luciferase reporter experiments, and biological effects in GBM were explored by colony formation assays, EDU cell proliferation assays, flow cytometric analysis and intracranial GBM mouse model. Changes in protein expression were analyzed using western blot. We examined the regulatory mechanism of ANXA2 on miR155HG in GBM by gene expression profiling analysis, double immunofluorescence staining, chromatin immunoprecipitation and luciferase reporter assays. RESULTS: We found that miR155HG was upregulated in GBM tissues and cell lines. Bioinformatic analyses of three GBM databases showed that miR155HG expression levels were closely associated with genes involved in cell proliferation and apoptosis. Knocking down miR155HG suppressed GBM cell proliferation in vitro, induced a G1/S-phase cell cycle arrest, and increased apoptosis. We also found that miR155HG functions as a competing endogenous RNA for miR-185. Moreover, miR-185 directly targets and inhibits ANXA2, which exhibits oncogenic functions in GBM. We also found that ANXA2 promoted miR155HG expression via STAT3 phosphorylation. CONCLUSION: Our results demonstrated that overexpressed miR155HG in GBM can sponge miR-185 to promote ANXA2 expression, and ANXA2 stimulates miR155HG level through phosphorylated STAT3 binding to the miR155HG promoter. We establish the miR155HG/miR185/ANXA2 loop as a mechanism that underlies the biological functions of miR155HG and ANXA2 in GBM and further suggest this loop may serve as a therapeutic target and/or prognostic biomarker for GBM.


Asunto(s)
Anexina A2/genética , Neoplasias Encefálicas/patología , Glioblastoma/patología , MicroARNs/genética , ARN Largo no Codificante/genética , Animales , Neoplasias Encefálicas/genética , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Progresión de la Enfermedad , Transición Epitelial-Mesenquimal , Regulación Neoplásica de la Expresión Génica , Glioblastoma/genética , Humanos , Ratones , Trasplante de Neoplasias
12.
Am J Transl Res ; 11(8): 4851-4865, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31497204

RESUMEN

Glioblastoma (GBM) is the most frequently occurred malignant human tumor that arise in brain with a poor prognosis. microRNAs (miRNAs) are vital small molecules during GBM initiation and progression. However, the expression of miR-940 and its potential function in GBM remain poor. Our study demonstrated that miR-940 was dramatically decreased in GBM cells and glioma tissues. Introduction of miR-940 significantly repressed proliferative ability of GBM cells. Notably, treatment of miR-940 dramatically suppressed tumor growth in an animal model, accompanied by decreased Ki67 expression. Functional experiments showed CKS1 as a target of miR-940, knockdown of CKS1 significantly induced the cell cycle arrest and restrained GBM cells proliferation, consistent with miR-940 treatment. Furthermore, reintroduction of CKS1 into glioma cells effectively rescued the tumor suppressive effect of miR-940. Correlation analysis indicated that miR-940 expression was inversely related to CKS1 mRNA levels in NBTs and gliomas. Together, miR-940/CKS1 signaling may be required for GBM progression and provide a new insight in diagnosis and prognosis of GBM patients.

13.
Am J Transl Res ; 11(12): 7351-7363, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31934283

RESUMEN

MicroRNAs have been found ectopically expressed in many cancers and play essential roles in tumor EMT progress. Recent studies identified decreased miR-940 expression in glioma cells and may serve as a tumor-suppressor. However, whether miR-940 involve in glioma EMT remain poorly understood. Here we confirmed that miR-940 was significantly reduced in glioma cells and tissues. Introduction of miR-940 dramatically suppressed invasion and migration of glioma cells. Gain-of-function experiments showed ZEB2 as a direct target of miR-940, knockdown of ZEB2 evidently repressed invasive capacity of glioma cells through EMT. Moreover, reintroduction of ZEB2 effectively reversed the tumor suppressive effect of miR-940 treatment. In vivo study showed reduced tumor cell motion in miR-940-injected groups. Spearman's correlation analysis indicated inversely correlated expression of ZEB2 and miR-940 in gliomas and NBTs. Altogether, miR-940-ZEB2 cascade may play important roles in glioma cells invasion and EMT progression, and might provide new therapeutic approaches for better outcomes of GBM patients.

14.
Neuro Oncol ; 21(4): 462-473, 2019 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-30590814

RESUMEN

BACKGROUND: Epithelial cell transformation sequence 2 (ECT2) is upregulated in glioma and promotes glioma cell proliferation. A preliminary experiment showed a positive correlation between ECT2 and pituitary tumor-transforming gene 1 (PTTG1). The aim of this study was to explore how ECT2 affects PTTG1 to influence the proliferation of glioma cells. METHODS: The expression of ECT2 in glioma was detected by western blot and reverse transcription PCR. The effect of ECT2 on glioma proliferation was examined using cell proliferation-related assays and in vivo experiments. The effect of ECT2 on the stability of E2F transcription factor 1 (E2F1) and the expression of PTTG1 were examined by western blot, co-immunoprecipitation, and in vivo ubiquitination assays. RESULTS: ECT2 was upregulated in gliomas and was negatively correlated with prognosis; its downregulation inhibited glioma cell proliferation. Furthermore, ECT2 regulated PTTG1 expression by affecting the stability of E2F1, thereby affecting the glioma cell proliferation. In addition, the deubiquitinating enzyme proteasome 26S subunit, non-ATPase 14 (PSMD14) affected the degradation of E2F1 and regulated the stability of E2F1. Interestingly, ECT2 regulated the expression of PSMD14. CONCLUSION: In this study, we clarify a new mechanism by which ECT2 regulates the expression of PTTG1 and thus affects the proliferation of glioma cells: ECT2 influences the stability of E2F1 by regulating the expression of the deubiquitinating enzyme PSMD14, thereby affecting the expression of PTTG1. Intensive and extensive understanding of the mechanism of ECT2 in glioma proliferation may provide an opportunity for the development of new molecular therapeutic targets for glioma treatment.


Asunto(s)
Neoplasias Encefálicas/patología , Glioma/patología , Proteínas Proto-Oncogénicas/metabolismo , Securina/metabolismo , Animales , Neoplasias Encefálicas/metabolismo , Proliferación Celular/fisiología , Factor de Transcripción E2F1/metabolismo , Glioma/metabolismo , Xenoinjertos , Humanos , Ratones , Ratones Desnudos , Complejo de la Endopetidasa Proteasomal/metabolismo , Transducción de Señal/fisiología , Transactivadores/metabolismo
15.
Am J Transl Res ; 11(7): 4584-4601, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31396362

RESUMEN

Glioma is one of the most prevalent primary malignant brain tumours among adults, and accumulating evidence has shown that dysregulation of microRNAs (miRNAs) is associated with various types of cancers, including glioma. It is necessary to gain a better understanding of the roles and mechanisms of action of miRNAs in WNT-driven glioblastoma multiforme (GBM). Here, we report that miR-206 inhibits the WNT/ß-catenin pathway by directly targeting Frizzled 7 (FZD7) mRNA and functions as a tumour suppressor in glioma. The expression of miR-206 in human glioma samples and glioma cells was assessed by reverse-transcription quantitative PCR, fluorescence in situ hybridisation, and histological analysis. Cell Counting Kit-8, colony formation, 5-ethynyl-2'-deoxyuridine incorporation, flow-cytometric, wound healing, Transwell invasion, and three-dimensional migration assays were performed to examine glioma cell proliferation, migration, and invasion in vitro. The effects of miR-206 in vivo were investigated in a xenograft nude-mouse model. MiR-206 expression was significantly lower in glioma specimens than in normal control samples. FZD7 was confirmed as a direct target gene of miR-206. GBM cell proliferation, migration, and invasion were blocked after restoration of miR-206 expression. Moreover, intracranial glioma models revealed an inhibitory effect of miR-206 on intracranial glioma tumour growth. Our results suggest that miR-206 plays a key role in the blockade of the WNT/ß-catenin signalling pathway by down-regulating FZD7 and may be a promising therapeutic agent against malignant glioma and other WNT-driven tumours.

16.
Oncogene ; 38(15): 2706-2721, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30542120

RESUMEN

Temozolomide was recognized as the first-line therapy for glioblastoma to prolong the survival of patients noticeably, while recent clinical studies found that some patients were not sensitive to temozolomide treatment. The possible mechanisms seemed to be methylguanine-DNA-methyltransferase (MGMT), mismatch repair, PARP, etc. And the abnormal expression of MGMT might be the most direct factor. In this study, we provide evidence that Fstl1 plays a vital role in temozolomide resistance by sequentially regulating DIP2A protein distribution, H3K9 acetylation (H3K9Ac), and MGMT transcription. As a multifunctional protein widely distributed in cells, DIP2A cooperates with the HDAC2-DMAP1 complex to enhance H3K9Ac deacetylation, prevent MGMT transcription, and increase temozolomide sensitivity. Fstl1, a glycoprotein highly expressed in glioblastoma, competitively binds DIP2A to block DIP2A nuclear translocation, so as to hinder DIP2A from binding the HDAC2-DMAP1 complex. The overexpression of Fstl1 promoted the expression of MGMT in association with increased promoter H3K9Ac. Upregulation of Fstl1 enhanced temozolomide resistance, whereas Fstl1 silencing obviously sensitized GBM cells to temozolomide both in vivo and in vitro. Moreover, DIP2A depletion abolished the effects of Fstl1 on MGMT expression and temozolomide resistance. These findings highlight an important role of Fstl1 in the regulation of temozolomide resistance by modulation of DIP2A/MGMT signaling.


Asunto(s)
Proteínas Portadoras/genética , Metilasas de Modificación del ADN/genética , Enzimas Reparadoras del ADN/genética , Resistencia a Antineoplásicos/genética , Proteínas Relacionadas con la Folistatina/genética , Glioblastoma/tratamiento farmacológico , Glioblastoma/genética , Proteínas Nucleares/genética , Temozolomida/farmacología , Proteínas Supresoras de Tumor/genética , Acetilación , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica/genética , Histona Desacetilasa 2 , Humanos , Regiones Promotoras Genéticas/genética , Proteínas Represoras/genética , Transducción de Señal/genética , Transcripción Genética/genética , Regulación hacia Arriba/genética
17.
Cancer Lett ; 433: 210-220, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30008386

RESUMEN

The glioma-astrocyte interaction plays an important role in tumor microenvironment remodeling; however, the underlying mechanism has not been completely clarified. In this study, we show that glioma cells stimulate normal human astrocyte (NHA) into reactive astrocyte (RAS) in a non-contact manner. Additionally, the amount of O6-alkylguanine DNA alkyltransferase (MGMT) mRNA in exosomes (EXOs) released by RAS was significantly increased compared with that in non-reactive NHA. Importantly, MGMT-negative glioma cells can take up RAS-EXOs and acquire a temozolomide (TMZ)-resistant phenotype via the translation of exogenous exosomal MGMT mRNA both in vitro and in vivo. Our findings illuminate a novel phenomenon that may be a potent mechanism underlying glioma recurrence in which glioma-associated NHAs protect MGMT-negative glioma cells from TMZ-induced apoptosis by the functional intercellular transfer of exosomal MGMT mRNA.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Astrocitos/metabolismo , Neoplasias Encefálicas/patología , Metilasas de Modificación del ADN/genética , Metilasas de Modificación del ADN/metabolismo , Enzimas Reparadoras del ADN/genética , Enzimas Reparadoras del ADN/metabolismo , Resistencia a Antineoplásicos/genética , Exosomas/metabolismo , Glioma/patología , ARN Mensajero/genética , Temozolomida/farmacología , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Apoptosis/genética , Línea Celular Tumoral , Humanos , Microambiente Tumoral
18.
Oncotarget ; 8(16): 26637-26647, 2017 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-28460453

RESUMEN

Chromobox homolog 7 (CBX7) cooperates with other polycomb group (PcG) proteins to maintain target genes in a silenced state. However, the precise role of CBX7 in tumor progression is still controversial. We found that the expression of CBX7 in four public databases was significantly lower in high grade glioma (HGG). The reduced expression of CBX7 correlated with poor outcome in HGG patients. Both KEGG and GO analyses indicated that genes that were negatively correlated to CBX7 were strongly associated with the cell cycle pathway. We observed that decreased CBX7 protein levels enhanced glioma cells proliferation, migration and invasion. Then, we verified that CBX7 overexpression arrested cells in the G0/G1 phase. Moreover, we demonstrated that the underlying mechanism involved in CBX7 induced repression of CCNE1 promoter requiring the recruitment of histone deacetylase 2 (HADC2). Finally, in vivo bioluminescence imaging and survival times of nude mice revealed that CBX7 behaved as a tumor suppressor in gliomas. In summary, our results validate the assumption that CBX7 is a tumor suppressor of gliomas. Moreover, CBX7 is a potential and novel prognostic biomarker in glioma patients. We also clarified that CBX7 silences CCNE1 via the combination of CCNE1 promoter and the recruitment of HDAC2.


Asunto(s)
Ciclina E/genética , Puntos de Control de la Fase G1 del Ciclo Celular/genética , Silenciador del Gen , Glioma/genética , Glioma/mortalidad , Proteínas Oncogénicas/genética , Complejo Represivo Polycomb 1/genética , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Glioma/patología , Xenoinjertos , Histona Desacetilasa 2/metabolismo , Humanos , Masculino , Ratones , Clasificación del Tumor , Complejo Represivo Polycomb 1/metabolismo , Pronóstico , Regiones Promotoras Genéticas , Unión Proteica , ARN Interferente Pequeño/genética
19.
Oncotarget ; 8(41): 71080-71094, 2017 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-29050344

RESUMEN

Glioblastoma multiforme is the most common primary malignancy in the brain and confers a uniformly poor prognosis. MicroRNAs have been shown to activate or inhibit tumorigenesis. Abnormalities in the p53 signaling pathway are found in various cancers and correlate with tumor formation. We examined the expression of microRNA-141-3p (miR-141-3p) in glioma of different grades by analysis of expression profiling databases and clinical specimens. Cell proliferation and flow cytometry assays were performed to evaluate the promotion of miR-141-3p in proliferation, cell cycle, apoptosis, and temozolomide resistance of glioblastoma cells in vitro. Bioinformatics analyses, luciferase reporter assays, and immunoblotting showed that p53 is a target gene of miR-141-3p. A significant inverse correlation was observed between expression of miR-141-3p and p53 in glioma and normal brain tissues (R2=0.506, P<0.0001). Rescue experiments indicated that overexpression of p53 significantly reversed the alterations in proliferation, cell cycle distribution, and temozolomide resistance measured by cell apoptosis induced by miR-141-3p overexpression. In an orthotopic mouse model of human glioma, inhibition of miRNA-141-3p reduced the proliferation and growth of glioma cells in the brain and significantly prolonged the survival of glioma-bearing mice. We suggest that miR-141-3p promotes glioblastoma progression and temozolomide resistance by altering p53 expression and therefore may serve as a new diagnostic marker and therapeutic target for glioma in the future.

20.
Oncol Lett ; 13(4): 2583-2590, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28454437

RESUMEN

Polycomb group (PcG) proteins form at least two key complexes, namely polycomb repressive complex 1 and polycomb repressive complex 2. These complexes are involved in the progression of various cancers. Systematic research has not been conducted on the aberrant expression of PcG members in gliomas. Using the Chinese Glioma Genome Atlas data set, PcG expression patterns between normal brain tissues and glioma samples were analyzed, and a PcG-based classifier was then developed using BRB Cox regression and risk-score model. These results were validated in an independent GSE16011 set. A total of six PcGs [chromobox protein homolog (CBX) 6, CBX7, PHD finger protein 1, enhancer of zeste homolog 2 (EZH2), DNA (cytosine-5-)-methyltransferase 3ß (DNMT3B) and polyhomeotic-like protein 2] were identified to be associated with glioma grade. Survival analysis then revealed a five-PcG gene signature one protective gene (enhancer of zeste homolog 1) and four risky genes (EZH2, PHD finger protein 19, DNMT3A and DNMT3B), which may identify patients with high risk of poor prognosis of glioma. Multivariate Cox analysis indicated that the five-PcG signature was an independent prognostic biomarker. These findings indicated that a novel prognostic classifier, five-PcG signature, served as an independent prognostic marker for patients with glioma.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA