Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 144
Filtrar
1.
Asian Pac J Cancer Prev ; 22(12): 3959-3965, 2021 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-34967577

RESUMEN

OBJECTIVE: BCR ABL oncogene encodes the BCR-ABL chimeric protein, which is a constitutively activated non-receptor tyrosine kinase. The BCR-ABL oncoprotein is a key molecular basis for the pathogenesis of chronic myeloid leukemia (CML) via activation of several downstream signaling pathways including JAK/STAT pathway. Development of leukemia involves constitutive activation of signaling molecules including, JAK2, STAT3, STAT5A and STAT5B. Thymoquinone (TQ) is a bioactive constituent of Nigella sativa that has shown anticancer properties in various cancers. The present study aimed to evaluate the effect of TQ on the expression of BCR ABL, JAK2, STAT3, STAT5A and STAT5B genes and their consequences on the cell proliferation and apoptosis in K562 CML cells. METHODS: BCR-ABL positive K562 CML cells were treated with TQ. Cytotoxicity was determined by Trypan blue exclusion assay. Apoptosis assay was performed by annexin V-FITC/PI staining assay and analyzed by flow cytometry. Transcription levels of BCR ABL, JAK2, STAT3, STAT5A and STAT5B genes were evaluated by reverse transcription-quantitative polymerase chain reaction (RT-qPCR). Protein levels of JAK2 and STAT5 were determined by Jess Assay analysis. RESULTS: TQ markedly decreased the cell proliferation and induced apoptosis in K562 cells (P < 0.001) in a concentration dependent manner. TQ caused a significant decrease in the transcriptional levels of BCR ABL, JAK2, STAT3, STAT5A and STAT5B genes (P < 0.001). TQ induced a significant decrease in JAK2 and STAT5 protein levels (P < 0.001). CONCLUSION: our results indicated that TQ inhibited cell growth of K562 cells via downregulation of BCR ABL/ JAK2/STAT3 and STAT5 signaling and reducing JAK2 and STAT5 protein levels.


Asunto(s)
Antineoplásicos/farmacología , Benzoquinonas/farmacología , Genes abl/efectos de los fármacos , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Apoptosis/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Janus Quinasa 2/efectos de los fármacos , Células K562 , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Factor de Transcripción STAT3/efectos de los fármacos , Factor de Transcripción STAT5/efectos de los fármacos , Transducción de Señal/genética , Proteínas Supresoras de Tumor/efectos de los fármacos
2.
Int J Biol Sci ; 17(14): 3981-3992, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34671213

RESUMEN

Enolase 1 (ENO1) is a moonlighting protein, function as a glycolysis enzyme, a plasminogen receptor and a DNA binding protein. ENO1 play an important role in the process of cancer development. The transcription, translation, post-translational modifying activities and the immunoregulatory role of ENO1 at the cancer development is receiving increasing attention. Some function model studies have shown that ENO1 is a potential target for cancer treatment. In this review, we provide a comprehensive overview of the characterization, function, related transduction cascades of ENO1 and its roles in the pathophysiology of cancers, which is a consequence of ENO1 signaling dysregulation. And the development of novels anticancer agents that targets ENO1 may provide a more attractive option for the treatment of cancers. The data of sarcoma and functional cancer models indicates that ENO1 may become a new potential target for anticancer therapy.


Asunto(s)
Antineoplásicos/farmacología , Proteínas de Unión al ADN/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Fosfopiruvato Hidratasa/efectos de los fármacos , Proteínas Supresoras de Tumor/efectos de los fármacos , Antineoplásicos/uso terapéutico , Biomarcadores de Tumor/química , Biomarcadores de Tumor/metabolismo , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Humanos , Fosfopiruvato Hidratasa/química , Fosfopiruvato Hidratasa/metabolismo , Procesamiento Proteico-Postraduccional , Transducción de Señal , Relación Estructura-Actividad , Proteínas Supresoras de Tumor/química , Proteínas Supresoras de Tumor/metabolismo
3.
Clin Transl Med ; 11(8): e503, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34459131

RESUMEN

Intrinsic resistance to CDK4/6 inhibitors hinders their clinical utility in cancer treatment. Furthermore, the predictive markers of CDK4/6 inhibitors in gastric cancer (GC) remain incompletely described. Here, we found that PAX6 expression was negatively correlated with the response to palbociclib in vitro and in vivo in GC. We observed that the PAX6 expression level was negatively correlated with the overall survival of GC patients and further showed that PAX6 can promote GC cell proliferation and the cell cycle. The cell cycle is regulated by the interaction of cyclins with their partner serine/threonine cyclin-dependent kinases (CDKs), and the G1/S-phase transition is the main target of CDK4/6 inhibitors. Therefore, we tested whether PAX6 expression was correlated with the GC response to palbociclib. We found that PAX6 hypermethylates the promoter of LATS2 and inactivates the Hippo pathway, which upregulates cyclin D1 (CCND1) expression. This results in a suppressed response to palbociclib in GC. Furthermore, we found that the induction of the Hippo signaling pathway or treatment with a DNA methylation inhibitor could overcome PAX6-induced palbociclib resistance in GC. These findings uncover a tumor promoter function of PAX6 in GC and establish overexpressed PAX6 as a mechanism of resistance to palbociclib.


Asunto(s)
Quinasa 4 Dependiente de la Ciclina/efectos de los fármacos , Quinasa 6 Dependiente de la Ciclina/efectos de los fármacos , Vía de Señalización Hippo/efectos de los fármacos , Factor de Transcripción PAX6/efectos de los fármacos , Piperazinas/farmacología , Proteínas Serina-Treonina Quinasas/efectos de los fármacos , Piridinas/farmacología , Neoplasias Gástricas/tratamiento farmacológico , Proteínas Supresoras de Tumor/efectos de los fármacos , Anciano , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , China , Quinasa 4 Dependiente de la Ciclina/genética , Quinasa 6 Dependiente de la Ciclina/genética , Modelos Animales de Enfermedad , Femenino , Vía de Señalización Hippo/genética , Humanos , Masculino , Ratones , Ratones Desnudos , Persona de Mediana Edad , Oncogenes/efectos de los fármacos , Oncogenes/genética , Factor de Transcripción PAX6/genética , Proteínas Serina-Treonina Quinasas/genética , Neoplasias Gástricas/genética , Proteínas Supresoras de Tumor/genética
4.
Molecules ; 26(14)2021 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-34299435

RESUMEN

KRIBB11, an HSF1 inhibitor, was shown to sensitize various types of cancer cells to treatment with several anticancer drugs. However, the exclusive effects of KRIBB11 in preventing the growth of glioblastoma cells and the related mechanisms have not been elucidated yet. Herein, we aimed to examine the potential of KRIBB11 as an anticancer agent for glioblastoma. Using MTT and colony formation assays and Western blotting for c-PARP, we demonstrated that KRIBB11 substantially inhibits the growth of A172 glioma cells by inducing apoptosis. At the molecular level, KRIBB11 decreased anti-apoptotic protein MCL-1 levels, which was attributable to the increase in MULE ubiquitin ligase levels. However, the constitutive activity of HSF1 in A172 cells was not influenced by the exclusive treatment with KRIBB11. Additionally, based on cycloheximide chase assay, we found that KRIBB11 markedly retarded the degradation of MULE. In conclusion, stabilization of MULE upon KRIBB11 treatment is apparently an essential step for degradation of MCL-1 and the subsequent induction of apoptosis in A172 cells. Our results have expanded the knowledge on molecular pathways controlled by KRIBB11 and could be potentially effective for developing an inhibitory therapeutic strategy for glioblastoma.


Asunto(s)
Aminopiridinas/farmacología , Glioblastoma/tratamiento farmacológico , Indazoles/farmacología , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Aminopiridinas/metabolismo , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/metabolismo , Línea Celular Tumoral , Glioblastoma/metabolismo , Glioma/tratamiento farmacológico , Glioma/metabolismo , Humanos , Indazoles/metabolismo , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Supresoras de Tumor/efectos de los fármacos , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina , Ubiquitina-Proteína Ligasas/efectos de los fármacos , Ubiquitina-Proteína Ligasas/metabolismo
5.
BMC Cancer ; 21(1): 493, 2021 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-33941107

RESUMEN

BACKGROUND: Glioblastoma (GBM) is the deadliest and the most common primary brain tumor in adults. The invasiveness and proliferation of GBM cells can be decreased through the inhibition of Wnt/ß-catenin pathway. In this regard, celecoxib is a promising agent, but other COXIBs and 2,5-dimethylcelecoxib (2,5-DMC) await elucidation. Thus, the aim of this study was to analyze the impact of celecoxib, 2,5-DMC, etori-, rofe-, and valdecoxib on GBM cell viability and the activity of Wnt/ß-catenin pathway. In addition, the combination of the compounds with temozolomide (TMZ) was also evaluated. Cell cycle distribution and apoptosis, MGMT methylation level, COX-2 and PGE2 EP4 protein levels were also determined in order to better understand the molecular mechanisms exerted by these compounds and to find out which of them can serve best in GBM therapy. METHODS: Celecoxib, 2,5-DMC, etori-, rofe- and valdecoxib were evaluated using three commercially available and two patient-derived GBM cell lines. Cell viability was analyzed using MTT assay, whereas alterations in MGMT methylation level were determined using MS-HRM method. The impact of COXIBs, in the presence and absence of TMZ, on Wnt pathway was measured on the basis of the expression of ß-catenin target genes. Cell cycle distribution and apoptosis analysis were performed using flow cytometry. COX-2 and PGE2 EP4 receptor expression were evaluated using Western blot analysis. RESULTS: Wnt/ß-catenin pathway was attenuated by COXIBs and 2,5-DMC irrespective of the COX-2 expression profile of the treated cells, their MGMT methylation status, or radio/chemoresistance. Celecoxib and 2,5-DMC were the most cytotoxic. Cell cycle distribution was altered, and apoptosis was induced after the treatment with celecoxib, 2,5-DMC, etori- and valdecoxib in T98G cell line. COXIBs and 2,5-DMC did not influence MGMT methylation status, but inhibited COX-2/PGE2/EP4 pathway. CONCLUSIONS: Not only celecoxib, but also 2,5-DMC, etori-, rofe- and valdecoxib should be further investigated as potential good anti-GBM therapeutics.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Inhibidores de la Ciclooxigenasa 2/farmacología , Glioblastoma/metabolismo , Proteínas de Neoplasias/efectos de los fármacos , Pirazoles/farmacología , Sulfonamidas/farmacología , Vía de Señalización Wnt/efectos de los fármacos , Anciano , Antineoplásicos Alquilantes/farmacología , Apoptosis/efectos de los fármacos , Neoplasias Encefálicas/tratamiento farmacológico , Celecoxib/farmacología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Ciclooxigenasa 2/metabolismo , Metilasas de Modificación del ADN/efectos de los fármacos , Metilasas de Modificación del ADN/metabolismo , Enzimas Reparadoras del ADN/efectos de los fármacos , Enzimas Reparadoras del ADN/metabolismo , Dinoprostona/metabolismo , Relación Dosis-Respuesta a Droga , Etoricoxib/farmacología , Femenino , Glioblastoma/tratamiento farmacológico , Humanos , Isoxazoles/farmacología , Lactonas/farmacología , Masculino , Metilación , Persona de Mediana Edad , Proteínas de Neoplasias/metabolismo , Subtipo EP4 de Receptores de Prostaglandina E/efectos de los fármacos , Subtipo EP4 de Receptores de Prostaglandina E/metabolismo , Sulfonas/farmacología , Temozolomida/farmacología , Proteínas Supresoras de Tumor/efectos de los fármacos , Proteínas Supresoras de Tumor/metabolismo , beta Catenina/efectos de los fármacos , beta Catenina/metabolismo
6.
Arch Insect Biochem Physiol ; 107(1): e21785, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33818826

RESUMEN

Mutant lethal giant larvae (lgl) flies (Drosophila melanogaster) are known to develop epithelial tumors with invasive characteristics. The present study has been conducted to investigate the influence of melatonin (0.025 mM) on behavioral responses of lgl mutant flies as well as on biochemical indices (redox homeostasis, carbohydrate and lipid metabolism, transaminases, and minerals) in hemolymph, and head and intestinal tissues. Behavioral abnormalities were quantitatively observed in lgl flies but were found normalized among melatonin-treated lgl flies. Significantly decreased levels of lipid peroxidation products and antioxidants involved in redox homeostasis were observed in hemolymph and tissues of lgl flies, but had restored close to normalcy in melatonin-treated flies. Carbohydrates including glucose, trehalose, and glycogen were decreased and increased in the hemolymph and tissues of lgl and melatonin-treated lgl flies, respectively. Key enzymes of carbohydrate metabolism showed a significant increment in their levels in lgl mutants but had restored close to wild-type baseline levels in melatonin-treated flies. Variables of lipid metabolism showed significantly inverse levels in hemolymph and tissues of lgl flies, while normalization of most of these variables was observed in melatonin-treated mutants. Lipase, chitinase, transaminases, and alkaline phosphatase showed an increment in their activities and minerals exhibited decrement in lgl flies; reversal of changes was observed under melatonin treatment. The impairment of cognition, disturbance of redox homeostasis and metabolic reprogramming in lgl flies, and restoration of normalcy in all these cellular and behavioral processes indicate that melatonin could act as oncostatic and cytoprotective agents in Drosophila.


Asunto(s)
Drosophila melanogaster/efectos de los fármacos , Melatonina/farmacología , Animales , Antineoplásicos/farmacología , Carbohidratos/sangre , Cognición/efectos de los fármacos , Crioprotectores/farmacología , Proteínas de Drosophila/efectos de los fármacos , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Enzimas/sangre , Homeostasis/efectos de los fármacos , Larva/efectos de los fármacos , Larva/genética , Larva/metabolismo , Lípidos/sangre , Mutación , Oxidación-Reducción/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Proteínas Supresoras de Tumor/sangre , Proteínas Supresoras de Tumor/efectos de los fármacos , Proteínas Supresoras de Tumor/genética
7.
Clin Epigenetics ; 13(1): 35, 2021 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-33588926

RESUMEN

BACKGROUND: Epigenetic inactivation of O6-methylguanine DNA-methyltransferase (MGMT) is associated with increased sensitivity to alkylating chemotherapeutic agents in glioblastoma patients. The genetic background underlying MGMT gene methylation may explain individual differences in treatment response and provide a clue to a personalized treatment strategy. Making use of the longitudinal twin design, we aimed, for the first time, to estimate the genetic contributions to MGMT methylation in a Danish twin cohort. METHODS: DNA-methylation from whole blood (18 monozygotic (MZ) and 25 dizygotic (DZ) twin pairs) repeated 10 years apart from the Longitudinal Study of Aging Danish Twins (LSADT) were used to search for genetic and environmental contributions to DNA-methylation at 170 CpG sites of across the MGMT gene. Both univariate and bivariate twin models were applied. The intraclass correlations, performed on cross-sectional data (246 MZ twin pairs) from an independent study population, the Middle-Aged Danish Twins (MADT), were used to assess the genetic influence at each CpG site of MGMT for replication. RESULTS: Univariate twin model revealed twelve CpG sites showing significantly high heritability at intake (wave 1, h2 > 0.43), and seven CpG sites with significant heritability estimates at end of follow-up (wave 2, h2 > 0.5). There were six significant CpG sites, located at the gene body region, that overlapped among the two waves (h2 > 0.5), of which five remained significant in the bivariate twin model, which was applied to both waves. Within MZ pair correlation in these six CpGs from MADT demarks top level of genetic influence. There were 11 CpGs constantly have substantial common environmental component over the 10 years. CONCLUSIONS: We have identified 6 CpG sites linked to the MGMT gene with strong and persistent genetic control based on their DNA methylation levels. The genetic basis of MGMT gene methylation could help to explain individual differences in glioblastoma treatment response and most importantly, provide references for mapping the methylation Quantitative Trait Loci (meQTL) underlying the genetic regulation.


Asunto(s)
Metilasas de Modificación del ADN/genética , Enzimas Reparadoras del ADN/genética , ADN/metabolismo , Glioblastoma/genética , Guanina/análogos & derivados , Proteínas Supresoras de Tumor/genética , Anciano , Envejecimiento/genética , Antineoplásicos Alquilantes/uso terapéutico , Estudios de Cohortes , Islas de CpG/genética , Estudios Transversales , Metilación de ADN , Metilasas de Modificación del ADN/efectos de los fármacos , Enzimas Reparadoras del ADN/efectos de los fármacos , Dinamarca/epidemiología , Ambiente , Epigenómica , Femenino , Silenciador del Gen/efectos de los fármacos , Glioblastoma/tratamiento farmacológico , Guanina/metabolismo , Humanos , Estudios Longitudinales , Masculino , Metiltransferasas , Persona de Mediana Edad , Proteínas Supresoras de Tumor/efectos de los fármacos , Gemelos Dicigóticos/genética , Gemelos Monocigóticos/genética
8.
CNS Neurosci Ther ; 27(5): 552-563, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33460245

RESUMEN

BACKGROUND: Temozolomide (TMZ) is a first-line chemotherapy drug for the treatment of malignant glioma and resistance to it poses a major challenge. Receptor-interacting protein 2 (RIP2) is associated with the malignant character of cancer cells. However, it remains unclear whether RIP2 is involved in TMZ resistance in glioma. METHODS: RIP2 expression was inhibited in TMZ-resistant glioma cells and normal glioma cells by using small interfering RNA (siRNA) against RIP2. Plasmid transfection method was used to overexpress RIP2. Cell counting kit-8 assays were performed to evaluate cell viability. Western blotting or immunofluorescence was performed to determine RIP2, NF-κB, and MGMT expression in cells. Flow cytometry was used to investigate cell apoptosis. TMZ-resistant glioma xenograft models were established to evaluate the role of the RIP2/NF-κB/MGMT signaling pathway in drug resistance. RESULTS: We observed that RIP2 expression was upregulated in TMZ-resistant glioma cells, whereas silencing of RIP2 expression enhanced cellular sensitivity to TMZ. Similarly, upon the induction of RIP2 overexpression, glioma cells developed resistance to TMZ. The molecular mechanism underlying the process indicated that RIP2 can activate the NF-κB signaling pathway and upregulate the expression of O6-methylguanine-DNA methyltransferase (MGMT), following which the glioma cells develop drug resistance. In the TMZ-resistant glioma xenograft model, treatment with JSH-23 (an NF-κB inhibitor) and lomeguatrib (an MGMT inhibitor) could enhance the sensitivity of the transplanted tumor to TMZ. CONCLUSION: We report that the RIP2/NF-κB/MGMT signaling pathway is involved in the regulation of TMZ resistance. Interference with NF-κB or MGMT activity could constitute a novel strategy for the treatment of RIP2-positive TMZ-resistant glioma.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Metilasas de Modificación del ADN/efectos de los fármacos , Metilasas de Modificación del ADN/genética , Enzimas Reparadoras del ADN/efectos de los fármacos , Enzimas Reparadoras del ADN/genética , Resistencia a Antineoplásicos/genética , Glioma/tratamiento farmacológico , FN-kappa B/efectos de los fármacos , FN-kappa B/genética , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor/efectos de los fármacos , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor/genética , Transducción de Señal/efectos de los fármacos , Temozolomida/farmacología , Proteínas Supresoras de Tumor/efectos de los fármacos , Proteínas Supresoras de Tumor/genética , Animales , Apoptosis/efectos de los fármacos , Neoplasias Encefálicas/genética , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioma/genética , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , FN-kappa B/antagonistas & inhibidores , ARN Interferente Pequeño/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
9.
JCI Insight ; 5(2)2020 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-31996486

RESUMEN

Our integrative genomic and functional analysis identified transforming acidic coiled-coil-containing protein 2 (TACC2) as a chronic obstructive pulmonary disease (COPD) candidate gene. Here, we found that smokers with COPD exhibit a marked decrease in lung TACC2 protein levels relative to smokers without COPD. Single cell RNA sequencing reveals that TACC2 is expressed primarily in lung epithelial cells in normal human lungs. Furthermore, suppression of TACC2 expression impairs the efficiency of homologous recombination repair and augments spontaneous and cigarette smoke extract-induced (CSE-induced) DNA damage and cytotoxicity in immortalized human bronchial epithelial cells. By contrast, enforced expression of TACC2 attenuates the CSE effects. We also found that CSE enhances TACC2 degradation via the ubiquitin-proteasome system mediated by the ubiquitin E3 ligase subunit, F box L7. Furthermore, cellularly expressed TACC2 proteins harboring naturally occurring mutations exhibited altered protein lifespan coupled with modified DNA damage repair and cytotoxic responses. CS triggers emphysematous changes accompanied by accumulated DNA damage, apoptosis of alveolar epithelia, and lung inflammation in Tacc2-/- compared with Tacc2+/+ mice. Our results suggest that CS destabilizes TACC2 protein in lung epithelia by the ubiquitin proteasome system, leading to subsequent DNA damage, cytotoxicity, and emphysema.


Asunto(s)
Enfisema Pulmonar/inducido químicamente , Enfisema Pulmonar/genética , Humo/efectos adversos , Fumar/efectos adversos , Proteínas Supresoras de Tumor/efectos de los fármacos , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Anciano , Animales , Apoptosis/efectos de los fármacos , Proteínas Portadoras , Línea Celular , Daño del ADN/efectos de los fármacos , Reparación del ADN , Enfisema , Células Epiteliales/metabolismo , Femenino , Humanos , Pulmón/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Mutagénesis Sitio-Dirigida , Complejo de la Endopetidasa Proteasomal/metabolismo , Nicotiana/efectos adversos , Transcriptoma , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
10.
Clin Transl Oncol ; 22(7): 1138-1145, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31732915

RESUMEN

PURPOSE: N-myc downstream-regulated gene 2 (NDRG2) is down-regulated in grade-III meningioma [anaplastic meningioma (AM)] and associated with clinically aggressive behavior. Current therapies in the treatment of high-grade meningioma are lacking with limited success. This study aims to validate the effect of NDRG2-targeted therapy using structurally related bioactive triterpene compounds derived from the edible mushroom Ganoderma lucidum (ganoderic acid A:GA-A/ganoderic acid DM:GA-DM) in human AM in relevant pre-clinical models. METHODS: Tissue samples from the AM tumor regions of three human patients and control non-tumor samples were used to analyze the expression pattern of NDRG2. In vitro cell culture and in vivo cell-line-derived orthotopic xenograft animal models of AM were utilized to assess efficacy of treatment with GA-A/DM. RESULTS: Downregulation of NDRG2 expression was observed in surgically resected high-grade meningiomas compared to normal brain. These results prompt us to use NDRG2-targeting agents GA-A/DM. In vitro results showed that 72-h treatments of 25 µM GA-A/DM induced AM cell death, upregulate NDRG2 protein expression, downregulate NDRG2 promoter methylation in meningioma cells as compared to azacitidine and decitabine, the most commonly used demethylating agents. Our results also demonstrated that GA-A/DM does not have any detrimental effect on normal human neurons and arachnoid cells. GA-A/DM promoted apoptotic factors (Bax) while suppressing MMP-9, p-P13K, p-AKT, p-mTOR, and Wnt-2 protein expression. RNAi-mediated knockdown of NDRG2 protein expression increased tumor proliferation, while forced expression of wt-NDRG2 decreased proliferation in an in vitro model. Magnetic resonance (MR) imaging and Hematoxylin (H&E) staining demonstrated gross reduction of tumor volume in GA-A/DM treated mice at 5 weeks when compared with saline-treated orthotopic AM xenografted controls. There was an overall decrease in tumor cell proliferation with increased survival in GA-A/DM-treated animals. Enzyme assays showed that GA-A/DM did not negatively impact hepatic function. CONCLUSION: GA-A/DM may be a promising natural therapeutic reagent in the treatment of AM by suppressing growth via NDRG2 modulation and altering of intracellular signal pathways. We have shown it could potentially be an effective treatment for AM with decreased cellular proliferation in vitro, decreased tumor volume and increased survival in vivo.


Asunto(s)
Ácidos Heptanoicos/farmacología , Lanosterol/análogos & derivados , Neoplasias Meníngeas/metabolismo , Meningioma/metabolismo , Triterpenos/farmacología , Proteínas Supresoras de Tumor/efectos de los fármacos , Anciano , Anaplasia , Animales , Antimetabolitos Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Azacitidina/farmacología , Muerte Celular/efectos de los fármacos , Metilación de ADN/efectos de los fármacos , Decitabina/farmacología , Regulación hacia Abajo , Técnicas de Silenciamiento del Gen , Humanos , Técnicas In Vitro , Lanosterol/farmacología , Metaloproteinasa 9 de la Matriz/efectos de los fármacos , Metaloproteinasa 9 de la Matriz/metabolismo , Neoplasias Meníngeas/patología , Meningioma/patología , Ratones , Ratones SCID , Persona de Mediana Edad , Terapia Molecular Dirigida , Clasificación del Tumor , Fosfatidilinositol 3-Quinasas/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-akt/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Serina-Treonina Quinasas TOR/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proteína wnt2/efectos de los fármacos , Proteína wnt2/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Proteína X Asociada a bcl-2/efectos de los fármacos , Proteína X Asociada a bcl-2/metabolismo
11.
Rev. Hosp. Ital. B. Aires (2004) ; 39(4): 146-148, dic. 2019. ilus
Artículo en Español | LILACS | ID: biblio-1099838

RESUMEN

Los anticuerpos monoclonales que inhiben los puntos de control PD-1 y CTLA-4 se usan actualmente en el tratamiento del melanoma y cáncer metastásico de pulmón de células no pequeñas, entre otros. Se refiere el caso de una paciente con cáncer de pulmón en tratamiento con pembrolizumab. La paciente se presentó con edema facial y parálisis facial periférica. En el laboratorio se observó la hormona tirotrofina (TSH) elevada y se llegó al diagnóstico de hipotiroidismo por pembrolizumab. Inició tratamiento con levotiroxina con mejoría clínica. Se presenta este caso por el importante papel del dermatólogo en el manejo multidisciplinario del paciente oncológico. (AU)


Monoclonal antibodies that inhibit PD-1 and CTLA-4 control points are currently used in the treatment of melanoma and metastatic non-small cell lung cancer, among others. The case of a patient, with lung cancer being treated with Pembrolizumab. The patient was presented with facial edema and peripheral facial paralysis and in the laboratory the elevated hormone Tyrotrophin (TSH) was observed, the diagnosis of pembrolizumab hypothyroidism was reached. She started treatment with levothyroxine with clinical improvement. This case is presented by the important role of the dermatologist in the multidisciplinary management of the cancer patient. (AU)


Asunto(s)
Humanos , Femenino , Persona de Mediana Edad , Puntos de Control de la Fase M del Ciclo Celular/efectos de los fármacos , Inmunoterapia/efectos adversos , Anticuerpos Monoclonales/efectos adversos , Tiroxina/uso terapéutico , Neoplasias Encefálicas/complicaciones , Neoplasias Encefálicas/tratamiento farmacológico , Tirotropina/análisis , Carboplatino/administración & dosificación , Carcinoma de Pulmón de Células no Pequeñas/complicaciones , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Proteínas Supresoras de Tumor/efectos de los fármacos , Dermatología , Traumatismos Faciales , Parálisis Facial , Antígeno CTLA-4/efectos de los fármacos , Antígeno CTLA-4/fisiología , Receptor de Muerte Celular Programada 1/efectos de los fármacos , Receptor de Muerte Celular Programada 1/fisiología , Pemetrexed/administración & dosificación , Melanoma/complicaciones , Melanoma/tratamiento farmacológico , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/uso terapéutico , Metástasis de la Neoplasia/tratamiento farmacológico
12.
Cell Prolif ; 52(6): e12699, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31599032

RESUMEN

OBJECTIVES: The chemotherapy drug resistance is a major challenge for non-small-cell lung cancer (NSCLC) treatment. Combination of immunotherapy and chemotherapy has shown promise for cancer. The goal of this study was to evaluate the anti-tumour efficacy of interleukin-7 (IL-7) combining cisplatin against NSCLC. MATERIALS AND METHODS: Cell proliferation was analysed using CCK-8 assay, EdU proliferation assay and colony-forming assay. Cell apoptosis was evaluated using HOECHST 33342 assay and flow cytometry. The protein expression levels were analysed by Western blot. The blocking antibody against the IL-7 receptor and the inhibitors of STAT5 and JAK3 were used to investigate the pathway involved. A xenograft model was established to assess the anti-tumour efficacy of IL-7 combining cisplatin in vivo. RESULTS: Here we found IL-7R was increased in A549/DDP cells compared with A549 cells. The block of IL-7R reversed the inhibitory effects of IL-7 combined with cisplatin and decreased the numbers of apoptosis cells induced by treatment of IL-7 combined with cisplatin. The JAK3 inhibitor and STAT5 inhibitor were used to identify the pathway involved. The results showed that JAK3/STAT5 pathway was involved in enhancing role of cisplatin sensitivity of NSCLC cells by IL-7. In vivo, cisplatin significantly inhibited tumour growth and IL-7 combined with cisplatin achieved the best therapeutic effect. CONCLUSION: Together, IL-7 promoted the sensitivity of NSCLC cells to cisplatin via IL-7R-JAK3/STAT5 signalling pathway.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Resistencia a Antineoplásicos/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Interleucina-7/farmacología , Células A549/efectos de los fármacos , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Humanos , Janus Quinasa 3/efectos de los fármacos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Factor de Transcripción STAT5/efectos de los fármacos , Proteínas Supresoras de Tumor/efectos de los fármacos
13.
Stroke ; 49(12): 3020-3029, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30571407

RESUMEN

Background and Purpose- Heme and iron are considered to be key factors responsible for secondary insults after intracerebral hemorrhage (ICH). Our previous study showed that LRP1 (low-density lipoprotein receptor-related protein-1)-Hx (hemopexin) facilitates removal of heme. The TLR7 (Toll-like receptor 7)-BTK (Bruton tyrosine kinase)-CRT (calreticulin) pathway regulates the expression of LRP1-Hx. This study is designed to clarify whether TLR7 activation facilitates heme scavenging and to establish the potential role of the BTK-CRT-LRP1-Hx signaling pathway in the pathophysiology of ICH. Methods- ICH was induced by stereotactic, intrastriatal injection of type VII collagenase. Mice received TLR7 agonist (imiquimod) via intraperitoneal injection after ICH induction. TLR7 inhibitor (ODN2088), BTK inhibitor (LFM-A13), and CRT agonist (thapsigargin) were given in different groups to further evaluate the underlying pathway. Mice were randomly divided into sham, ICH+vehicle (normal saline), ICH+Imiquimod (2.5, 5, and 10 µg/g), ICH+ODN2088, ICH+LFM-A13, ICH+thapsigargin, and ICH+ODN2088+thapsigargin. Imiquimod was administered twice daily starting at 6 hours after ICH; ODN2088 was administered by intracerebroventricular injection at 30 minutes, and LFM-A13 or thapsigargin was administered by intraperitoneal injection at 3 hours after ICH induction. Neurological scores, cognitive abilities, as well as brain edema, blood-brain barrier permeability, hemoglobin level, brain expression of TLR7/BTK/CRT/LRP1/Hx were analyzed. Results- Low dosage imiquimod significantly attenuated hematoma volume, brain edema, BBB permeability, and neurological deficits after ICH. Imiquimod also increased protein expressions of TLR7, BTK, CRT, LRP1, and Hx; ODN2088 reduced TLR7, BTK, CRT, LRP1, and Hx expressions. Conclusions- TLR7 plays an important role in heme scavenging after ICH by modulating the BTK-CRT-LRP1-Hx pathway. TLR7 may offer protective effects by promoting heme resolution and reduction of brain edema after ICH.


Asunto(s)
Agammaglobulinemia Tirosina Quinasa/metabolismo , Encéfalo/metabolismo , Calreticulina/metabolismo , Hemorragia Cerebral/metabolismo , Hemo/metabolismo , Hemopexina/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores de LDL/metabolismo , Receptor Toll-Like 7/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Agammaglobulinemia Tirosina Quinasa/antagonistas & inhibidores , Agammaglobulinemia Tirosina Quinasa/efectos de los fármacos , Amidas/farmacología , Animales , Barrera Hematoencefálica/efectos de los fármacos , Encéfalo/efectos de los fármacos , Edema Encefálico/metabolismo , Calreticulina/agonistas , Calreticulina/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Hemopexina/efectos de los fármacos , Imiquimod/farmacología , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad , Glicoproteínas de Membrana/agonistas , Glicoproteínas de Membrana/antagonistas & inhibidores , Glicoproteínas de Membrana/efectos de los fármacos , Ratones , Nitrilos/farmacología , Oligodesoxirribonucleótidos/farmacología , Receptores de LDL/efectos de los fármacos , Transducción de Señal , Tapsigargina/farmacología , Receptor Toll-Like 7/agonistas , Receptor Toll-Like 7/antagonistas & inhibidores , Receptor Toll-Like 7/efectos de los fármacos , Proteínas Supresoras de Tumor/efectos de los fármacos
14.
Biol Res ; 51(1): 22, 2018 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-30012208

RESUMEN

BACKGROUND: Our study aimed to investigate the roles of autophagy against high glucose induced response in retinal pigment epithelium (ARPE-19 cells). METHODS: The morphological changes and reactive oxygen species (ROS) generation in ARPE-19 cells under high glucose treatment were respectively detected using the transmission electron microscopy and flow cytometry. The expression levels of Parkin, PINK1, BNIP3L, LC3-I and LC3-II in ARPE-19 cells received high glucose treatment were measured by western blot after pretreatment of carbonyl cyanide m-chlorophenylhydrazone (CCCP), 3-methyladenine (3-MA), N-acetyl cysteine (NAC) or cyclosporin A (CsA) followed by high glucose treatment. RESULTS: ARPE-19 cells subjected to high glucose stress showed an obvious reduction in the LC3-I expression and significant increase in the number of autophagosomes, in the intracellular ROS level, and in the expression levels of Parkin, PINK1, BNIP3L and LC3-II (p < 0.05). Pretreatment with CCCP significantly reduced the LC3-I expression and increased the expression levels of Parkin, PINK1, BNIP3L and LC3-II (p < 0.05). ARPE-19 cells pretreated with CsA under high glucose stress showed markedly down-regulated expressions of Parkin, PINK1 and BNIP3L compared with the cells treated with high glucose (p < 0.05). Pretreatment of ARPE-19 cells with NAC or 3-MA under high glucose stress resulted in a marked reduction in the expression levels of PINK1, BNIP3L and LC3-II (p < 0.05). Meanwhile, the expression level of Parkin in the ARPE-19 cells pretreated with NAC under high glucose stress was comparable with that in the control cells. CONCLUSION: Autophagy might have protective roles against high glucose induced injury in ARPE19 cells via regulating PINK1/Parkin pathway and BNIP3L.


Asunto(s)
Autofagia/efectos de los fármacos , Glucosa/farmacología , Proteínas de la Membrana/efectos de los fármacos , Proteínas Quinasas/efectos de los fármacos , Proteínas Proto-Oncogénicas/efectos de los fármacos , Epitelio Pigmentado de la Retina/efectos de los fármacos , Proteínas Supresoras de Tumor/efectos de los fármacos , Ubiquitina-Proteína Ligasas/efectos de los fármacos , Autofagia/fisiología , Línea Celular , Citometría de Flujo , Humanos , Proteínas de la Membrana/metabolismo , Microscopía Electrónica de Transmisión , Proteínas Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Epitelio Pigmentado de la Retina/citología , Transducción de Señal/fisiología , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
15.
Brain Behav Immun ; 70: 36-47, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29545118

RESUMEN

Epidemiological evidence suggests that people with bipolar disorder prescribed lithium exhibit a lower risk of Alzheimer's disease (AD) relative to those prescribed other mood-stabilizing medicines. Lithium chloride (LiCl) reduces brain ß-amyloid (Aß) levels, and the brain clearance of Aß is reduced in AD. Therefore, the purpose of this study was to assess whether the cognitive benefits of LiCl are associated with enhanced brain clearance of exogenously-administered Aß. The brain clearance of intracerebroventricularly (icv) administered 125I-Aß42 was assessed in male Swiss outbred mice administered daily oral NaCl or LiCl (300 mg/kg for 21 days). LiCl exhibited a 31% increase in the brain clearance of 125I-Aß42 over 10 min, which was associated with a 1.6-fold increase in brain microvascular expression of the blood-brain barrier efflux transporter low density lipoprotein receptor-related protein 1 (LRP1) and increased cerebrospinal fluid (CSF) bulk-flow. 8-month-old female wild type (WT) and APP/PS1 mice were also administered daily NaCl or LiCl for 21 days, which was followed by cognitive assessment by novel object recognition and water maze, and measurement of soluble Aß42, plaque-associated Aß42, and brain efflux of 125I-Aß42. LiCl treatment restored the long-term spatial memory deficit observed in APP/PS1 mice as assessed by the water maze (back to similar levels of escape latency as WT mice), but the short-term memory deficit remained unaffected by LiCl treatment. While LiCl did not affect plaque-associated Aß42, soluble Aß42 levels were reduced by 49.9% in APP/PS1 mice receiving LiCl. The brain clearance of 125I-Aß42 decreased by 27.8% in APP/PS1 mice, relative to WT mice, however, LiCl treatment restored brain 125I-Aß42 clearance in APP/PS1 mice to a rate similar to that observed in WT mice. These findings suggest that the cognitive benefits and brain Aß42 lowering effects of LiCl are associated with enhanced brain clearance of Aß42, possibly via brain microvascular LRP1 upregulation and increased CSF bulk-flow, identifying a novel mechanism of protection by LiCl for the treatment of AD.


Asunto(s)
Péptidos beta-Amiloides/efectos de los fármacos , Cognición/efectos de los fármacos , Cloruro de Litio/uso terapéutico , Enfermedad de Alzheimer , Precursor de Proteína beta-Amiloide , Animales , Barrera Hematoencefálica/efectos de los fármacos , Encéfalo , Modelos Animales de Enfermedad , Cloruro de Litio/farmacología , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad , Masculino , Memoria/efectos de los fármacos , Ratones , Ratones Transgénicos , Placa Amiloide , Presenilina-1 , Receptores de LDL/efectos de los fármacos , Receptores de LDL/fisiología , Proteínas Supresoras de Tumor/efectos de los fármacos , Proteínas Supresoras de Tumor/fisiología
16.
Biosci Biotechnol Biochem ; 82(1): 57-64, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29191087

RESUMEN

Alzheimer's disease (AD) is the most common cause of dementia among elderly population. Deranged ß-amyloid (Aß) trafficking across the blood-brain barrier is known to be a critical element in the pathogenesis of AD. In the vascular endothelial cells of hippocampus, Aß transport is mainly mediated by low-density lipoprotein-associated protein 1 (LRP1) and the receptor for advanced glycation end (RAGE) products; therefore, LRP1 and RAGE endothelial cells are potential therapeutic targets for AD. In this study, we explored the effects of Formononetin (FMN) on learning and memory improvement in APP/PS1 mice and the related mechanisms. We found that FMN significantly improved learning and memory ability by suppressing Aß production from APP processing, RAGE-dependent inflammatory signaling and promoted LRP1-dependent cerebral Aß clearance pathway. Moreover, FMN treatment alleviated ultrastructural changes in hippocampal vascular endothelial cells. In conclusion, we believe that FMN may be an efficacious and promising treatment for AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Sistemas de Liberación de Medicamentos , Isoflavonas/uso terapéutico , Fármacos Neuroprotectores , Animales , Humanos , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad , Ratones , Fármacos Neuroprotectores/uso terapéutico , Receptor para Productos Finales de Glicación Avanzada/efectos de los fármacos , Receptores de LDL/efectos de los fármacos , Proteínas Supresoras de Tumor/efectos de los fármacos
17.
Biol. Res ; 51: 22, 2018. graf
Artículo en Inglés | LILACS | ID: biblio-950906

RESUMEN

BACKGROUND: Our study aimed to investigate the roles of autophagy against high glucose induced response in retinal pigment epithelium (ARPE-19 cells). METHODS: The morphological changes and reactive oxygen species (ROS) generation in ARPE-19 cells under high glucose treatment were respectively detected using the transmission electron microscopy and flow cytometry. The expression levels of Parkin, PINK1, BNIP3L, LC3-I and LC3-II in ARPE-19 cells received high glucose treatment were measured by western blot after pretreatment of carbonyl cyanide m-chlorophenylhydrazone (CCCP), 3-methyladenine (3-MA), N-acetyl cysteine (NAC) or cyclosporin A (CsA) followed by high glucose treatment. RESULTS: ARPE-19 cells subjected to high glucose stress showed an obvious reduction in the LC3-I expression and significant increase in the number of autophagosomes, in the intracellular ROS level, and in the expression levels of Parkin, PINK1, BNIP3L and LC3-II (p < 0.05). Pretreatment with CCCP significantly reduced the LC3-I expression and increased the expression levels of Parkin, PINK1, BNIP3L and LC3-II (p < 0.05). ARPE-19 cells pretreated with CsA under high glucose stress showed markedly down-regulated expressions of Parkin, PINK1 and BNIP3L compared with the cells treated with high glucose (p < 0.05). Pretreatment of ARPE-19 cells with NAC or 3-MA under high glucose stress resulted in a marked reduction in the expression levels of PINK1, BNIP3L and LC3-II (p < 0.05). Meanwhile, the expression level of Parkin in the ARPE-19 cells pretreated with NAC under high glucose stress was comparable with that in the control cells. CONCLUSION: Autophagy might have protective roles against high glucose induced injury in ARPE19 cells via regulating PINK1/Parkin pathway and BNIP3L.


Asunto(s)
Humanos , Proteínas Quinasas/efectos de los fármacos , Autofagia/efectos de los fármacos , Proteínas Proto-Oncogénicas/efectos de los fármacos , Proteínas Supresoras de Tumor/efectos de los fármacos , Ubiquitina-Proteína Ligasas/efectos de los fármacos , Epitelio Pigmentado de la Retina/efectos de los fármacos , Glucosa/farmacología , Proteínas de la Membrana/efectos de los fármacos , Proteínas Quinasas/metabolismo , Autofagia/fisiología , Transducción de Señal/fisiología , Línea Celular , Proteínas Proto-Oncogénicas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Microscopía Electrónica de Transmisión , Epitelio Pigmentado de la Retina/citología , Citometría de Flujo , Proteínas de la Membrana/metabolismo
18.
Toxicol Appl Pharmacol ; 330: 30-39, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28688920

RESUMEN

Gene 33 (Mig6, ERRFI1) is an adaptor protein with multiple cellular functions. We recently reported that depletion of this protein promotes lung epithelial cell transformation induced by hexavalent chromium [Cr(VI)]. However, the early molecular events that mediate this process are not clear. In the present study, we used single-cell RNA sequencing to compare gene expression profiles between BEAS-2B lung epithelial cells chronically exposed to a sublethal dose of Cr(VI) with or without CRISPR/cas9-mediated deletion of Gene 33. Our data reveal 83 differentially expressed genes. The most notable changes are genes associated with cell adhesion, oxidative stresses, protein ubiquitination, epithelial-mesenchymal transition/metastasis, and WNT signaling. Up-regulation of some neuro-specific genes is also evident, particularly ubiquitin carboxyl-terminal hydrolase L1 (UCHL1), a deubiquitinase and potential biomarker for lung cancer. Gene 33 deletion and/or Cr(VI) exposure did not cause discernable changes in cell morphology. However, Gene 33 deletion led to a modest but significant reduction of cells in the G2/M phase of the cell cycle regardless of Cr(VI) exposure. Gene 33 deletion also significantly reduced cell proliferation. Interestingly, Cr(VI) exposure eliminated the difference in cell proliferation between the two genotypes. Gene 33 deletion also significantly elevated cell migration. Our data indicate that combined Gene 33 deletion and chronic Cr(VI) exposure produces a gene expression pattern and a phenotype resemble those of the transformed lung epithelial cells. Given the known association of UCHL1 with lung cancer, we propose that UCHL1 is an important player in the early stage of lung epithelial cell transformation and tumorigenesis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Sistemas CRISPR-Cas/genética , Carcinógenos/toxicidad , Cromo/toxicidad , Contaminantes Ambientales/toxicidad , Expresión Génica/efectos de los fármacos , Expresión Génica/genética , ARN/química , Proteínas Supresoras de Tumor/genética , Proteínas Adaptadoras Transductoras de Señales/efectos de los fármacos , Sistemas CRISPR-Cas/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica , Humanos , ARN/efectos de los fármacos , Análisis de Secuencia de ARN , Proteínas Supresoras de Tumor/efectos de los fármacos
19.
Neuro Oncol ; 19(8): 1119-1126, 2017 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-28371907

RESUMEN

BACKGROUND: Radiation with concurrent and adjuvant (6 cycles) temozolomide (TMZ) is the established standard of postsurgical care for newly diagnosed glioblastoma (GBM). This regimen has been adopted with variations, including extending TMZ beyond 6 cycles. The optimal duration of maintenance therapy remains controversial. METHODS: We performed pooled analysis of individual patient data from 4 randomized trials for newly diagnosed GBM. All patients who were progression free 28 days after cycle 6 were included. The decision to continue TMZ was per local practice and standards, and at the discretion of the treating physician. Patients were grouped into those treated with 6 cycles and those who continued beyond 6 cycles. Progression-free and overall survival were compared, adjusted by age, performance status, resection extent, and MGMT methylation. RESULTS: A total of 2214 GBM patients were included in the 4 trials. Of these, 624 qualified for analysis 291 continued maintenance TMZ until progression or up to 12 cycles, while 333 discontinued TMZ after 6 cycles. Adjusted for prognostic factors, treatment with more than 6 cycles of TMZ was associated with a somewhat improved progression-free survival (hazard ratio [HR] 0.80 [0.65-0.98], P = .03), in particular for patients with methylated MGMT (n = 342, HR 0.65 [0.50-0.85], P < .01). However, overall survival was not affected by the number of TMZ cycles (HR = 0.92 [0.71-1.19], P = .52), including the MGMT methylated subgroup (HR = 0.89 [0.63-1.26], P = .51). CONCLUSIONS: Continuing TMZ beyond 6 cycles was not shown to increase overall survival for newly diagnosed GBM.


Asunto(s)
Antineoplásicos Alquilantes/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Dacarbazina/análogos & derivados , Glioblastoma/tratamiento farmacológico , Adulto , Anciano , Anciano de 80 o más Años , Terapia Combinada/métodos , Dacarbazina/uso terapéutico , Progresión de la Enfermedad , Supervivencia sin Enfermedad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Temozolomida , Proteínas Supresoras de Tumor/efectos de los fármacos
20.
Antiviral Res ; 142: 1-11, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28286234

RESUMEN

Viral infection induces translocation of the nucleolar protein GLTSCR2 from the nucleus to the cytoplasm, resulting in attenuation of the type I interferon IFN-ß. Addressing the role of GLTSCR2 in viral replication, we detect that knocking down GLTSCR2 by shRNAs results in significant suppression of viral replication in mammalian and chicken cells. Injection of chicken embryo with the GLTSCR2-specific shRNA-1370 simultaneously or 24 h prior to infection with Newcastle disease virus (NDV) substantially reduces viral replication in chicken embryo fibroblasts. Injection of shRNA-1370 into chicken embryo also reduces the replication of avian influenza virus (AIV). In contrast, GLTSCR2-derived protein G4-T, forming α-helical dimers, increases replication of seven various DNA and RNA viruses in cells. Our studies reveal that alteration of the function of cellular GLTSCR2 plays a role in supporting viral replication. GLTSCR2 should be seriously considered as a therapeutic target for developing broad spectrum antiviral agents to effectively control viral infection.


Asunto(s)
Antivirales/farmacología , Proteínas Supresoras de Tumor/efectos de los fármacos , Proteínas Supresoras de Tumor/fisiología , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/farmacología , Animales , Línea Celular , Embrión de Pollo , Chlorocebus aethiops , Virus ADN/efectos de los fármacos , Perros , Fibroblastos/virología , Técnicas de Silenciamiento del Gen , Células HEK293 , Células HeLa , Humanos , Virus de la Influenza A/efectos de los fármacos , Virus de la Influenza A/genética , Interferón Tipo I/metabolismo , Interferón-alfa/metabolismo , Células de Riñón Canino Madin Darby , Enfermedad de Newcastle/virología , Virus de la Enfermedad de Newcastle/efectos de los fármacos , Virus de la Enfermedad de Newcastle/genética , Virus de la Enfermedad de Newcastle/fisiología , Conformación Proteica en Hélice alfa/efectos de los fármacos , Virus ARN/efectos de los fármacos , ARN Interferente Pequeño/genética , Proteínas Recombinantes , Proteínas Supresoras de Tumor/genética , Células Vero , Replicación Viral/efectos de los fármacos , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA