Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 449
Filtrar
1.
Biomed Res Int ; 2022: 1948657, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35141331

RESUMEN

OBJECTIVE: Studies have unveiled that the components of Tripterygium wilfordii Hook F (TWHF) such as celastrol could attenuate apoptosis and proliferation of various tumor cells. This study is focused on the radiosensitization effect and apoptotic pathways of celastrol via the inhibition of the c-myc gene and the influence of which combined with radiotherapy on the proliferation, apoptosis, invasion, and metastasis of chondrosarcoma cells. METHODS: A variety of bioinformatic tools were applied to explore the expression level and prognosis of the c-myc gene in different tumor cells and chondrosarcoma cells. We used pharmacology network to analyze the components, pathways, targets, molecular functions of TWHF and explore the relevant effective components over the MYC gene. Clone formation assay, CCK-8 assay, flow cytometry, and transwell migration assay were applied to detect the effects of celastrol on the expression of c-myc gene, cell apoptosis, and cell cycle. Radiation therapy was used to observe the radiosensitization effect of celastrol on chondrosarcoma. RESULTS: This study shows that the c-myc gene is overexpressed in various tumor cells and bone tumor cells to varying degrees. Celastrol can significantly inhibit the expression of the c-myc gene, induce G2/M phase arrest through regulation of G2/M phase-related proteins, and promote SW1353 cell apoptosis through the mitochondrial signaling pathway. In addition, we also found that the use of triptorubin to inhibit c-myc gene expression in combination with radiotherapy can increase the osteosarcoma cells' apoptosis rate through the mitochondrial signaling pathway significantly. CONCLUSIONS: Our study validated the radiosensitization effect of celastrol through knocking down the expression of the c-myc gene to induce G2/M phase arrest and provides a new idea for the treatment of refractory or recurrent chondrosarcoma that is not sensitive to radiotherapy.


Asunto(s)
Puntos de Control del Ciclo Celular/efectos de los fármacos , Condrosarcoma/tratamiento farmacológico , Genes myc/efectos de los fármacos , Triterpenos Pentacíclicos/farmacología , Fármacos Sensibilizantes a Radiaciones/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Biología Computacional , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Transducción de Señal
2.
Clin Transl Med ; 12(1): e703, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35073459

RESUMEN

BACKGROUND: Chemoresistance to cisplatin (DDP) remains a major challenge in advanced gastric cancer (GC) treatment. Although accumulating evidence suggests an association between dysregulation of long non-coding RNAs (lncRNAs) and chemoresistance, the regulatory functions and complexities of lncRNAs in modulating DDP-based chemotherapy in GC remain under-investigated. This study was designed to explore the critical chemoresistance-related lncRNAs in GC and identify novel therapeutic targets for patients with chemoresistant GC. METHODS: Chemoresistance-related lncRNAs were identified through microarray and verified through a quantitative real-time polymerase chain reaction (qRT-PCR). Proteins bound by lncRNAs were identified through a human proteome array and validated through RNA immunoprecipitation (RIP) and RNA pull-down assays. Co-immunoprecipitation and ubiquitination assays were performed to explore the molecular mechanisms of the Musashi2 (MSI2) post-modification. The effects of LINC00942 (LNC942) and MSI2 on DDP-based chemotherapy were investigated through MTS, apoptosis assays and xenograft tumour formation in vivo. RESULTS: LNC942 was found to be up-regulated in chemoresistant GC cells, and its high expression was positively correlated with the poor prognosis of patients with GC. Functional studies indicated that LNC942 confers chemoresistance to GC cells by impairing apoptosis and inducing stemness. Mechanically, LNC942 up-regulated the MSI2 expression by preventing its interaction with SCFß-TRCP E3 ubiquitin ligase, eventually inhibiting ubiquitination. Then, LNC942 stabilized c-Myc mRNA in an N6-methyladenosine (m6 A)-dependent manner. As a potential m6 A recognition protein, MSI2 stabilized c-Myc mRNA with m6 A modifications. Moreover, inhibition of the LNC942-MSI2-c-Myc axis was found to restore chemosensitivity both in vitro and in vivo. CONCLUSIONS: These results uncover a chemoresistant accelerating function of LNC942 in GC, and disrupting the LNC942-MSI2-c-Myc axis could be a novel therapeutic strategy for GC patients undergoing chemoresistance.


Asunto(s)
Cisplatino/metabolismo , Resistencia a Medicamentos/efectos de los fármacos , Genes myc/efectos de los fármacos , ARN Largo no Codificante/agonistas , Proteínas de Unión al ARN/antagonistas & inhibidores , Cisplatino/uso terapéutico , Genes myc/fisiología , Humanos , ARN Largo no Codificante/genética , ARN Largo no Codificante/uso terapéutico , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/farmacología , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/genética
3.
Sci Rep ; 12(1): 855, 2022 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-35039581

RESUMEN

The proto-oncogene MYC is important for development and cell growth, however, its abnormal regulation causes cancer. Recent studies identified distinct enhancers of MYC in various cancers, but any MYC enhancer(s) in hepatocellular carcinoma (HCC) remain(s) elusive. By analyzing H3K27ac enrichment and enhancer RNA (eRNA) expression in cultured HCC cells, we identified six putative MYC enhancer regions. Amongst these, two highly active enhancers, located ~ 800 kb downstream of the MYC gene, were identified by qRT-PCR and reporter assays. We functionally confirmed these enhancers by demonstrating a significantly reduced MYC expression and cell proliferation upon CRISPR/Cas9-based deletion and/or antisense oligonucleotide (ASO)-mediated inhibition. In conclusion, we identified potential MYC enhancers of HCC and propose that the associated eRNAs may be suitable targets for HCC treatment.


Asunto(s)
Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Proliferación Celular/genética , Elementos de Facilitación Genéticos/genética , Elementos de Facilitación Genéticos/fisiología , Genes myc/efectos de los fármacos , Genes myc/fisiología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , ARN no Traducido , Línea Celular Tumoral , Humanos
4.
Int J Oncol ; 60(1)2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34970694

RESUMEN

Methyltransferase N6­adenosine (METTL5) is a methyltransferase that specifically catalyzes 18S rRNA N6 methylation at adenosine 1832 (m6A1832), which is located in a critical position in the decoding center, therefore suggesting its potential importance in the regulation of translation. However, the underlying mechanism of METTL5­mediated translation regulation of specific genes and its biological functions are largely undefined. To the best of our knowledge, the present study demonstrated for the first time that METTL5 was an oncogene that promoted cell proliferation, migration, invasion and tumorigenesis in pancreatic cancer. In addition, the oncogenic function of METTL5 may involve an increase in c­Myc translation, as evidenced by the fact that the oncogenic effect caused by METTL5 overexpression could be abolished by c­Myc knockdown. Notably, m6A modifications at the 5' untranslated region (5'UTR) and coding DNA sequence region (near the 5'UTR) of c­Myc mRNA played a critical role in the specific translation regulation by METTL5. In addition, it was further demonstrated that METTL5 and its cofactor tRNA methyltransferase activator subunit 11­2 synergistically promote pancreatic cancer progression. These findings revealed important roles for METTL5 in the development of pancreatic cancer and present the METTL5/c­Myc axis as a novel therapeutic strategy for treatment.


Asunto(s)
Genes myc/efectos de los fármacos , Metiltransferasas/efectos adversos , Neoplasias Pancreáticas/genética , ARN Ribosómico 18S/efectos adversos , Carcinogénesis/genética , Progresión de la Enfermedad , Genes myc/genética , Humanos , Metiltransferasas/metabolismo , ARN Ribosómico 18S/metabolismo
5.
Clin Transl Med ; 11(12): e674, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34954904

RESUMEN

BACKGROUND AND PURPOSE: The aim of this study is to decipher the underlying mechanisms of CCAAT/enhancer-binding protein delta (CEBPD)-enhanced glycolysis as well as the biological significance of CEBPD and MYC coamplification in urothelial carcinoma (UC). METHODS: In vitro analyses were conducted to examine the effects of altered CEBPD or MYC expression on UC cells. The in vivo effects of CEBPD overexpression in a high-glucose environment on tumour growth were investigated in xenografted induced diabetic severe combined immunodeficiency/beige mice. Data mining was used to cross-validate the associations between CEBPD and MYC copy number and transcriptional expression, quantitative reverse transcription-polymerase chain reaction, immunohistochemistry, chromogenic in situ hybridization, and in situ hybridization targeting microRNA were performed on 635 UC patient samples and xenograft samples. UC patient survival in relation to diabetes was validated by using the National Health Insurance Research Database. RESULTS: CEBPD and MYC coamplification (29.6%) occurred at a high frequency, MYC expression promoted chromosomal instability, facilitating CEBPD copy number gain and expression. CEBPD promoted glucose uptake and lactate production by upregulating SLC2A1 and HK2, leading to mitochondrial fission, increased extracellular acidification rate and decreased oxygen consumption rate to fuel cell growth. CEBPD upregulated HK2 expression through multiple regulation pathways including MYC stabilization, suppression of FBXW7 transactivation and MYC-independent transcriptional suppression of hsa-miR-429. Clinical and xenografted experiments confirmed the growth advantage of CEBPD in relation to glucose metabolic dysregulation and the significant correlations between the expression of these genes. CONCLUSIONS: We confirmed that CEBPD has an oncogenic role in UC by activating AKT signalling and initiating metabolic reprogramming from mitochondrial oxidative phosphorylation to glycolysis to satisfy glucose addiction. These novel CEBPD- and MYC-centric multilayered positive feedback loops enhance cancer growth that could complement theranostic approaches.


Asunto(s)
Proteína delta de Unión al Potenciador CCAAT/efectos de los fármacos , Genes myc/efectos de los fármacos , Neoplasias de la Vejiga Urinaria/genética , Urotelio/fisiopatología , Animales , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Retroalimentación Fisiológica/efectos de los fármacos , Retroalimentación Fisiológica/fisiología , Ratones , Urotelio/anomalías , Efecto Warburg en Oncología
6.
BMC Cancer ; 21(1): 1112, 2021 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-34657603

RESUMEN

BACKGROUND: Tumor cells exhibit enhanced metabolism of nutrients to satisfy the demand of sustained proliferation in vivo. Seminal reports have presented evidence that tryptophan (Trp) metabolic reprogramming induced by aberrant indoleamine 2,3-dioxygenases could promote tumor development in several cancer types. However, the underlying mechanism of Trp metabolism associated tumor progression is not fully understood. MATERIALS AND METHODS: Prostatic cell lines LNCaP and VCaP were purchased from the Cell Bank of the Chinese Academy of Sciences (China). Human prostatic tumor tissue samples were obtained from the Tongji Hospital. Female NOD-SCID mice (6 ~ 8 weeks) were purchased from Huafukang Co. (China) and raised in SPF room. Commercial kits and instruments were used for cell apoptosis analysis, real-time PCR, western blotting, ELISA analysis and other experiments. RESULT: Comparing the tumor tissues from prostatic cancer patients, we found elevated expression of tryptophan 2, 3-dioxygenase 2 (TDO2), and elevated Trp metabolism in chemo-resistant tumor tissues. In vitro, overexpression of TDO2 significantly promoted the Trp metabolism in prostatic cancer cell lines LNCaP and VCap, resulting in the multidrug resistance development. Mechanistically, we demonstrated that Trp metabolite kynurenine (Kyn) promoted the upregulation and nuclear translocation of transcription factor aryl hydrocarbon receptor (AhR). Subsequently, AhR collaborated with NF-κB to facilitate the activation of c-Myc. In turn, c-Myc promoted the up-regulation of ATP-binding cassette (ABC) transporters and Trp transporters, thereby contributing to chemoresistance and strengthened Trp metabolism in prostatic cancer. Interrupt of Trp/TDO2/Kyn/AhR/c-Myc loop with c-Myc inhibitor Mycro-3 efficiently suppressed the chemoresistance and improved the outcome of chemotherapy, which described a new strategy in clinical prostatic cancer treatment. CONCLUSION: Our study demonstrates that elevated TOD2 expression promoted Trp metabolism and metabolite Kyn production, thus resulting in the activation of AhR/c-Myc/ABC-SLC transporters signaling pathway. Interrupt of Trp metabolism/c-Myc loop efficiently suppressed the drugs resistance induced by TDO2, which represented potential target to improve the outcome in drug-resistant prostatic cancer treatment.


Asunto(s)
Resistencia a Antineoplásicos , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Neoplasias de la Próstata/tratamiento farmacológico , Triptófano/metabolismo , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Línea Celular Tumoral , Núcleo Celular/metabolismo , Progresión de la Enfermedad , Resistencia a Múltiples Medicamentos , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Expresión Génica , Genes myc/efectos de los fármacos , Humanos , Quinurenina/metabolismo , Masculino , Ratones Endogámicos NOD , Ratones SCID , FN-kappa B/metabolismo , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Receptores de Hidrocarburo de Aril/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Activación Transcripcional , Regulación hacia Arriba
7.
J Hematol Oncol ; 14(1): 121, 2021 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-34372899

RESUMEN

MYC oncogene is a transcription factor with a wide array of functions affecting cellular activities such as cell cycle, apoptosis, DNA damage response, and hematopoiesis. Due to the multi-functionality of MYC, its expression is regulated at multiple levels. Deregulation of this oncogene can give rise to a variety of cancers. In this review, MYC regulation and the mechanisms by which MYC adjusts cellular functions and its implication in hematologic malignancies are summarized. Further, we also discuss potential inhibitors of MYC that could be beneficial for treating hematologic malignancies.


Asunto(s)
Genes myc , Neoplasias Hematológicas/genética , Proteínas Proto-Oncogénicas c-myc/genética , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Descubrimiento de Drogas , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genes myc/efectos de los fármacos , Neoplasias Hematológicas/diagnóstico , Neoplasias Hematológicas/tratamiento farmacológico , Neoplasias Hematológicas/metabolismo , Humanos , Modelos Moleculares , Pronóstico , Proteínas Proto-Oncogénicas c-myc/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-myc/química , Proteínas Proto-Oncogénicas c-myc/metabolismo
8.
Methods Mol Biol ; 2318: 337-346, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34019301

RESUMEN

Oncoproteins encoded by dominant oncogenes have long been considered as targets for chemotherapeutic intervention. However, oncogenic transcription factors have often been dismissed as "undruggable." Members of the Myc family of transcription factors have been identified as promising targets for cancer chemotherapy in multiple publications reporting the requirement of Myc proteins for maintenance of almost every type of tumor. Here, we describe cell-based approaches to identify c-Myc small molecule inhibitors by screening complex libraries of diverse small molecules based on Myc functionality and specificity.


Asunto(s)
Ensayos de Selección de Medicamentos Antitumorales/métodos , Genes myc/efectos de los fármacos , Proteínas Proto-Oncogénicas c-myc/antagonistas & inhibidores , Línea Celular Tumoral , Genes myc/genética , Genes myc/fisiología , Humanos , Proteínas Oncogénicas/efectos de los fármacos , Proteínas Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Factores de Transcripción/efectos de los fármacos , Factores de Transcripción/metabolismo
9.
Biomed Pharmacother ; 139: 111545, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33873145

RESUMEN

Tacrolimus (TAC, FK506) is a major calcineurin inhibitor and has been commonly used in treatments of patients with organ transplants and immune diseases. Moreover, tacrolimus is recommended by the treatment guidelines for oral potentially malignant disorders (OPMDs) such as oral lichen planus (OLP). However, whether tacrolimus increases the risk of cancer remains controversial. We observed that in a 4-Nitroquinoline N-oxide (4NQO)-induced oral carcinogenesis model, tacrolimus treatment was associated with a significantly lower ratio of cancer formation (52.94% vs. 90%) and a lower proportion of Ki67 and proliferation cell nuclear antigen (PCNA) -positive cells in lesion areas (P < 0.001). Liver, kidney, and lung functions of rats and the tumor immune microenvironment of the tongue were not affected. These observations suggest that tacrolimus blocked oral carcinogenesis through epithelial cell proliferation inhibition, independent of its immunosuppressive effects. As a processing factor, tacrolimus decreased tumor formation and cell proliferation in different stages of oral squamous cell carcinoma (OSCC) progression in vivo and in vitro. Furthermore, we investigated effects on the cell cycle and expression of related proteins. Tacrolimus induced G1/S phase arrest and significantly downregulated the expression of cyclinD1, cyclinE1, and c-Myc. These results suggest that tacrolimus induces G1/S phase arrest via inhibition of cyclinD1, cyclinE1, and c-Myc expression and retards oral cell carcinogenesis in vitro and in vivo. Thus, application of tacrolimus is a safe therapeutic strategy for treating OPMDs.


Asunto(s)
Anticarcinógenos/farmacología , Ciclo Celular/efectos de los fármacos , Neoplasias de la Boca/prevención & control , Tacrolimus/farmacología , 4-Nitroquinolina-1-Óxido , Animales , Carcinógenos , Microambiente Celular/efectos de los fármacos , Ciclinas/antagonistas & inhibidores , Ciclinas/biosíntesis , Genes myc/efectos de los fármacos , Antígeno Ki-67 , Masculino , Ratones , Ratones Endogámicos BALB C , Neoplasias de la Boca/inducido químicamente , Neoplasias de la Boca/patología , Antígeno Nuclear de Célula en Proliferación/análisis , Ratas , Ratas Sprague-Dawley , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Carcinoma de Células Escamosas de Cabeza y Cuello/prevención & control , Ensayos Antitumor por Modelo de Xenoinjerto
10.
ChemMedChem ; 16(10): 1667-1679, 2021 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-33508167

RESUMEN

Lead-optimization strategies for compounds targeting c-Myc G-quadruplex (G4) DNA are being pursued to develop anticancer drugs. Here, we investigate the structure-activity- relationship (SAR) of a newly synthesized series of molecules based on the pyrrolidine-substituted 5-nitro indole scaffold to target G4 DNA. Our synthesized series allows modulation of flexible elements with a structurally preserved scaffold. Biological and biophysical analyses illustrate that substituted 5-nitroindole scaffolds bind to the c-Myc promoter G-quadruplex. These compounds downregulate c-Myc expression and induce cell-cycle arrest in the sub-G1/G1 phase in cancer cells. They further increase the concentration of intracellular reactive oxygen species. NMR spectra show that three of the newly synthesized compounds interact with the terminal G-quartets (5'- and 3'-ends) in a 2 : 1 stoichiometry.


Asunto(s)
Antineoplásicos/farmacología , G-Cuádruplex/efectos de los fármacos , Genes myc/efectos de los fármacos , Indoles/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Células HeLa , Humanos , Indoles/síntesis química , Indoles/química , Estructura Molecular , Relación Estructura-Actividad , Células Tumorales Cultivadas
11.
Invest New Drugs ; 39(2): 587-594, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33052557

RESUMEN

Background The MYC oncogene is one of the most frequently altered driver genes in cancer. MYC is thus a potential target for cancer treatment as well as a biomarker for the disease. However, as a target for treatment, MYC has traditionally been regarded as "undruggable" or difficult to target. We set out to evaluate the efficacy of a novel MYC inhibitor known as MYCMI-6, which acts by preventing MYC from interacting with its cognate partner MAX. Methods MYCMI-6 response was assessed in a panel of breast cancer cell lines using MTT assays and flow cytometry. MYC gene amplification, mRNA and protein expression was analysed using the TCGA and METABRIC databases. Results MYCMI-6 inhibited cell growth in breast cancer cell lines with IC50 values varying form 0.3 µM to >10 µM. Consistent with its ability to decrease cell growth, MYCMI-6 was found to induce apoptosis in two cell lines in which growth was inhibited but not in two cell lines that were resistant to growth inhibition. Across all breast cancers, MYC was found to be amplified in 15.3% of cases in the TCGA database and 26% in the METABRIC database. Following classification of the breast cancers by their molecular subtypes, MYC was most frequently amplified and exhibited highest expression at both mRNA and protein level in the basal subtype. Conclusions Based on these findings, we conclude that for patients with breast cancer, anti-MYC therapy is likely to be most efficacious in patients with the basal subtype.


Asunto(s)
Acridinas/farmacología , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/patología , Proliferación Celular/efectos de los fármacos , Genes myc/efectos de los fármacos , Piridinas/farmacología , Biomarcadores de Tumor , Ciclo Celular , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Regulación Neoplásica de la Expresión Génica , Humanos , Concentración 50 Inhibidora , Peso Molecular , ARN Mensajero
12.
Neuroendocrinology ; 111(8): 739-751, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32615570

RESUMEN

INTRODUCTION: Dysregulation of the mechanistic target of rapamycin complex 1 (mTORC1)-dependent pathways in pancreatic neuroendocrine neoplasms (PanNENs) underlies the introduction of the mTORC1 inhibitor everolimus as treatment of advanced progressive PanNENs. Although everolimus significantly increases progression-free survival, most patients acquire secondary resistance to the drug. This study aimed at identifying mechanisms involved in acquisition of resistance to everolimus. METHODS: BON-1 and everolimus-resistant (ER) BON-1 cells were used as in vitro system of sensitivity and acquired resistance. Transcriptome changes occurring in BON-1 and ER-BON-1 were investigated by RNA sequencing and validated by quantitative PCR analysis. RNA extracted from patients' biopsies was used to validate MYC upregulation. Drug screening and functional assays were performed using ER-BON-1 cells. Cell cycle progression was evaluated by FACS analysis. RESULTS: Our results show that MYC overexpression is a key event in the development of secondary resistance to everolimus in PanNEN cell lines and in metastatic lesions from neuroendocrine neoplasm patients. MYC knockdown restored ER-BON-1 sensitivity to everolimus. Pharmacological inhibition of MYC mediated by the cyclin-dependent kinase inhibitor dinaciclib strongly reduced viability of ER-BON-1. Dinaciclib synergized with everolimus and inhibited ER-BON-1 cell cycle progression. DISCUSSION: Our findings suggest that MYC upregulation drives the development of secondary resistance to everolimus in PanNENs and that its inhibition is an exploitable vulnerability. Indeed, our results indicate that combined treatments with cyclin-dependent kinase and mTOR inhibitors may counteract secondary resistance to everolimus in PanNENs and may pave the ground for new therapeutic regimens for these tumors.


Asunto(s)
Antineoplásicos/farmacología , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Everolimus/farmacología , Genes myc/efectos de los fármacos , Tumores Neuroendocrinos/tratamiento farmacológico , Neoplasias Pancreáticas/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Línea Celular Tumoral/efectos de los fármacos , Humanos , Regulación hacia Arriba
13.
Medicine (Baltimore) ; 99(23): e20181, 2020 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-32501970

RESUMEN

BACKGROUND: This study will examine the effects of oxymatrine on the proliferation of human liver cancer Bel-7404 cells (HLCBC). METHODS: This study will search electronic bibliographic databases available in PUBMED, EMBASE, Cochrane Library, Scopus, Cumulative Index to Nursing and Allied Health Literature, China Biology Medicine, and China National Knowledge Infrastructure. We attempt to search case-controlled studies (CCSs) or randomized controlled studies (RCSs) pertaining to HLCBC from their inception to the February 29, 2020 without limitations of language and publication time. We will include any CCSs or RCSs on exploring oxymatrine on the proliferation of HLCBC. We will assess the methodological quality of CCSs by Newcastle-Ottawa Scale, and RCSs by Cochrane risk of bias tool. Review Manager 5.3 software will be utilized for statistical analysis. RESULTS: The current study will summarize most recent eligible studies to investigate the effects of oxymatrine on the proliferation of HLCBC. CONCLUSION: Its results may provide reliable scientific evidence on effects of oxymatrine on the proliferation of HLCBC. SYSTEMATIC REVIEW REGISTRATION: INPLASY202040026.


Asunto(s)
Alcaloides/farmacología , Neoplasias Hepáticas/tratamiento farmacológico , Quinolizinas/farmacología , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Factor de Transcripción E2F1/biosíntesis , Expresión Génica , Genes myc/efectos de los fármacos , Humanos , Reacción en Cadena en Tiempo Real de la Polimerasa , Proyectos de Investigación , Metaanálisis como Asunto
14.
Nat Commun ; 11(1): 740, 2020 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-32029739

RESUMEN

Primary and acquired drug resistance imposes a major threat to achieving optimized clinical outcomes during cancer treatment. Aberrant changes in epigenetic modifications are closely involved in drug resistance of tumor cells. Using BET inhibitor (BETi) resistant leukemia cells as a model system, we demonstrated herein that genome-wide enhancer remodeling played a pivotal role in driving therapeutic resistance via compensational re-expression of pro-survival genes. Capitalizing on the CRISPR interference technology, we identified the second intron of IncRNA, PVT1, as a unique bona fide gained enhancer that restored MYC transcription independent of BRD4 recruitment in leukemia. A combined BETi and CDK7 inhibitor treatment abolished MYC transcription by impeding RNAPII loading without affecting PVT1-mediated chromatin looping at the MYC locus in BETi-resistant leukemia cells. Together, our findings have established the feasibility of targeting enhancer plasticity to overcome drug resistance associated with epigenetic therapies.


Asunto(s)
Leucemia Experimental/tratamiento farmacológico , Leucemia Experimental/genética , Proteínas Nucleares/antagonistas & inhibidores , Factores de Transcripción/antagonistas & inhibidores , Animales , Proteínas de Ciclo Celular/antagonistas & inhibidores , Línea Celular Tumoral , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Resistencia a Antineoplásicos/genética , Sinergismo Farmacológico , Elementos de Facilitación Genéticos , Femenino , Genes myc/efectos de los fármacos , Compuestos Heterocíclicos de 4 o más Anillos/administración & dosificación , Humanos , Células Jurkat , Células K562 , Leucemia Experimental/metabolismo , Ratones , Modelos Genéticos , Fenilendiaminas/administración & dosificación , Pirimidinas/administración & dosificación , ARN Polimerasa II/metabolismo , ARN Largo no Codificante/genética , Quinasa Activadora de Quinasas Ciclina-Dependientes
15.
J Med Chem ; 63(2): 656-675, 2020 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-31858797

RESUMEN

WD repeat domain 5 (WDR5) is a member of the WD40-repeat protein family that plays a critical role in multiple chromatin-centric processes. Overexpression of WDR5 correlates with a poor clinical outcome in many human cancers, and WDR5 itself has emerged as an attractive target for therapy. Most drug-discovery efforts center on the WIN site of WDR5 that is responsible for the recruitment of WDR5 to chromatin. Here, we describe discovery of a novel WDR5 WIN site antagonists containing a dihydroisoquinolinone bicyclic core using a structure-based design. These compounds exhibit picomolar binding affinity and selective concentration-dependent antiproliferative activities in sensitive MLL-fusion cell lines. Furthermore, these WDR5 WIN site binders inhibit proliferation in MYC-driven cancer cells and reduce MYC recruitment to chromatin at MYC/WDR5 co-bound genes. Thus, these molecules are useful probes to study the implication of WDR5 inhibition in cancers and serve as a potential starting point toward the discovery of anti-WDR5 therapeutics.


Asunto(s)
Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/síntesis química , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Quinolonas/síntesis química , Quinolonas/farmacología , Repeticiones WD40/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular , Cromatina/efectos de los fármacos , Cromatina/genética , Cristalografía por Rayos X , Diseño de Fármacos , Descubrimiento de Drogas , Represión Epigenética/efectos de los fármacos , Genes myc/efectos de los fármacos , Humanos , Relación Estructura-Actividad
16.
Bioconjug Chem ; 30(12): 3038-3045, 2019 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-31663728

RESUMEN

We herein report a cell-membrane-permeable molecular probe ADG, prepared by conjugating guanosine with anthracene, selectively interacts with c-MYC G-quadruplex over other promoter and telomeric quadruplexes as well as duplex DNA. NMR spectroscopy suggests that ADG interacts with terminal G-quartets as well as with the nearby G-rich tract (G13-G14-G15 and G8-G9-G10) of c-MYC quadruplex. In vitro cellular studies indicate that ADG represses c-MYC expression by stabilizing its promoter G-quadruplex and alters c-MYC-related cellular events. ADG suppresses hTERT and BCL2 gene expressions in a promoter-independent manner, inhibits elongation of telomere length, and activates apoptotic cascades in cancer cells.


Asunto(s)
Apoptosis/efectos de los fármacos , G-Cuádruplex , Genes myc/efectos de los fármacos , Sondas Moleculares/química , Homeostasis del Telómero/efectos de los fármacos , Antracenos/química , Línea Celular Tumoral , Permeabilidad de la Membrana Celular , Genes myc/genética , Guanosina/química , Humanos , Sondas Moleculares/metabolismo , Sondas Moleculares/farmacología , Neoplasias/patología , Regiones Promotoras Genéticas/efectos de los fármacos
17.
Chemistry ; 25(41): 9691-9700, 2019 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-31087710

RESUMEN

Guanine-rich sequences of DNA are known to readily fold into tetra-stranded helical structures known as G-quadruplexes (G4). Due to their biological relevance, G4s are potential anticancer drug targets and therefore there is significant interest in molecules with high affinity for these structures. Most G4 binders are polyaromatic planar compounds which π-π stack on the G4's guanine tetrad. However, many of these compounds are not very selective since they can also intercalate into duplex DNA. Herein we report a new class of binder based on an octahedral cobalt(III) complex that binds to G4 via a different mode involving hydrogen bonding, electrostatic interactions and π-π stacking. We show that this new compound binds selectivity to G4 over duplex DNA (particularly to the G-rich sequence of the c-myc promoter). This new octahedral complex also has the ability to template the formation of G4 DNA from the unfolded sequence. Finally, we show that upon binding to G4, the complex prevents helicase Pif1-p from unfolding the c-myc G4 structure.


Asunto(s)
Cobalto/química , Cobalto/farmacología , Complejos de Coordinación/química , Complejos de Coordinación/farmacología , ADN/química , G-Cuádruplex/efectos de los fármacos , Animales , Bovinos , ADN/genética , ADN/metabolismo , ADN Helicasas/metabolismo , Genes myc/efectos de los fármacos , Humanos , Ligandos , Simulación del Acoplamiento Molecular , Fenilendiaminas/química , Fenilendiaminas/farmacología , Regiones Promotoras Genéticas/efectos de los fármacos
18.
Biochem Pharmacol ; 166: 108-119, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31075264

RESUMEN

Aflatoxin B1 (AFB1), a member of the aflatoxin family, is a common contaminant in foods and feeds, and AFB1 exposure is associated with various clinical conditions. Thus far, research on the toxicity of AFB1 has mainly focused on its induction of liver cancer, but little research has been reported on renal toxicity, especially with regards to the underlying molecular mechanisms. In this study, we found that AFB1 treatment significantly induced kidney damage and reduced kidney weight. The human kidney cell line HEK293T was used to further study the molecular mechanism of the toxicity of AFB1 to kidney cells. We found that AFB1 significantly and dose-dependently induced S phase arrest and upregulated p21 mRNA and protein expression. Upstream of p21, three negative regulators, PLK1, MYC, and PLD1, were significantly downregulated under AFB1 treatment. Consistently, p21 was upregulated, and PLK1, MYC and PLD1 were downregulated in mouse kidney after AFB1 treatment. Interestingly, AFB1 also decreased the physical interaction between PLK1 and MYC and weakened the stability of the MYC protein. Importantly, overexpression of PLK1, MYC and PLD1 significantly blocked the upregulation of p21 and attenuated the S phase arrest caused by AFB1. In summary, AFB1 markedly induces kidney damage and strongly induces S phase arrest by upregulating the expression of p21 via PLK1, PLD1 and MYC, which represents a noval mechanism of the renal toxicity of AFB1.


Asunto(s)
Aflatoxina B1/farmacología , Proteínas de Ciclo Celular/biosíntesis , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/biosíntesis , Genes myc/efectos de los fármacos , Fosfolipasa D/biosíntesis , Proteínas Serina-Treonina Quinasas/biosíntesis , Proteínas Proto-Oncogénicas/biosíntesis , Fase S/efectos de los fármacos , Animales , Puntos de Control del Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/fisiología , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Relación Dosis-Respuesta a Droga , Expresión Génica , Genes myc/fisiología , Células HEK293 , Humanos , Masculino , Ratones , Fosfolipasa D/antagonistas & inhibidores , Fosfolipasa D/genética , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/genética , Fase S/fisiología , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología , Quinasa Tipo Polo 1
19.
Nucleic Acids Res ; 47(13): e73, 2019 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-30949695

RESUMEN

Regions of genomic instability are not random and often co-localize with DNA sequences that can adopt alternative DNA structures (i.e. non-B DNA, such as H-DNA). Non-B DNA-forming sequences are highly enriched at translocation breakpoints in human cancer genomes, representing an endogenous source of genetic instability. However, a further understanding of the mechanisms involved in non-B DNA-induced genetic instability is needed. Small molecules that can modulate the formation/stability of non-B DNA structures, and therefore the subsequent mutagenic outcome, represent valuable tools to study DNA structure-induced genetic instability. To this end, we have developed a tunable Förster resonance energy transfer (FRET)-based assay to detect triplex/H-DNA-destabilizing and -stabilizing ligands. The assay was designed by incorporating a fluorophore-quencher pair in a naturally-occurring H-DNA-forming sequence from a chromosomal breakpoint hotspot in the human c-MYC oncogene. By tuning triplex stability via buffer composition, the assay functions as a dual-reporter that can identify stabilizers and destabilizers, simultaneously. The assay principle was demonstrated using known triplex stabilizers, BePI and coralyne, and a complementary oligonucleotide to mimic a destabilizer, MCRa2. The potential of the assay was validated in a 384-well plate with 320 custom-assembled compounds. The discovery of novel triplex stabilizers/destabilizers may allow the regulation of genetic instability in human genomes.


Asunto(s)
Alcaloides de Berberina/farmacología , Puntos de Rotura del Cromosoma , ADN/efectos de los fármacos , Transferencia Resonante de Energía de Fluorescencia/métodos , Genes myc , Inestabilidad Genómica/efectos de los fármacos , Indoles/farmacología , Conformación de Ácido Nucleico/efectos de los fármacos , Piridinas/farmacología , Alcanosulfonatos/análisis , Compuestos Azo/análisis , Tampones (Química) , Dicroismo Circular , Relación Dosis-Respuesta a Droga , Doxorrubicina/farmacología , Colorantes Fluorescentes/análisis , Genes myc/efectos de los fármacos , Genoma Humano , Humanos , Ligandos , Oligodesoxirribonucleótidos/química
20.
Nat Commun ; 10(1): 620, 2019 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-30728358

RESUMEN

Elevated MYC expression sensitizes tumor cells to apoptosis but the therapeutic potential of this mechanism remains unclear. We find, in a model of MYC-driven breast cancer, that pharmacological activation of AMPK strongly synergizes with BCL-2/BCL-XL inhibitors to activate apoptosis. We demonstrate the translational potential of an AMPK and BCL-2/BCL-XL co-targeting strategy in ex vivo and in vivo models of MYC-high breast cancer. Metformin combined with navitoclax or venetoclax efficiently inhibited tumor growth, conferred survival benefits and induced tumor infiltration by immune cells. However, withdrawal of the drugs allowed tumor re-growth with presentation of PD-1+/CD8+ T cell infiltrates, suggesting immune escape. A two-step treatment regimen, beginning with neoadjuvant metformin+venetoclax to induce apoptosis and followed by adjuvant metformin+venetoclax+anti-PD-1 treatment to overcome immune escape, led to durable antitumor responses even after drug withdrawal. We demonstrate that pharmacological reactivation of MYC-dependent apoptosis is a powerful antitumor strategy involving both tumor cell depletion and immunosurveillance.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Genes myc/efectos de los fármacos , Inmunoterapia , Compuestos de Anilina/farmacología , Animales , Anticuerpos Monoclonales Humanizados , Apoptosis/inmunología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Linfocitos T CD8-positivos , Línea Celular Tumoral/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Combinación de Medicamentos , Femenino , Células HEK293 , Xenoinjertos , Humanos , Metformina/farmacología , Ratones , Ratones Transgénicos , Proteínas Proto-Oncogénicas c-bcl-2 , Sulfonamidas/farmacología , Proteína bcl-X
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA