Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
1.
Signal Transduct Target Ther ; 6(1): 299, 2021 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-34373448

RESUMO

Aflatoxin exposure is a crucial factor in promoting the development of primary hepatocellular carcinoma (HCC) in individuals infected with the hepatitis virus. However, the molecular pathways leading to its bioactivation and subsequent toxicity in hepatocytes have not been well-defined. Here, we carried out a genome-wide CRISPR-Cas9 genetic screen to identify aflatoxin B1 (AFB1) targets. Among the most significant hits was the aryl hydrocarbon receptor (AHR), a ligand-binding transcription factor regulating cell metabolism, differentiation, and immunity. AHR-deficient cells tolerated high concentrations of AFB1, in which AFB1 adduct formation was significantly decreased. AFB1 triggered AHR nuclear translocation by directly binding to its N-terminus. Furthermore, AHR mediated the expression of P450 induced by AFB1. AHR expression was also elevated in primary tumor sections obtained from AFB1-HCC patients, which paralleled the upregulation of PD-L1, a clinically relevant immune regulator. Finally, anti-PD-L1 therapy exhibited greater efficacy in HCC xenografts derived from cells with ectopic expression of AHR. These results demonstrated that AHR was required for the AFB1 toxicity associated with HCC, and implicate the immunosuppressive regimen of anti-PD-L1 as a therapeutic option for the treatment of AFB1-associated HCCs.


Assuntos
Antígeno B7-H1/genética , Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/genética , Receptores de Hidrocarboneto Arílico/genética , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Aflatoxina B1/farmacologia , Animais , Antígeno B7-H1/antagonistas & inibidores , Sistemas CRISPR-Cas/efeitos dos fármacos , Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/terapia , Carcinoma Hepatocelular/virologia , Linhagem Celular Tumoral , Genoma Humano/efeitos dos fármacos , Vírus de Hepatite/patogenicidade , Hepatócitos/efeitos dos fármacos , Humanos , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/terapia , Neoplasias Hepáticas/virologia , Camundongos , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Cells ; 10(5)2021 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-34065904

RESUMO

(1) Background: Chronic inflammation has been regarded as a risk factor for the onset and progression of human cancer, but the critical molecular mechanisms underlying this pathological process have yet to be elucidated. (2) Methods: In this study, we investigated whether interleukin (IL)-17-mediated inflammation was involved in cigarette smoke-induced genomic instability. (3) Results: Higher levels of both IL-17 and the DNA damage response (DDR) were found in the lung tissues of smokers than in those of non-smokers. Similarly, elevated levels of IL-17 and the DDR were observed in mice after cigarette smoke exposure, and a positive correlation was observed between IL-17 expression and the DDR. In line with these observations, the DDR in the mouse lung was diminished in IL-17 KO when exposed to cigarette smoke. Besides this, the treatment of human bronchial epithelium cells with IL-17 led to increased levels of the DDR and chromosome breakage. (4) Conclusions: These results suggest that cigarette smoke induces genomic instability at least partially through IL-17-mediated inflammation, implying that IL-17 could play an important role in the development of lung cancer.


Assuntos
Genoma Humano/efeitos dos fármacos , Inflamação/induzido quimicamente , Interleucina-17/metabolismo , Fumar/efeitos adversos , Produtos do Tabaco/efeitos adversos , Animais , Brônquios/citologia , Células Cultivadas , Dano ao DNA , Células Epiteliais/citologia , Instabilidade Genômica , Humanos , Mediadores da Inflamação/metabolismo , Pulmão/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fumaça
3.
Eur J Pharmacol ; 889: 173641, 2020 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-33045196

RESUMO

Although majority of acute promyelocytic leukemia (APL) patients achieve complete remission after the standard treatment, 5-10% of patients are shown to relapse or develop resistance to treatment. In such cases, medications that target epigenetic processes could become an appealing supplementary approach. In this study, we tested the anti-leukemic activity of histone deacetylase inhibitor Belinostat (PXD101) and histone methyltransferase inhibitor 3-Deazaneplanocin A combined with all-trans retinoic acid in APL cells NB4, promyelocytes resembling HL-60 cells and APL patients' cells. After HL-60 and NB4 cell treatment, ChIP-sequencing was performed using antibodies against hyper-acetylated histone H4. Hyper-acetylated histone H4 distribution peaks were compared in treated vs untreated HL-60 and NB4 cells. Results demonstrated that in treated HL-60 cells, the majority of peaks were distributed within the regions of proximal promoters, whereas in treated NB4 cells, hyper-acetylated histone H4 peaks were mainly localized in gene body regions. Further ChIP-seq data analysis revealed the changes in histone H4 hyper-acetylation in promoter/gene body regions of genes involved in cancer signaling pathways. In addition, quantitative gene expression analysis proved changes in various cellular pathways important for carcinogenesis. Epigenetic treatment down-regulated the expression of MTOR, LAMTOR1, WNT2B, VEGFR3, FGF2, FGFR1, TGFA, TGFB1, TGFBR1, PDGFA, PDGFRA and PDGFRB genes in NB4, HL-60 and APL patients' cells. In addition, effect of epigenetic treatment on protein expression of aforementioned signaling pathways was confirmed with mass spectrometry analysis. Taken together, these results provide supplementary insights into molecular changes that occur during epigenetic therapy application in in vitro promyelocytic leukemia cell model.


Assuntos
Epigênese Genética/genética , Genoma Humano/genética , Inibidores de Histona Desacetilases/uso terapêutico , Histonas/antagonistas & inibidores , Histonas/genética , Leucemia Promielocítica Aguda/genética , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Epigênese Genética/efeitos dos fármacos , Genoma Humano/efeitos dos fármacos , Células HL-60 , Inibidores de Histona Desacetilases/farmacologia , Humanos , Ácidos Hidroxâmicos/farmacologia , Ácidos Hidroxâmicos/uso terapêutico , Leucemia Promielocítica Aguda/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Sulfonamidas/farmacologia , Sulfonamidas/uso terapêutico
4.
BMC Cancer ; 20(1): 880, 2020 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-32928150

RESUMO

BACKGROUND: Tobacco smoking is associated with a unique mutational signature in the human cancer genome. It is unclear whether tobacco smoking-altered DNA methylations and gene expressions affect smoking-related mutational signature. METHODS: We systematically analyzed the smoking-related DNA methylation sites reported from five previous casecontrol studies in peripheral blood cells to identify possible target genes. Using the mediation analysis approach, we evaluated whether the association of tobacco smoking with mutational signature is mediated through altered DNA methylation and expression of these target genes in lung adenocarcinoma tumor tissues. RESULTS: Based on data obtained from 21,108 blood samples, we identified 374 smoking-related DNA methylation sites, annotated to 248 target genes. Using data from DNA methylations, gene expressions and smoking-related mutational signature generated from ~ 7700 tumor tissue samples across 26 cancer types from The Cancer Genome Atlas (TCGA), we found 11 of the 248 target genes whose expressions were associated with smoking-related mutational signature at a Bonferroni-correction P < 0.001. This included four for head and neck cancer, and seven for lung adenocarcinoma. In lung adenocarcinoma, our results showed that smoking increased the expression of three genes, AHRR, GPR15, and HDGF, and decreased the expression of two genes, CAPN8, and RPS6KA1, which were consequently associated with increased smoking-related mutational signature. Additional evidence showed that the elevated expression of AHRR and GPR15 were associated with smoking-altered hypomethylations at cg14817490 and cg19859270, respectively, in lung adenocarcinoma tumor tissues. Lastly, we showed that decreased expression of RPS6KA1, were associated with poor survival of lung cancer patients. CONCLUSIONS: Our findings provide novel insight into the contributions of tobacco smoking to carcinogenesis through the underlying mechanisms of the elevated mutational signature by altered DNA methylations and gene expressions.


Assuntos
Metilação de DNA/efeitos dos fármacos , Epigênese Genética/genética , Neoplasias/genética , Fumar Tabaco/efeitos adversos , Ilhas de CpG/efeitos dos fármacos , Metilação de DNA/genética , Epigênese Genética/efeitos dos fármacos , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genoma Humano/efeitos dos fármacos , Humanos , Masculino , Análise de Mediação , Mutação/genética , Proteínas de Neoplasias/genética , Neoplasias/sangue , Neoplasias/induzido quimicamente , Neoplasias/patologia
5.
Signal Transduct Target Ther ; 5(1): 152, 2020 08 19.
Artigo em Inglês | MEDLINE | ID: mdl-32811807

RESUMO

Laboratory research and pharmacoepidemiology provide support for metformin as a potential antitumor agent. However, the lack of a clear understanding of the indications of metformin limits its efficacy. Here, we performed a genome-wide CRISPR knockout negative screen to identify potential targets that might synergize with metformin. Next-generation sequencing of pooled genomic DNAs isolated from surviving cells after 18 days of metformin treatment (T18) compared to those of the untreated cells at day 0 (T0) yielded candidate genes. Knockdown of a group of cyclin-dependent kinases (CDKs), including CDK1, CDK4, and CDK6, confirmed the results of the screen. Combination treatment of the CDKs inhibitor abemaciclib with metformin profoundly inhibited tumor viability in vitro and in vivo. Although cell cycle parameters were not further altered under the combination treatment, investigation of the metabolome revealed significant changes in cell metabolism, especially with regard to fatty acid oxidation, the tricarboxylic acid cycle and aspartate metabolism. Such changes appeared to be mediated through inhibition of the mTOR pathway. Collectively, our study suggests that the combination of CDKs inhibitor with metformin could be recognized as a potential therapy in future clinical applications.


Assuntos
Proteína Quinase CDC2/genética , Quinase 4 Dependente de Ciclina/genética , Quinase 6 Dependente de Ciclina/genética , Metformina/farmacologia , Neoplasias/tratamento farmacológico , Animais , Ácido Aspártico/metabolismo , Proteína Quinase CDC2/antagonistas & inibidores , Sistemas CRISPR-Cas/genética , Ciclo do Ácido Cítrico , Quinase 4 Dependente de Ciclina/antagonistas & inibidores , Quinase 6 Dependente de Ciclina/antagonistas & inibidores , Sinergismo Farmacológico , Ácidos Graxos/metabolismo , Técnicas de Silenciamento de Genes , Genoma Humano/efeitos dos fármacos , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Células MCF-7 , Camundongos , Neoplasias/genética , Neoplasias/patologia , Inibidores de Proteínas Quinases/farmacologia , Serina-Treonina Quinases TOR/genética
6.
Clin Cancer Res ; 26(20): 5477-5486, 2020 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-32816946

RESUMO

PURPOSE: Malignant pleural mesothelioma (MPM) is considered an orphan disease with few treatment options. Despite multimodality therapy, the majority of MPMs recur and eventually become refractory to any systemic treatment. One potential mechanism underlying therapeutic resistance may be intratumor heterogeneity (ITH), making MPM challenging to eradicate. However, the ITH architecture of MPM and its clinical impact have not been well studied. EXPERIMENTAL DESIGN: We delineated the immunogenomic ITH by multiregion whole-exome sequencing and T-cell receptor (TCR) sequencing of 69 longitudinal MPM specimens from nine patients with resectable MPM, who were treated with dasatinib. RESULTS: The median total mutation burden before dasatinib treatment was 0.65/Mb, similar with that of post-dasatinib treatment (0.62/Mb). The median proportion of mutations shared by any given pair of two tumor regions within the same tumors was 80% prior to and 83% post-dasatinib treatment indicating a relatively homogenous genomic landscape. T-cell clonality, a parameter indicating T-cell expansion and reactivity, was significantly increased in tumors after dasatinib treatment. Furthermore, on average, 82% of T-cell clones were restricted to individual tumor regions, with merely 6% of T-cell clones shared by all regions from the same tumors indicating profound TCR heterogeneity. Interestingly, patients with higher T-cell clonality and higher portion of T cells present across all tumor regions in post-dasatinib-treated tumors had significantly longer survival. CONCLUSIONS: Despite the homogeneous genomic landscape, the TCR repertoire is extremely heterogeneous in MPM. Dasatinib may potentially induce T-cell response leading to improved survival.


Assuntos
Dasatinibe/administração & dosagem , Mesotelioma Maligno/tratamento farmacológico , Recidiva Local de Neoplasia/tratamento farmacológico , Linfócitos T/efeitos dos fármacos , Adulto , Idoso , Evolução Clonal/genética , Dasatinibe/efeitos adversos , Evolução Molecular , Feminino , Heterogeneidade Genética , Genoma Humano/efeitos dos fármacos , Genômica , Humanos , Masculino , Mesotelioma Maligno/genética , Mesotelioma Maligno/patologia , Pessoa de Meia-Idade , Mutação/genética , Proteínas de Neoplasias/genética , Recidiva Local de Neoplasia/genética , Recidiva Local de Neoplasia/patologia , Intervalo Livre de Progressão , Receptores de Antígenos de Linfócitos T/genética , Linfócitos T/patologia , Sequenciamento do Exoma
7.
Artigo em Inglês | MEDLINE | ID: mdl-32522347

RESUMO

Recent years have witnessed an expansion of mutagenesis research focusing on experimentally modeled genome-scale mutational signatures of carcinogens and of endogenous processes. Experimental mutational signatures can explain etiologic links to patterns found in human tumors that may be linked to same exposures, and can serve as biomarkers of exposure history and may even provide insights on causality. A number of innovative exposure models have been employed and reported, based on cells cultured in monolayers or in 3-D, on organoids, induced pluripotent stem cells, non-mammalian organisms, microorganisms and rodent bioassays. Here we discuss some of the latest developments and pros and cons of these experimental systems used in mutational signature analysis. Integrative designs that bring together multiple exposure systems (in vitro, in vivo and in silico pan-cancer data mining) started emerging as powerful tools to identify robust mutational signatures of the tested cancer risk agents. We further propose that devising a new generation of cell-based models is warranted to streamline systematic testing of carcinogen effects on the cell genomes, while seeking to increasingly supplant animal with non-animal systems to address relevant ethical issues and accentuate the 3R principles. We conclude that the knowledge accumulating from the growing body of signature modelling investigations has considerable power to advance cancer etiology studies and to support cancer prevention efforts through streamlined characterization of cancer-causing agents and the recognition of their specific effects.


Assuntos
Carcinógenos/toxicidade , Mutagênese/efeitos dos fármacos , Mutação/efeitos dos fármacos , Neoplasias/induzido quimicamente , Animais , Análise Mutacional de DNA/métodos , Genoma Humano/efeitos dos fármacos , Genoma Humano/genética , Humanos , Mutagênese/genética , Mutação/genética
8.
Nature ; 580(7804): 517-523, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32322066

RESUMO

A high tumour mutational burden (hypermutation) is observed in some gliomas1-5; however, the mechanisms by which hypermutation develops and whether it predicts the response to immunotherapy are poorly understood. Here we comprehensively analyse the molecular determinants of mutational burden and signatures in 10,294 gliomas. We delineate two main pathways to hypermutation: a de novo pathway associated with constitutional defects in DNA polymerase and mismatch repair (MMR) genes, and a more common post-treatment pathway, associated with acquired resistance driven by MMR defects in chemotherapy-sensitive gliomas that recur after treatment with the chemotherapy drug temozolomide. Experimentally, the mutational signature of post-treatment hypermutated gliomas was recapitulated by temozolomide-induced damage in cells with MMR deficiency. MMR-deficient gliomas were characterized by a lack of prominent T cell infiltrates, extensive intratumoral heterogeneity, poor patient survival and a low rate of response to PD-1 blockade. Moreover, although bulk analyses did not detect microsatellite instability in MMR-deficient gliomas, single-cell whole-genome sequencing analysis of post-treatment hypermutated glioma cells identified microsatellite mutations. These results show that chemotherapy can drive the acquisition of hypermutated populations without promoting a response to PD-1 blockade and supports the diagnostic use of mutational burden and signatures in cancer.


Assuntos
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/terapia , Glioma/genética , Glioma/terapia , Mutação , Animais , Antineoplásicos Alquilantes/farmacologia , Antineoplásicos Alquilantes/uso terapêutico , Neoplasias Encefálicas/imunologia , Reparo de Erro de Pareamento de DNA/genética , Frequência do Gene , Genoma Humano/efeitos dos fármacos , Genoma Humano/genética , Glioma/imunologia , Humanos , Masculino , Camundongos , Repetições de Microssatélites/efeitos dos fármacos , Repetições de Microssatélites/genética , Mutagênese/efeitos dos fármacos , Mutação/efeitos dos fármacos , Fenótipo , Prognóstico , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Análise de Sequência de DNA , Temozolomida/farmacologia , Temozolomida/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto
9.
J Comput Aided Mol Des ; 34(3): 219-230, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31925639

RESUMO

Small molecules binding at any of the multiple regulatory sites on the molecular surface of a protein kinase may stabilize or disrupt the corresponding interaction, leading to consequent modulation of the kinase cellular activity. As such, each of these sites represents a potential drug target. Even targeting sites outside the immediate ATP site, the so-called exosites, may cause desirable biological effects through an allosteric mechanism. Targeting exosites can alleviate adverse effects and toxicity that is common when ATP-site compounds bind promiscuously to many other types of kinases. In this study we have identified, catalogued, and annotated all potentially druggable exosites on the protein kinase domains within the existing structural human kinome. We then priority-ranked these exosites by those most amenable to drug design. In order to identify pockets that are either consistent across the kinome, or unique and specific to a particular structure, we have also implemented a normalized representation of all pockets, and displayed these graphically. Finally, we have built a database and designed a web-based interface for users interested in accessing the 3-dimensional representations of these pockets. We envision this information will assist drug discovery efforts searching for untargeted binding pockets in the human kinome.


Assuntos
Sítios de Ligação/genética , Desenho de Fármacos , Genoma Humano/efeitos dos fármacos , Proteínas Quinases/genética , Trifosfato de Adenosina/química , Trifosfato de Adenosina/genética , Sítios de Ligação/efeitos dos fármacos , Genoma Humano/genética , Humanos , Ligação Proteica/genética , Domínios Proteicos/genética , Proteínas Quinases/química , Propriedades de Superfície/efeitos dos fármacos
10.
Sci Rep ; 9(1): 15222, 2019 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-31645597

RESUMO

Recent advances in pharmacogenomics have generated a wealth of data of different types whose analysis have helped in the identification of signatures of different cellular sensitivity/resistance responses to hundreds of chemical compounds. Among the different data types, gene expression has proven to be the more successful for the inference of drug response in cancer cell lines. Although effective, the whole transcriptome can introduce noise in the predictive models, since specific mechanisms are required for different drugs and these realistically involve only part of the proteins encoded in the genome. We analyzed the pharmacogenomics data of 961 cell lines tested with 265 anti-cancer drugs and developed different machine learning approaches for dissecting the genome systematically and predict drug responses using both drug-unspecific and drug-specific genes. These methodologies reach better response predictions for the vast majority of the screened drugs using tens to few hundreds genes specific to each drug instead of the whole genome, thus allowing a better understanding and interpretation of drug-specific response mechanisms which are not necessarily restricted to the drug known targets.


Assuntos
Antineoplásicos/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Neoplasias/genética , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Genoma Humano/efeitos dos fármacos , Humanos , Aprendizado de Máquina , Modelos Biológicos , Farmacogenética , Transcriptoma/efeitos dos fármacos
11.
Cancer Biol Ther ; 20(11): 1398-1402, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31423907

RESUMO

Medulloblastoma is an aggressive primitive neuroectodermal tumor of the cerebellum that is more common in children than in adults. In the past decade, advances in understanding the molecular drivers of medulloblastoma have identified four molecular subgroups defined by experimental gene expression profiles: the WNT pathway, sonic hedgehog (SHH) pathway, and subgroups 3 and 4 (non-SHH/WNT).  Medulloblastoma of adults belong primarily to the SHH category. Vismodegib, an SHH-pathway inhibitor, FDA-approved in 2012 for treatment of basal cell carcinoma, has been used successfully in the setting of chemorefractory medulloblastoma, but not as a first-line therapy. In 2016, we reported a case of an adult patient with a sustained response of an unresectable multifocal form of adult medulloblastoma to vismodegib. Molecular analysis in that case revealed mutations in TP53 and a cytogenetic abnormality, i17q, that is prevalent and most often associated with subgroup 4 rather than the SHH-activated form of medulloblastoma. Here, we report further whole-genome analysis of that patient (designated Patient A) as well as an additional adult patient (Patient B) whose tumor harbored the SHH molecular subgroup but which was unresponsive to visgmodegib therapy. Comparison of these disparate responses highlights the challenges to tailoring SHH-targeted treatment in individual patients with adult medulloblastoma.


Assuntos
Anilidas/administração & dosagem , Metilação de DNA/genética , Proteínas Hedgehog/genética , Meduloblastoma/tratamento farmacológico , Piridinas/administração & dosagem , Adulto , Encéfalo/diagnóstico por imagem , Encéfalo/patologia , Metilação de DNA/efeitos dos fármacos , Genoma Humano/efeitos dos fármacos , Humanos , Imageamento por Ressonância Magnética , Masculino , Meduloblastoma/diagnóstico por imagem , Meduloblastoma/genética , Meduloblastoma/patologia , Pessoa de Meia-Idade , Mutação/efeitos dos fármacos , Via de Sinalização Wnt/efeitos dos fármacos
12.
Steroids ; 151: 108463, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31344408

RESUMO

Paxillin is extensively involved in focal adhesion signaling and kinase signaling throughout the plasma membrane and cytoplasm. However, recent studies in prostate cancer suggest that paxillin also plays a critical role in regulating gene expression within the nucleus, serving as a liaison between cytoplasmic and nuclear MAPK and Androgen Receptor (AR) signaling. Here we used RNA-seq to examine the paxillin-regulated transcriptome in several human prostate cancer cell lines. First, we examined paxillin effects on androgen-mediated transcription in control or paxillin-depleted AR-positive LNCaP and C4-2 human prostate cancer cells. In androgen-dependent LNCaP cells, we found over 1000 paxillin-dependent androgen-responsive genes, some of which are involved in endocrine therapy resistance. Most paxillin-dependent AR-mediated genes in LNCaP cells were no longer paxillin-dependent in androgen-sensitive, castration-resistant C4-2 cells, suggesting that castration-resistance may markedly alter paxillin effects on genomic AR signaling. To examine the paxillin-regulated transcriptome in the absence of androgen signaling, we performed RNA-seq in AR-negative PC3 human prostate cancer cells. Paxillin enhanced several pro-proliferative pathways, including the CyclinD/Rb/E2F and DNA replication/repair pathways. Additionally, paxillin suppressed pro-apoptotic genes, including CASP1 and TNFSF10. Quantitative PCR confirmed that these pathways are similarly regulated by paxillin in LNCaP and C4-2 cells. Functional studies showed that, while paxillin stimulated cell proliferation, it had minimum effect on apoptosis. Thus, paxillin appears to be an important transcriptional regulator in prostate cancer, and analysis of its transcriptome might lead to novel approaches toward the diagnosis and treatment of this important disease.


Assuntos
Redes Reguladoras de Genes/efeitos dos fármacos , Genoma Humano/efeitos dos fármacos , Paxilina/farmacologia , Neoplasias da Próstata/genética , Apoptose/efeitos dos fármacos , Apoptose/genética , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Humanos , Masculino , Neoplasias da Próstata/patologia
13.
Psychiatry Clin Neurosci ; 73(6): 323-330, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30821055

RESUMO

AIM: Glucocorticoids play a major role in regulating the stress response, and an imbalance of glucocorticoids has been implicated in stress-related disorders. Within mouse models, CpGs across the genome have been shown to be differentially methylated in response to glucocorticoid treatment, and using the Infinium 27K array, it was shown that humans given synthetic glucocorticoids had DNA methylation (DNAm) changes in blood. However, further investigation of the extent to which glucocorticoids affect DNAm across a larger proportion of the genome is needed. METHODS: Buccal samples were collected before and after synthetic glucocorticoid treatment in the context of a dental procedure. This included 30 tooth extraction surgery patients who received 10 mg of dexamethasone. Genome-wide DNAm was assessed with the Infinium HumanMethylationEPIC array. RESULTS: Five CpGs showed genome-wide significant DNAm changes that were >10%. These differentially methylated CpGs were in or nearest the following genes: ZNF438, KLHDC10, miR-544 or CRABP1, DPH5, and WDFY2. Using previously published datasets of human blood gene expression changes following dexamethasone exposure, a significant proportion of genes with false-discovery-rate-adjusted significant CpGs were also differentially expressed. A pathway analysis of the genes with false-discovery-rate-adjusted significant CpGs revealed significant enrichment of olfactory transduction, pentose and glucuronate interconversions, ascorbate and aldarate metabolism, and steroid hormone biosynthesis pathways. CONCLUSION: High-dose synthetic glucocorticoid administration in the setting of a dental procedure was significantly associated with DNAm changes within buccal samples. These findings are consistent with prior findings of an influence of glucocorticoids on DNAm in humans.


Assuntos
Ilhas de CpG/efeitos dos fármacos , Metilação de DNA/efeitos dos fármacos , Dexametasona/farmacologia , Expressão Gênica/efeitos dos fármacos , Genoma Humano/efeitos dos fármacos , Glucocorticoides/farmacologia , Adulto , Dexametasona/administração & dosagem , Feminino , Glucocorticoides/administração & dosagem , Humanos , Masculino , Mucosa Bucal , Procedimentos Cirúrgicos Bucais , Adulto Jovem
14.
Cancer Genomics Proteomics ; 15(5): 365-378, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30194077

RESUMO

BACKGROUND/AIM: Genomic DNA copy number alterations (CNAs) are frequent in tumors and have been catalogued by The Cancer Genome Atlas project. Emergence of chemoresistance frequently renders drug therapies ineffective. MATERIALS AND METHODS: We analyzed how CNAs recurrently found in the genomes of TCGA patients of thirty-one tumor types affect protein targets of antineoplastic (AN) agents. RESULTS: CNA deletions more frequently affected the targets of AN agents than CNA amplifications. Interestingly, in seven tumors we observed signs of compensatory CNAs. For example, in glioblastoma multiforme, two target genes (FLT1, FLT3) of the experimental drug sorafenib were recurrently deleted, whereas another target (KDR) of sorafenib was recurrently amplified. In renal clear cell carcinoma, the target FLT1 of pazopanib, sunitinib, sorafenib, and axitinib was recurrently deleted, whereas FLT4 bound by the same drugs, was recurrently amplified. CONCLUSION: Deletions of AN target proteins can be compensated by amplification of alternative targets.


Assuntos
Carcinoma de Células Renais/tratamento farmacológico , Variações do Número de Cópias de DNA/genética , Resistencia a Medicamentos Antineoplásicos/genética , Glioblastoma/tratamento farmacológico , Antineoplásicos/efeitos adversos , Axitinibe , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/patologia , Feminino , Genoma Humano/efeitos dos fármacos , Glioblastoma/genética , Glioblastoma/patologia , Humanos , Imidazóis/uso terapêutico , Indazóis/uso terapêutico , Indóis/uso terapêutico , Masculino , Terapia de Alvo Molecular , Niacinamida/análogos & derivados , Niacinamida/uso terapêutico , Compostos de Fenilureia/uso terapêutico , Pirróis/uso terapêutico , Sorafenibe , Sunitinibe , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/genética , Tirosina Quinase 3 Semelhante a fms/genética
15.
Biogerontology ; 19(6): 579-602, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-29907918

RESUMO

Hutchinson-Gilford progeria syndrome (HGPS) is a rare and fatal premature ageing disease in children. HGPS is one of several progeroid syndromes caused by mutations in the LMNA gene encoding the nuclear structural proteins lamins A and C. In classic HGPS the mutation G608G leads to the formation of a toxic lamin A protein called progerin. During post-translational processing progerin remains farnesylated owing to the mutation interfering with a step whereby the farnesyl moiety is removed by the enzyme ZMPSTE24. Permanent farnesylation of progerin is thought to be responsible for the proteins toxicity. Farnesyl is generated through the mevalonate pathway and three drugs that interfere with this pathway and hence the farnesylation of proteins have been administered to HGPS children in clinical trials. These are a farnesyltransferase inhibitor (FTI), statin and a bisphosphonate. Further experimental studies have revealed that other drugs such as N-acetyl cysteine, rapamycin and IGF-1 may be of use in treating HGPS through other pathways. We have shown previously that FTIs restore chromosome positioning in interphase HGPS nuclei. Mis-localisation of chromosomes could affect the cells ability to regulate proper genome function. Using nine different drug treatments representing drug regimes in the clinic we have shown that combinatorial treatments containing FTIs are most effective in restoring specific chromosome positioning towards the nuclear periphery and in tethering telomeres to the nucleoskeleton. On the other hand, rapamycin was found to be detrimental to telomere tethering, it was, nonetheless, the most effective at inducing DNA damage repair, as revealed by COMET analyses.


Assuntos
Dano ao DNA/efeitos dos fármacos , Difosfonatos/uso terapêutico , Farnesiltranstransferase/antagonistas & inibidores , Genoma Humano/efeitos dos fármacos , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Progéria/tratamento farmacológico , Sirolimo/uso terapêutico , Linhagem Celular , Ensaio Cometa , Difosfonatos/farmacologia , Quimioterapia Combinada , Feminino , Fibroblastos/efeitos dos fármacos , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Fator de Crescimento Insulin-Like I/farmacologia , Lamina Tipo A/genética , Laminas/genética , Proteínas de Membrana/genética , Metaloendopeptidases/genética , Mutação , Progéria/genética , Progéria/metabolismo , Processamento de Proteína Pós-Traducional , Sirolimo/farmacologia
16.
Food Chem Toxicol ; 109(Pt 1): 714-720, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28887092

RESUMO

Cadmium (Cd) exposure is not easily avoidable; it is a common contaminant found in many food sources, accumulates throughout life and, in high doses, is a significant health hazard for humans. Women are highly vulnerable to Cd because of their relatively higher absorption rate than men. High levels of Cd accumulated in the mother could potentially cause harm to both the mother and new-born child. The foetal genome is vulnerable to external signals; Cd partially crosses the placental barrier and can impact on foetal development, potentially, through epigenetic mechanisms causing changes to foetal gene expression. This review explores current research on Cd induced methylation changes to maternal and foetal genomes. Cd is significantly associated with differential methylation of both maternal and foetal genomes. Some studies have described infant sex-specific changes in DNA methylation in association with maternal Cd burden. However, research on methylation changes to the foetal genome due to prenatal Cd exposure is scarce. More research is required to explore the impact of maternal Cd accumulation on differential methylation of the foetal genome.


Assuntos
Cádmio/toxicidade , Feto/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Exposição Materna/efeitos adversos , Adolescente , Adulto , Idoso , Peso ao Nascer , Cádmio/metabolismo , Criança , Pré-Escolar , Metilação de DNA , Poluentes Ambientais/metabolismo , Poluentes Ambientais/toxicidade , Epigênese Genética , Feminino , Desenvolvimento Fetal/efeitos dos fármacos , Feto/metabolismo , Genoma Humano/efeitos dos fármacos , Humanos , Lactente , Rim/química , Rim/metabolismo , Fígado/química , Fígado/metabolismo , Pulmão/química , Pulmão/metabolismo , Masculino , Pessoa de Meia-Idade , Placenta/efeitos dos fármacos , Placenta/metabolismo , Gravidez , Adulto Jovem
17.
Chem Biol Interact ; 273: 257-265, 2017 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-28666766

RESUMO

During the recent years, microarray analysis of gene expression has become an inevitable tool for exploring toxicity of drugs and other chemicals on biological systems. Therefore, toxicogenomics is considered as a fruitful area for searching cellular pathways and mechanisms including cancer, immunological diseases, environmental responses, gene-gene interactions and chemical toxicity. In this work, we examined toxic effects of Tungsten Borides NPs on gene expression profiling of the human lung alveolar epithelial cells (HPAEpiC). In line with this purpose, a single crystal of tungsten boride (mixture of WB and W2B) nanoparticles was synthesized by means of zone melting method, and characterized via using X-ray crystallography (XRD), transmission electron microscope (TEM), scanning electron microscope (SEM) and energy-dispersive X-ray spectroscopy (EDX) techniques. Cell viability and cytotoxicity were determined by 3-(4,5-dimethyl-thiazol-2-yl) 2,5-diphenyltetrazolium bromide (MTT), neutral red (NR) and lactate dehydrogenase (LDH) release tests. The whole genome microarray expression analysis was performed to find out the effects of WB and W2B NPs mixture on gene expression of the HPAEpiC cell culture. 123 of 40,000 gene probes were assigned to characterize expression profile for WB/W2B NPs exposure. According to results; 70 genes were up-regulated and 53 genes were down-regulated (≥2 fold change). For further investigations, these genes were functionally classified by using DAVID (The Database for Annotation, Visualization and Integrated Discovery) with gene ontology (GO) analysis. In the light of the data gained from this study, it could be concluded that the mixture of WB/W2B NPs can affect cytokine/chemokine metabolism, angiogenesis and prevent migration/invasion by activating various genes.


Assuntos
Células Epiteliais Alveolares/efeitos dos fármacos , Células Epiteliais Alveolares/metabolismo , Compostos de Boro/toxicidade , Genoma Humano/genética , Nanopartículas/toxicidade , Tungstênio/toxicidade , Células Epiteliais Alveolares/citologia , Compostos de Boro/química , Morte Celular/efeitos dos fármacos , Morte Celular/genética , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Células Cultivadas , Citocinas/metabolismo , Perfilação da Expressão Gênica , Genoma Humano/efeitos dos fármacos , Humanos , Nanopartículas/química , Análise de Sequência com Séries de Oligonucleotídeos , Tamanho da Partícula , Propriedades de Superfície , Toxicogenética , Tungstênio/química
18.
J Cancer Res Ther ; 13(1): 9-15, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28508826

RESUMO

The therapeutic options of nonsmall cell lung cancer (NSCLC) therapy has been changed since the first discovery of activating epidermal growth factor receptor (EGFR) mutations and the development of specific EGFR tyrosine kinase inhibitors, which resulted in the evolution of "personalized medicine." There are a considerable number of genomic aberrations in NSCLC serving as potential predictive biomarkers and drug targets and still more. We summarized the molecular pathways, potential targets, and possible impact on disease outcome in NSCLC.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Receptores ErbB/genética , Genoma Humano/genética , Terapia de Alvo Molecular , Biomarcadores Tumorais/genética , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Receptores ErbB/antagonistas & inibidores , Genoma Humano/efeitos dos fármacos , Genômica , Humanos , Mutação , Medicina de Precisão , Transdução de Sinais/efeitos dos fármacos , Resultado do Tratamento
19.
Clin Cancer Res ; 23(17): 5003-5014, 2017 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-28490462

RESUMO

Purpose: The past decade has witnessed the rapid development of personalized targeted therapies in lung cancer. It is still unclear whether epigenetic changes are involved in the response to tyrosine kinase inhibitor (TKI) treatment in epidermal growth factor receptor (EGFR)-mutated lung cancer.Experimental Design: Methyl-sensitive cut counting sequencing (MSCC) was applied to investigate the methylation changes in paired tissues before and after erlotinib treatment for 42 days with partial response (PR) from stage IIIa (N2) lung adenocarcinoma patients (N = 2) with EGFR 19 deletion. The Sequenom EpiTYPER assay was used to validate the changed methylated candidate genes. Up- or downregulation of the candidate gene was performed to elucidate the potential mechanism in the regulation of erlotinib treatment response.Results: Sixty aberrant methylated genes were screened using MSCC sequencing. Two aberrant methylated genes, CBFA2T3 and GABBR2, were clearly validated. A same differential methylated region (DMR) between exon 2 and exon 3 of GABBR2 gene was confirmed consistently in both patients. GABBR2 was significantly downregulated in EGFR 19 deletion cells, HCC4006 and HCC827, but remained conserved in EGFR wild-type A549 cells after erlotinib treatment. Upregulation of GABBR2 expression significantly rescued erlotinib-induced apoptosis in HCC827 cells. GABBR2 was significantly downregulated, along with the reduction of S6, p-p70 S6, and p-ERK1/2, demonstrating that GABBR2 may play an important role in EGFR signaling through the ERK1/2 pathway.Conclusions: We demonstrated that GABBR2 gene might be a novel potential epigenetic treatment target with induction erlotinib treatment for stage IIIa (N2) EGFR 19 deletion lung adenocarcinoma. Clin Cancer Res; 23(17); 5003-14. ©2017 AACR.


Assuntos
Adenocarcinoma/genética , Metilação de DNA/genética , Receptores ErbB/genética , Neoplasias Pulmonares/genética , Receptores de GABA-B/genética , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/patologia , Adenocarcinoma de Pulmão , Apoptose/genética , Linhagem Celular Tumoral , Proliferação de Células , Metilação de DNA/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Epigênese Genética/genética , Cloridrato de Erlotinib/administração & dosagem , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genoma Humano/efeitos dos fármacos , Genoma Humano/genética , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Masculino , Pessoa de Meia-Idade , Mutação , Inibidores de Proteínas Quinases/administração & dosagem , Proteínas Repressoras/genética , Deleção de Sequência , Transdução de Sinais/efeitos dos fármacos , Resultado do Tratamento , Proteínas Supressoras de Tumor/genética
20.
Angew Chem Int Ed Engl ; 56(23): 6483-6487, 2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28474855

RESUMO

Cisplatin derivatives can form various types of DNA lesions (DNA-Pt) and trigger pleiotropic DNA damage responses. Here, we report a strategy to visualize DNA-Pt with high resolution, taking advantage of a novel azide-containing derivative of cisplatin we named APPA, a cellular pre-extraction protocol and the labeling of DNA-Pt by means of click chemistry in cells. Our investigation revealed that pretreating cells with the histone deacetylase (HDAC) inhibitor SAHA led to detectable clusters of DNA-Pt that colocalized with the ubiquitin ligase RAD18 and the replication protein PCNA. Consistent with activation of translesion synthesis (TLS) under these conditions, SAHA and cisplatin cotreatment promoted focal accumulation of the low-fidelity polymerase Polη that also colocalized with PCNA. Remarkably, these cotreatments synergistically triggered mono-ubiquitination of PCNA and apoptosis in a RAD18-dependent manner. Our data provide evidence for a role of chromatin in regulating genome targeting with cisplatin derivatives and associated cellular responses.


Assuntos
Antineoplásicos/farmacologia , Cromatina/fisiologia , Cisplatino/farmacologia , Genoma Humano/efeitos dos fármacos , Linhagem Celular Tumoral , Cisplatino/análogos & derivados , Química Click , DNA/efeitos dos fármacos , Dano ao DNA , DNA Polimerase Dirigida por DNA/metabolismo , Inibidores de Histona Desacetilases/farmacologia , Humanos , Sondas Moleculares , Antígeno Nuclear de Célula em Proliferação/metabolismo , Ubiquitinação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA