Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 114
Filtrar
1.
Nature ; 620(7972): 209-217, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37438531

RESUMEN

The human genome functions as a three-dimensional chromatin polymer, driven by a complex collection of chromosome interactions1-3. Although the molecular rules governing these interactions are being quickly elucidated, relatively few proteins regulating this process have been identified. Here, to address this gap, we developed high-throughput DNA or RNA labelling with optimized Oligopaints (HiDRO)-an automated imaging pipeline that enables the quantitative measurement of chromatin interactions in single cells across thousands of samples. By screening the human druggable genome, we identified more than 300 factors that influence genome folding during interphase. Among these, 43 genes were validated as either increasing or decreasing interactions between topologically associating domains. Our findings show that genetic or chemical inhibition of the ubiquitous kinase GSK3A leads to increased long-range chromatin looping interactions in a genome-wide and cohesin-dependent manner. These results demonstrate the importance of GSK3A signalling in nuclear architecture and the use of HiDRO for identifying mechanisms of spatial genome organization.


Asunto(s)
Cromatina , Posicionamiento de Cromosoma , Cromosomas Humanos , Genoma Humano , Glucógeno Sintasa Quinasas , Ensayos Analíticos de Alto Rendimiento , Análisis de la Célula Individual , Humanos , Cromatina/efectos de los fármacos , Cromatina/genética , Cromatina/metabolismo , Posicionamiento de Cromosoma/efectos de los fármacos , Cromosomas Humanos/efectos de los fármacos , Cromosomas Humanos/genética , Cromosomas Humanos/metabolismo , ADN/análisis , ADN/metabolismo , Genoma Humano/efectos de los fármacos , Genoma Humano/genética , Glucógeno Sintasa Quinasas/antagonistas & inhibidores , Glucógeno Sintasa Quinasas/deficiencia , Glucógeno Sintasa Quinasas/genética , Ensayos Analíticos de Alto Rendimiento/métodos , Interfase , Reproducibilidad de los Resultados , ARN/análisis , ARN/metabolismo , Transducción de Señal/efectos de los fármacos , Análisis de la Célula Individual/métodos , Cohesinas
2.
Environ Mol Mutagen ; 63(1): 37-63, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35023215

RESUMEN

This review considers antiviral nucleoside analog drugs, including ribavirin, favipiravir, and molnupiravir, which induce genome error catastrophe in SARS-CoV or SARS-CoV-2 via lethal mutagenesis as a mode of action. In vitro data indicate that molnupiravir may be 100 times more potent as an antiviral agent than ribavirin or favipiravir. Molnupiravir has recently demonstrated efficacy in a phase 3 clinical trial. Because of its anticipated global use, its relative potency, and the reported in vitro "host" cell mutagenicity of its active principle, ß-d-N4-hydroxycytidine, we have reviewed the development of molnupiravir and its genotoxicity safety evaluation, as well as the genotoxicity profiles of three congeners, that is, ribavirin, favipiravir, and 5-(2-chloroethyl)-2'-deoxyuridine. We consider the potential genetic risks of molnupiravir on the basis of all available information and focus on the need for additional human genotoxicity data and follow-up in patients treated with molnupiravir and similar drugs. Such human data are especially relevant for antiviral NAs that have the potential of permanently modifying the genomes of treated patients and/or causing human teratogenicity or embryotoxicity. We conclude that the results of preclinical genotoxicity studies and phase 1 human clinical safety, tolerability, and pharmacokinetics are critical components of drug safety assessments and sentinels of unanticipated adverse health effects. We provide our rationale for performing more thorough genotoxicity testing prior to and within phase 1 clinical trials, including human PIG-A and error corrected next generation sequencing (duplex sequencing) studies in DNA and mitochondrial DNA of patients treated with antiviral NAs that induce genome error catastrophe via lethal mutagenesis.


Asunto(s)
Antivirales/efectos adversos , Tratamiento Farmacológico de COVID-19 , Citidina/análogos & derivados , Daño del ADN/efectos de los fármacos , Hidroxilaminas/efectos adversos , Nucleósidos/efectos adversos , SARS-CoV-2/genética , Amidas/efectos adversos , Amidas/uso terapéutico , Antivirales/uso terapéutico , Citidina/efectos adversos , Citidina/uso terapéutico , Desoxiuridina/efectos adversos , Desoxiuridina/análogos & derivados , Desoxiuridina/uso terapéutico , Genoma Humano/efectos de los fármacos , Humanos , Hidroxilaminas/uso terapéutico , Mutagénesis/efectos de los fármacos , Nucleósidos/uso terapéutico , Pirazinas/efectos adversos , Pirazinas/uso terapéutico , Ribavirina/efectos adversos , Ribavirina/uso terapéutico , SARS-CoV-2/efectos de los fármacos
3.
Nutrients ; 13(11)2021 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-34835928

RESUMEN

Diet bioactive components, in the concept of nutrigenetics and nutrigenomics, consist of food constituents, which can transfer information from the external environment and influence gene expression in the cell and thus the function of the whole organism. It is crucial to regard food not only as the source of energy and basic nutriments, crucial for living and organism development, but also as the factor influencing health/disease, biochemical mechanisms, and activation of biochemical pathways. Bioactive components of the diet regulate gene expression through changes in the chromatin structure (including DNA methylation and histone modification), non-coding RNA, activation of transcription factors by signalling cascades, or direct ligand binding to the nuclear receptors. Analysis of interactions between diet components and human genome structure and gene activity is a modern approach that will help to better understand these relations and will allow designing dietary guidances, which can help maintain good health.


Asunto(s)
Factores Biológicos/genética , Dieta/efectos adversos , Alimentos/efectos adversos , Regulación de la Expresión Génica/efectos de los fármacos , Fenómenos Fisiológicos de la Nutrición/genética , Metilación de ADN/efectos de los fármacos , Análisis de los Alimentos , Genoma Humano/efectos de los fármacos , Código de Histonas/efectos de los fármacos , Humanos , Nutrigenómica , Receptores Citoplasmáticos y Nucleares/genética , Transducción de Señal/efectos de los fármacos , Factores de Transcripción/genética
4.
Signal Transduct Target Ther ; 6(1): 299, 2021 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-34373448

RESUMEN

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.


Asunto(s)
Antígeno B7-H1/genética , Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/genética , Receptores de Hidrocarburo de Aril/genética , Transporte Activo de Núcleo Celular/efectos de los fármacos , Aflatoxina B1/farmacología , Animales , Antígeno B7-H1/antagonistas & inhibidores , Sistemas CRISPR-Cas/efectos de los fármacos , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/terapia , Carcinoma Hepatocelular/virología , Línea Celular Tumoral , Genoma Humano/efectos de los fármacos , Virus de Hepatitis/patogenicidad , Hepatocitos/efectos de los fármacos , Humanos , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/terapia , Neoplasias Hepáticas/virología , Ratones , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Cells ; 10(5)2021 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-34065904

RESUMEN

(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.


Asunto(s)
Genoma Humano/efectos de los fármacos , Inflamación/inducido químicamente , Interleucina-17/metabolismo , Fumar/efectos adversos , Productos de Tabaco/efectos adversos , Animales , Bronquios/citología , Células Cultivadas , Daño del ADN , Células Epiteliales/citología , Inestabilidad Genómica , Humanos , Mediadores de Inflamación/metabolismo , Pulmón/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Humo
7.
Nucleic Acids Res ; 49(D1): D1138-D1143, 2021 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-33068428

RESUMEN

The public Comparative Toxicogenomics Database (CTD; http://ctdbase.org/) is an innovative digital ecosystem that relates toxicological information for chemicals, genes, phenotypes, diseases, and exposures to advance understanding about human health. Literature-based, manually curated interactions are integrated to create a knowledgebase that harmonizes cross-species heterogeneous data for chemical exposures and their biological repercussions. In this biennial update, we report a 20% increase in CTD curated content and now provide 45 million toxicogenomic relationships for over 16 300 chemicals, 51 300 genes, 5500 phenotypes, 7200 diseases and 163 000 exposure events, from 600 comparative species. Furthermore, we increase the functionality of chemical-phenotype content with new data-tabs on CTD Disease pages (to help fill in knowledge gaps for environmental health) and new phenotype search parameters (for Batch Query and Venn analysis tools). As well, we introduce new CTD Anatomy pages that allow users to uniquely explore and analyze chemical-phenotype interactions from an anatomical perspective. Finally, we have enhanced CTD Chemical pages with new literature-based chemical synonyms (to improve querying) and added 1600 amino acid-based compounds (to increase chemical landscape). Together, these updates continue to augment CTD as a powerful resource for generating testable hypotheses about the etiologies and molecular mechanisms underlying environmentally influenced diseases.


Asunto(s)
Bases de Datos Factuales , Interacción Gen-Ambiente , Genoma Humano/efectos de los fármacos , Genómica/métodos , Medicamentos bajo Prescripción/farmacología , Xenobióticos/toxicidad , Bases de Datos de Compuestos Químicos , Bases de Datos Genéticas , Genotipo , Humanos , Internet , Bases del Conocimiento , Especificidad de Órganos , Fenotipo , Medicamentos bajo Prescripción/química , Programas Informáticos , Toxicogenética/métodos , Xenobióticos/química
8.
Nucleic Acids Res ; 49(D1): D1144-D1151, 2021 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-33237278

RESUMEN

The Drug-Gene Interaction Database (DGIdb, www.dgidb.org) is a web resource that provides information on drug-gene interactions and druggable genes from publications, databases, and other web-based sources. Drug, gene, and interaction data are normalized and merged into conceptual groups. The information contained in this resource is available to users through a straightforward search interface, an application programming interface (API), and TSV data downloads. DGIdb 4.0 is the latest major version release of this database. A primary focus of this update was integration with crowdsourced efforts, leveraging the Drug Target Commons for community-contributed interaction data, Wikidata to facilitate term normalization, and export to NDEx for drug-gene interaction network representations. Seven new sources have been added since the last major version release, bringing the total number of sources included to 41. Of the previously aggregated sources, 15 have been updated. DGIdb 4.0 also includes improvements to the process of drug normalization and grouping of imported sources. Other notable updates include the introduction of a more sophisticated Query Score for interaction search results, an updated Interaction Score, the inclusion of interaction directionality, and several additional improvements to search features, data releases, licensing documentation and the application framework.


Asunto(s)
Colaboración de las Masas , Bases de Datos Factuales , Bases de Datos Genéticas , Drogas en Investigación/farmacología , Genoma Humano/efectos de los fármacos , Medicamentos bajo Prescripción/farmacología , Bases de Datos de Compuestos Químicos , Drogas en Investigación/química , Genotipo , Humanos , Internet , Bases del Conocimiento , Fenotipo , Medicamentos bajo Prescripción/química , Programas Informáticos
9.
PLoS One ; 15(12): e0243905, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33351840

RESUMEN

Pyrrole-imidazole (Py-Im) polyamides are synthetic molecules that can be rationally designed to target specific DNA sequences to both disrupt and recruit transcriptional machinery. While in vitro binding has been extensively studied, in vivo effects are often difficult to predict using current models of DNA binding. Determining the impact of genomic architecture and the local chromatin landscape on polyamide-DNA sequence specificity remains an unresolved question that impedes their effective deployment in vivo. In this report we identified polyamide-DNA interaction sites across the entire genome, by covalently crosslinking and capturing these events in the nuclei of human LNCaP cells. This technique confirms the ability of two eight ring hairpin-polyamides, with similar architectures but differing at a single ring position (Py to Im), to retain in vitro specificities and display distinct genome-wide binding profiles.


Asunto(s)
Proteínas de Unión al ADN/genética , Genoma Humano/efectos de los fármacos , Conformación de Ácido Nucleico/efectos de los fármacos , Nylons/farmacología , Proteínas de Unión al ADN/antagonistas & inhibidores , Humanos , Imidazoles/farmacología , Pirroles/farmacología
10.
Eur J Pharmacol ; 889: 173641, 2020 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-33045196

RESUMEN

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.


Asunto(s)
Epigénesis Genética/genética , Genoma Humano/genética , Inhibidores de Histona Desacetilasas/uso terapéutico , Histonas/antagonistas & inhibidores , Histonas/genética , Leucemia Promielocítica Aguda/genética , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Epigénesis Genética/efectos de los fármacos , Genoma Humano/efectos de los fármacos , Células HL-60 , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Ácidos Hidroxámicos/farmacología , Ácidos Hidroxámicos/uso terapéutico , Leucemia Promielocítica Aguda/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Sulfonamidas/farmacología , Sulfonamidas/uso terapéutico
11.
JCI Insight ; 5(20)2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-32970634

RESUMEN

The integration of HIV DNA into the host genome contributes to lifelong infection in most individuals. Few studies have examined integration in lymphoid tissue, where HIV predominantly persists before and after antiretroviral treatment (ART). Of particular interest is whether integration site distributions differ between infection stages with paired blood and tissue comparisons. Here, we profiled HIV integration site distributions in sorted memory, tissue-resident, and/or follicular helper CD4+ T cell subsets from paired blood and lymphoid tissue samples from acute, chronic, and ART-treated individuals. We observed minor differences in the frequency of nonintronic and nondistal intergenic sites, varying with tissue and residency phenotypes during ART. Genomic and epigenetic annotations were generally similar. Clonal expansion of cells marked by identical integration sites was detected, with increased detection in chronic and ART-treated individuals. However, overlap between or within CD4+ T cell subsets or tissue compartments was only observed in 8 unique sites of the 3540 sites studied. Together, these findings suggest that shared integration sites between blood and tissue may, depending on the tissue site, be the exception rather than the rule and indicate that additional studies are necessary to fully understand the heterogeneity of tissue-sequestered HIV reservoirs.


Asunto(s)
ADN Viral/genética , Infecciones por VIH/genética , Interacciones Huésped-Patógeno/genética , Integración Viral/genética , Adulto , Antirretrovirales/administración & dosificación , Linfocitos T CD4-Positivos/virología , Genoma Humano/efectos de los fármacos , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/patología , Infecciones por VIH/virología , VIH-1/genética , VIH-1/patogenicidad , Humanos , Tejido Linfoide/virología , Masculino , Subgrupos de Linfocitos T/virología , Carga Viral/genética , Adulto Joven
12.
BMC Cancer ; 20(1): 880, 2020 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-32928150

RESUMEN

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.


Asunto(s)
Metilación de ADN/efectos de los fármacos , Epigénesis Genética/genética , Neoplasias/genética , Fumar Tabaco/efectos adversos , Islas de CpG/efectos de los fármacos , Metilación de ADN/genética , Epigénesis Genética/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genoma Humano/efectos de los fármacos , Humanos , Masculino , Análisis de Mediación , Mutación/genética , Proteínas de Neoplasias/genética , Neoplasias/sangre , Neoplasias/inducido químicamente , Neoplasias/patología
13.
Signal Transduct Target Ther ; 5(1): 152, 2020 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-32811807

RESUMEN

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.


Asunto(s)
Proteína Quinasa CDC2/genética , Quinasa 4 Dependiente de la Ciclina/genética , Quinasa 6 Dependiente de la Ciclina/genética , Metformina/farmacología , Neoplasias/tratamiento farmacológico , Animales , Ácido Aspártico/metabolismo , Proteína Quinasa CDC2/antagonistas & inhibidores , Sistemas CRISPR-Cas/genética , Ciclo del Ácido Cítrico , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 6 Dependiente de la Ciclina/antagonistas & inhibidores , Sinergismo Farmacológico , Ácidos Grasos/metabolismo , Técnicas de Silenciamiento del Gen , Genoma Humano/efectos de los fármacos , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Células MCF-7 , Ratones , Neoplasias/genética , Neoplasias/patología , Inhibidores de Proteínas Quinasas/farmacología , Serina-Treonina Quinasas TOR/genética
14.
Clin Cancer Res ; 26(20): 5477-5486, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-32816946

RESUMEN

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.


Asunto(s)
Dasatinib/administración & dosificación , Mesotelioma Maligno/tratamiento farmacológico , Recurrencia Local de Neoplasia/tratamiento farmacológico , Linfocitos T/efectos de los fármacos , Adulto , Anciano , Evolución Clonal/genética , Dasatinib/efectos adversos , Evolución Molecular , Femenino , Heterogeneidad Genética , Genoma Humano/efectos de los fármacos , Genómica , Humanos , Masculino , Mesotelioma Maligno/genética , Mesotelioma Maligno/patología , Persona de Mediana Edad , Mutación/genética , Proteínas de Neoplasias/genética , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/patología , Supervivencia sin Progresión , Receptores de Antígenos de Linfocitos T/genética , Linfocitos T/patología , Secuenciación del Exoma
15.
Artículo en Inglés | MEDLINE | ID: mdl-32522347

RESUMEN

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.


Asunto(s)
Carcinógenos/toxicidad , Mutagénesis/efectos de los fármacos , Mutación/efectos de los fármacos , Neoplasias/inducido químicamente , Animales , Análisis Mutacional de ADN/métodos , Genoma Humano/efectos de los fármacos , Genoma Humano/genética , Humanos , Mutagénesis/genética , Mutación/genética
16.
Crit Rev Eukaryot Gene Expr ; 30(1): 85-91, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32421987

RESUMEN

Intravenous immunoglobulin (IVIG)-resistant Kawasaki disease (KD) is a complex disease, leading to the damage of multiple systems. The pathogen that triggers this sophisticated disease is still unknown. The aim of this study was to identify gene signatures during IVIG-resistant KD and uncover their potential mechanisms. The gene expression profiles of GSE18606 were downloaded from the GEO database. The GSE18606 dataset contained eight IVIG-resistant KD samples and nine healthy age-appropriate controls. The gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes pathway analyses were performed, and protein-protein interaction (PPI) network of the differentially expressed genes (DEGs) was constructed by Cytoscape software. In total, 73 DEGs were identified in IVIG-resistant KD, including 58 upregulated genes and 15 downregulated genes. GO analysis results showed that DEGs were significantly enriched in biological processes of neutrophil degranulation, neutrophil mediated immunity, and neutrophil activation involved in immune response. Among them, 10 hub genes (S100A8, S100A9, S100A12, HGF, LCN2, LY96, CTGF, MMP8, IRAK3, and SLPI) with a high degree of connectivity were selected. The present study indicated that the identified DEGs and hub genes promote our understanding of the molecular mechanisms underlying the development of IVIG-resistant KD, and might be used as molecular targets and diagnostic biomarkers for the treatment of IVIG-resistant KD.


Asunto(s)
Resistencia a Medicamentos/genética , Inmunoglobulinas Intravenosas/efectos adversos , Síndrome Mucocutáneo Linfonodular/tratamiento farmacológico , Mapas de Interacción de Proteínas/genética , Preescolar , Biología Computacional , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Ontología de Genes , Genoma Humano/efectos de los fármacos , Genoma Humano/genética , Humanos , Inmunoglobulinas Intravenosas/administración & dosificación , Lactante , Masculino , Síndrome Mucocutáneo Linfonodular/genética , Síndrome Mucocutáneo Linfonodular/inmunología , Mapas de Interacción de Proteínas/efectos de los fármacos , Mapas de Interacción de Proteínas/inmunología , Programas Informáticos , Transcriptoma/genética
17.
Nature ; 580(7804): 517-523, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32322066

RESUMEN

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.


Asunto(s)
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/terapia , Glioma/genética , Glioma/terapia , Mutación , Animales , Antineoplásicos Alquilantes/farmacología , Antineoplásicos Alquilantes/uso terapéutico , Neoplasias Encefálicas/inmunología , Reparación de la Incompatibilidad de ADN/genética , Frecuencia de los Genes , Genoma Humano/efectos de los fármacos , Genoma Humano/genética , Glioma/inmunología , Humanos , Masculino , Ratones , Repeticiones de Microsatélite/efectos de los fármacos , Repeticiones de Microsatélite/genética , Mutagénesis/efectos de los fármacos , Mutación/efectos de los fármacos , Fenotipo , Pronóstico , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Análisis de Secuencia de ADN , Temozolomida/farmacología , Temozolomida/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto
18.
J Mol Endocrinol ; 64(4): R45-R56, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32229699

RESUMEN

Molecular endocrinology of vitamin D is based on the activation of the transcription factor vitamin D receptor (VDR) by the vitamin D metabolite 1α,25-dihydroxyvitamin D3. This nuclear vitamin D-sensing process causes epigenome-wide effects, such as changes in chromatin accessibility as well as in the contact of VDR and its supporting pioneer factors with thousands of genomic binding sites, referred to as vitamin D response elements. VDR binding enhancer regions loop to transcription start sites of hundreds of vitamin D target genes resulting in changes of their expression. Thus, vitamin D signaling is based on epigenome- and transcriptome-wide shifts in VDR-expressing tissues. Monocytes are the most responsive cell type of the immune system and serve as a paradigm for uncovering the chromatin model of vitamin D signaling. In this review, an alternative approach for selecting vitamin D target genes is presented, which are most relevant for understanding the impact of vitamin D endocrinology on innate immunity. Different scenarios of the regulation of primary upregulated vitamin D target genes are presented, in which vitamin D-driven super-enhancers comprise a cluster of persistent (constant) and/or inducible (transient) VDR-binding sites. In conclusion, the spatio-temporal VDR binding in the context of chromatin is most critical for the regulation of vitamin D target genes.


Asunto(s)
Cromatina/fisiología , Vitamina D/metabolismo , Vitamina D/farmacología , Animales , Sitios de Unión/efectos de los fármacos , Sitios de Unión/genética , Cromatina/efectos de los fármacos , Epigénesis Genética/efectos de los fármacos , Epigénesis Genética/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Genoma Humano/efectos de los fármacos , Genoma Humano/fisiología , Humanos , Unión Proteica/genética , Receptores de Calcitriol/fisiología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Activación Transcripcional/efectos de los fármacos , Vitamina D/análogos & derivados , Vitamina D/fisiología
19.
Genes (Basel) ; 11(4)2020 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-32260267

RESUMEN

Schizophrenia (SCZ) is a psychiatric disorder characterized by both positive and negative symptoms, including cognitive dysfunction, decline in motivation, delusion and hallucinations. Antipsychotic agents are currently the standard of care treatment for SCZ. However, only about one-third of SCZ patients respond to antipsychotic medications. In the current study, we have performed a meta-analysis of publicly available whole-genome expression datasets on Brodmann area 46 of the brain dorsolateral prefrontal cortex in order to prioritize potential pathways underlying SCZ pathology. Moreover, we have evaluated whether the differentially expressed genes in SCZ belong to specific subsets of cell types. Finally, a cross-tissue comparison at both the gene and functional level was performed by analyzing the transcriptomic pattern of peripheral blood mononuclear cells of SCZ patients. Our study identified a robust disease-specific set of dysfunctional biological pathways characterizing SCZ patients that could in the future be exploited as potential therapeutic targets.


Asunto(s)
Encéfalo/metabolismo , Corteza Prefrontal/metabolismo , Esquizofrenia/genética , Transcriptoma/genética , Antipsicóticos/uso terapéutico , Encéfalo/patología , Mapeo Encefálico , Disfunción Cognitiva/tratamiento farmacológico , Disfunción Cognitiva/genética , Disfunción Cognitiva/patología , Femenino , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica/efectos de los fármacos , Genoma Humano/efectos de los fármacos , Humanos , Leucocitos Mononucleares/metabolismo , Masculino , Corteza Prefrontal/patología , Esquizofrenia/tratamiento farmacológico , Esquizofrenia/patología , Transducción de Señal/efectos de los fármacos
20.
J Comput Aided Mol Des ; 34(3): 219-230, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31925639

RESUMEN

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.


Asunto(s)
Sitios de Unión/genética , Diseño de Fármacos , Genoma Humano/efectos de los fármacos , Proteínas Quinasas/genética , Adenosina Trifosfato/química , Adenosina Trifosfato/genética , Sitios de Unión/efectos de los fármacos , Genoma Humano/genética , Humanos , Unión Proteica/genética , Dominios Proteicos/genética , Proteínas Quinasas/química , Propiedades de Superficie/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA