Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Pharmacol Res ; 153: 104683, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32050092

RESUMO

Triple negative breast cancer (TNBC) is most aggressive subtype of breast cancers with high probability of metastasis as well as lack of specific targets and targeted therapeutics. TNBC is characterized with unique tumor microenvironment (TME), which differs from other subtypes. TME is associated with induction of proliferation, angiogenesis, inhibition of apoptosis and immune system suppression, and drug resistance. Exosomes are promising nanovesicles, which orchestrate the TME by communicating with different cells within TME. The components of TME including transformed ECM, soluble factors, immune suppressive cells, epigenetic modifications and re-programmed fibroblasts together hamper antitumor response and helps progression and metastasis of TNBCs. Therefore, TME could be a therapeutic target of TNBC. The current review presents latest updates on the role of exosomes in modulation of TME, approaches for targeting TME and combination of immune checkpoint inhibitors and target chemotherapeutics. Finally, we also discussed various phytochemicals that alter genetic, transcriptomic and proteomic profiles of TME along with current challenges and future implications. Thus, as TME is associated with the hallmarks of TNBC, the understanding of the impact of different components can improve the clinical benefits of TNBC patients.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Exoma/efeitos dos fármacos , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Microambiente Tumoral/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Epigênese Genética , Exoma/imunologia , Feminino , Humanos , Terapia de Alvo Molecular , Metástase Neoplásica , Células-Tronco Neoplásicas/efeitos dos fármacos , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/imunologia , Neoplasias de Mama Triplo Negativas/patologia , Microambiente Tumoral/genética , Microambiente Tumoral/imunologia
2.
Neurochem Int ; 130: 104348, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-30508560

RESUMO

Reserch progresses in understanding the pathogenicity of multiple sclerosis (MS) in the last couple of decade has enabled us to develop new drug entities available in the clinic. However, we still have not succeeded in preventing conversion from relapsing-remitting MS (RR-MS) to secondary progressive MS (SP-MS) and curing this intractable form of MS. Furthermore, diagnosis is usually retrospective and subjective, relying on gradual worsening of neurological signs/symptoms. This is obviously due to the lack of understanding for the pathogenicity driving disease progression in MS and of reliable biomarkers reflecting the progressive or stationary disease status. Two relevant components are involved in brain pathology of SP-MS, neurodegeneration and inflammation. Neurodegeneration may occur spontaneously in a neuron-intrinsic manner under chronic inflammation, such as glutamate excitotoxicity, mitochondrial/oxidative injury with iron deposit in the brain, and loss of trophic support. Meanwhile, inflammation is usually associated with recurrent relapse and the cumulative infiltration of immune cells, including T cells, B cells, and myeloid cells of peripheral or CNS origin, could ignite the processes of neurodegeneration. Especially, the higher frequency of leptomeningeal follicle-like structures observed in SP-MS patients suggests that immune cells sheltered behind a blood-brain barrier is still active under smoldering CNS inflammation. Recent successes in Ocrelizumab for primary progressive in MS (PP-MS) and Siponimod for SP-MS reappraised the importance of immune cells for pathogenesis progressive MS. Accordingly, our recent comparative analysis between MS and its animal model, experimental autoimmune encephalomyelitis (EAE), raises a new possibility that ectopic expression of eomesodermin (Eomes) in helper T (Th) cells constitutes a previously unappreciated subset of Th cells with cytotoxic potential against neuronal cells. In this review article, I will summarize the mechanisms proposed on pathogenesis of SP-MS and propose a new pathogenic mechanism for neurodegeneration mediated by unique cytotoxic Th cells.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Exoma/fisiologia , Fatores Imunológicos/administração & dosagem , Fatores Imunológicos/imunologia , Doenças Neurodegenerativas/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Animais , Anticorpos Monoclonais Humanizados/administração & dosagem , Anticorpos Monoclonais Humanizados/imunologia , Anticorpos Monoclonais Humanizados/metabolismo , Doença Crônica , Sistemas de Liberação de Medicamentos/tendências , Exoma/efeitos dos fármacos , Humanos , Fatores Imunológicos/metabolismo , Inflamação/tratamento farmacológico , Inflamação/imunologia , Inflamação/metabolismo , Doenças Neurodegenerativas/tratamento farmacológico , Doenças Neurodegenerativas/metabolismo , Linfócitos T Auxiliares-Indutores/efeitos dos fármacos , Linfócitos T Auxiliares-Indutores/metabolismo
3.
Cell Syst ; 7(6): 567-579.e6, 2018 12 26.
Artigo em Inglês | MEDLINE | ID: mdl-30503647

RESUMO

Mechanistic models are essential to deepen the understanding of complex diseases at the molecular level. Nowadays, high-throughput molecular and phenotypic characterizations are possible, but the integration of such data with prior knowledge on signaling pathways is limited by the availability of scalable computational methods. Here, we present a computational framework for the parameterization of large-scale mechanistic models and its application to the prediction of drug response of cancer cell lines from exome and transcriptome sequencing data. This framework is over 104 times faster than state-of-the-art methods, which enables modeling at previously infeasible scales. By applying the framework to a model describing major cancer-associated pathways (>1,200 species and >2,600 reactions), we could predict the effect of drug combinations from single drug data. This is the first integration of high-throughput datasets using large-scale mechanistic models. We anticipate this to be the starting point for development of more comprehensive models allowing a deeper mechanistic insight.


Assuntos
Antineoplásicos/farmacologia , Simulação por Computador , Modelos Biológicos , Neoplasias/tratamento farmacológico , Exoma/efeitos dos fármacos , Genômica , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Transdução de Sinais/efeitos dos fármacos , Biologia de Sistemas , Transcriptoma/efeitos dos fármacos
4.
Blood ; 130(14): 1639-1643, 2017 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-28827410

RESUMO

Recent studies suggest that the evolutionary history of a cancer is important in forecasting clinical outlook. To gain insight into the clonal dynamics of multiple myeloma (MM) and its possible influence on patient outcomes, we analyzed whole exome sequencing tumor data for 333 patients from Myeloma XI, a UK phase 3 trial and 434 patients from the CoMMpass study, all of which had received immunomodulatory drug (IMiD) therapy. By analyzing mutant allele frequency distributions in tumors, we found that 17% to 20% of MM is under neutral evolutionary dynamics. These tumors are associated with poorer patient survival in nonintensively treated patients, which is consistent with the reduced therapeutic efficacy of microenvironment-modulating IMiDs. Our findings provide evidence that knowledge of the evolutionary history of MM has relevance for predicting patient outcomes and personalizing therapy.


Assuntos
Frequência do Gene , Fatores Imunológicos/uso terapêutico , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/genética , Mutação , Talidomida/análogos & derivados , Talidomida/uso terapêutico , Exoma/efeitos dos fármacos , Feminino , Deriva Genética , Humanos , Imunossupressores/uso terapêutico , Estimativa de Kaplan-Meier , Lenalidomida , Masculino , Mieloma Múltiplo/diagnóstico , Mieloma Múltiplo/patologia , Prognóstico , Microambiente Tumoral/efeitos dos fármacos
5.
Carcinogenesis ; 37(8): 817-826, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27267998

RESUMO

Cholangiocarcinoma is a relatively rare cancer, but its incidence is increasing worldwide. Although several risk factors have been suggested, the etiology and pathogenesis of the majority of cholangiocarcinomas remain unclear. Recently, a high incidence of early-onset cholangiocarcinoma was reported among the workers of a printing company in Osaka, Japan. These workers underwent high exposure to organic solvents, mainly haloalkanes such as 1,2-dichloropropane (1,2-DCP) and/or dichloromethane. We performed whole-exome analysis on four cases of cholangiocarcinoma among the printing workers. An average of 44.8 somatic mutations was detected per Mb in the genome of the printing workers' cholangiocarcinoma tissues, approximately 30-fold higher than that found in control common cholangiocarcinoma tissues. Furthermore, C:G-to-T:A transitions with substantial strand bias as well as unique trinucleotide mutational changes of GpCpY to GpTpY and NpCpY to NpTpY or NpApY were predominant in all of the printing workers' cholangiocarcinoma genomes. These results were consistent with the epidemiological observation that they had been exposed to high concentrations of chemical compounds. Whole-genome analysis of Salmonella typhimurium strain TA100 exposed to 1,2-DCP revealed a partial recapitulation of the mutational signature in the printing workers' cholangiocarcinoma. Although our results provide mutational signatures unique to occupational cholangiocarcinoma, the underlying mechanisms of the disease should be further investigated by using appropriate model systems and by comparison with genomic data from other cancers.


Assuntos
Colangiocarcinoma/epidemiologia , Colangiocarcinoma/genética , Exoma/genética , Exposição Ocupacional , Adulto , Colangiocarcinoma/induzido quimicamente , Colangiocarcinoma/patologia , Exoma/efeitos dos fármacos , Humanos , Japão/epidemiologia , Masculino , Cloreto de Metileno/toxicidade , Mutação/efeitos dos fármacos , Impressão , Propano/análogos & derivados , Propano/toxicidade , Salmonella typhimurium/efeitos dos fármacos , Salmonella typhimurium/genética
6.
BMC Genomics ; 16: 866, 2015 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-26503232

RESUMO

BACKGROUND: N-ethyl-N-nitrosourea (ENU) mutagen has become the method of choice for inducing random mutations for forward genetics applications. However, distinguishing induced mutations from sequencing errors or sporadic mutations is difficult, which has hampered surveys of potential biases in the methodology in the past. Addressing this issue, we created a large cohort of mice with biological replicates enabling the confident calling of induced mutations, which in turn allowed us to conduct a comprehensive analysis of potential biases in mutation properties and genomic location. RESULTS: In the exome sequencing data we observe the known preference of ENU to cause A:T=>G:C transitions in longer genes. Mutations were frequently clustered and inherited in blocks hampering attempts to pinpoint individual causative mutations by genome analysis only. Furthermore, ENU mutations were biased towards areas in the genome that are accessible in testis, potentially limiting the scope of forward genetic approaches to only 1-10% of the genome. CONCLUSION: ENU provides a powerful tool for exploring the genome-phenome relationship, however forward genetic applications that require the mutation to be passed on through the germ line may be limited to explore only genes that are accessible in testis.


Assuntos
Etilnitrosoureia/toxicidade , Mutagênicos/toxicidade , Mutação/genética , Animais , Exoma/efeitos dos fármacos , Exoma/genética , Estudo de Associação Genômica Ampla , Masculino , Camundongos , Mutagênese/efeitos dos fármacos , Mutagênese/genética , Testículo/efeitos dos fármacos , Testículo/metabolismo
7.
Cancer Epidemiol Biomarkers Prev ; 24(12): 1873-81, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26383547

RESUMO

BACKGROUND: Dietary exposure to cytotoxic and carcinogenic aristolochic acid (AA) causes severe nephropathy typically associated with urologic cancers. Monitoring of AA exposure uses biomarkers such as aristolactam-DNA adducts, detected by mass spectrometry in the kidney cortex, or the somatic A>T transversion pattern characteristic of exposure to AA, as revealed by previous DNA-sequencing studies using fresh-frozen tumors. METHODS: Here, we report a low-coverage whole-exome sequencing method (LC-WES) optimized for multisample detection of the AA mutational signature, and demonstrate its utility in 17 formalin-fixed paraffin-embedded urothelial tumors obtained from 15 patients with endemic nephropathy, an environmental form of AA nephropathy. RESULTS: LC-WES identified the AA signature, alongside signatures of age and APOBEC enzyme activity, in 15 samples sequenced at the average per-base coverage of approximately 10×. Analysis at 3 to 9× coverage revealed the signature in 91% of the positive samples. The exome-wide distribution of the predominant A>T transversions exhibited a stochastic pattern, whereas 83 cancer driver genes were enriched for recurrent nonsynonymous A>T mutations. In two patients, pairs of tumors from different parts of the urinary tract, including the bladder, harbored overlapping mutation patterns, suggesting tumor dissemination via cell seeding. CONCLUSIONS: LC-WES analysis of archived tumor tissues is a reliable method applicable to investigations of both the exposure to AA and its biologic effects in human carcinomas. IMPACT: By detecting cancers associated with AA exposure in high-risk populations, LC-WES can support future molecular epidemiology studies and provide evidence-base for relevant preventive measures.


Assuntos
Ácidos Aristolóquicos/análise , Exoma/efeitos dos fármacos , Neoplasias/química , Neoplasias/genética , Carcinógenos/análise , Formaldeído , Humanos , Neoplasias/patologia , Inclusão em Parafina , Análise de Sequência de DNA/métodos , Fixação de Tecidos
8.
Gastroenterology ; 149(5): 1163-1176.e2, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26255561

RESUMO

In this review, we provide an update on genome-wide association studies (GWAS) in inflammatory bowel disease (IBD). In addition, we summarize progress in defining the functional consequences of associated alleles for coding and noncoding genetic variation. In the small minority of loci where major association signals correspond to nonsynonymous variation, we summarize studies defining their functional effects and implications for therapeutic targeting. Importantly, the large majority of GWAS-associated loci involve noncoding variation, many of which modulate levels of gene expression. Recent expression quantitative trait loci (eQTL) studies have established that the expression of most human genes is regulated by noncoding genetic variations. Significant advances in defining the epigenetic landscape have demonstrated that IBD GWAS signals are highly enriched within cell-specific active enhancer marks. Studies in European ancestry populations have dominated the landscape of IBD genetics studies, but increasingly, studies in Asian and African-American populations are being reported. Common variation accounts for only a modest fraction of the predicted heritability and the role of rare genetic variation of higher effects (ie, odds ratios markedly deviating from 1) is increasingly being identified through sequencing efforts. These sequencing studies have been particularly productive in more severe very early onset cases. A major challenge in IBD genetics will be harnessing the vast array of genetic discovery for clinical utility through emerging precision medical initiatives. In this article, we discuss the rapidly evolving area of direct-to-consumer genetic testing and the current utility of clinical exome sequencing, especially in very early onset, severe IBD cases. We summarize recent progress in the pharmacogenetics of IBD with respect to partitioning patient responses to anti-TNF and thiopurine therapies. Highly collaborative studies across research centers and across subspecialties and disciplines will be required to fully realize the promise of genetic discovery in IBD.


Assuntos
Expressão Gênica , Terapia Genética/métodos , Variação Genética , Estudo de Associação Genômica Ampla/métodos , Doenças Inflamatórias Intestinais/genética , Alelos , Triagem e Testes Direto ao Consumidor/métodos , Epigenômica/métodos , Exoma/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Testes Genéticos/métodos , Variação Genética/efeitos dos fármacos , Humanos , Farmacogenética/métodos , RNA não Traduzido/efeitos dos fármacos
9.
Mutat Res Genet Toxicol Environ Mutagen ; 775-776: 48-54, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25435355

RESUMO

Genetic mutations are known to drive cancer progression and certain tumors have mutation signatures that reflect exposures to environmental carcinogens. Benzo[a]pyrene (BaP) has a known mutation signature and has proven capable of inducing changes to DNA sequence that drives normal pre-stasis human mammary epithelial cells (HMEC) past a first tumor suppressor barrier (stasis) and toward immortality. We analyzed normal, pre-stasis HMEC, three independent BaP-derived post-stasis HMEC strains (184Aa, 184Be, 184Ce) and two of their immortal derivatives(184A1 and 184BE1) by whole exome sequencing. The independent post-stasis strains exhibited between 93 and 233 BaP-induced mutations in exons. Seventy percent of the mutations were C:G>A:T transversions, consistent with the known mutation spectrum of BaP. Mutations predicted to impact protein function occurred in several known and putative cancer drivers including p16, PLCG1, MED12, TAF1 in 184Aa; PIK3CG, HSP90AB1, WHSC1L1, LCP1 in 184Be and FANCA, LPP in 184Ce. Biological processes that typically harbor cancer driver mutations such as cell cycle, regulation of cell death and proliferation, RNA processing, chromatin modification and DNA repair were found to have mutations predicted to impact function in each of the post-stasis strains. Spontaneously immortalized HMEC lines derived from two of the BaP-derived post-stasis strains shared greater than 95% of their BaP-induced mutations with their precursor cells. These immortal HMEC had 10 or fewer additional point mutations relative to their post-stasis precursors, but acquired chromosomal anomalies during immortalization that arose independent of BaP. The results of this study indicate that acute exposures of HMEC to high dose BaP recapitulate mutation patterns of human tumors and can induce mutations in a number of cancer driver genes.


Assuntos
Benzo(a)pireno/toxicidade , Exoma/efeitos dos fármacos , Glândulas Mamárias Humanas/efeitos dos fármacos , Carcinógenos Ambientais , Células Cultivadas , Aberrações Cromossômicas , Células Epiteliais/efeitos dos fármacos , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Genes Neoplásicos , Humanos , Glândulas Mamárias Humanas/citologia , Mutação , Neoplasias/induzido quimicamente , Neoplasias/genética , Análise de Sequência de DNA , Adulto Jovem
10.
PLoS One ; 9(10): e110740, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25360671

RESUMO

Hydroxyurea has proven efficacy in children and adults with sickle cell anemia (SCA), but with considerable inter-individual variability in the amount of fetal hemoglobin (HbF) produced. Sibling and twin studies indicate that some of that drug response variation is heritable. To test the hypothesis that genetic modifiers influence pharmacological induction of HbF, we investigated phenotype-genotype associations using whole exome sequencing of children with SCA treated prospectively with hydroxyurea to maximum tolerated dose (MTD). We analyzed 171 unrelated patients enrolled in two prospective clinical trials, all treated with dose escalation to MTD. We examined two MTD drug response phenotypes: HbF (final %HbF minus baseline %HbF), and final %HbF. Analyzing individual genetic variants, we identified multiple low frequency and common variants associated with HbF induction by hydroxyurea. A validation cohort of 130 pediatric sickle cell patients treated to MTD with hydroxyurea was genotyped for 13 non-synonymous variants with the strongest association with HbF response to hydroxyurea in the discovery cohort. A coding variant in Spalt-like transcription factor, or SALL2, was associated with higher final HbF in this second independent replication sample and SALL2 represents an outstanding novel candidate gene for further investigation. These findings may help focus future functional studies and provide new insights into the pharmacological HbF upregulation by hydroxyurea in patients with SCA.


Assuntos
Anemia Falciforme/tratamento farmacológico , Anemia Falciforme/genética , Exoma/genética , Hemoglobina Fetal/metabolismo , Genômica , Hidroxiureia/farmacologia , Análise de Sequência de DNA , Anemia Falciforme/metabolismo , Criança , Estudos de Coortes , Proteínas de Ligação a DNA , Relação Dose-Resposta a Droga , Exoma/efeitos dos fármacos , Humanos , Hidroxiureia/efeitos adversos , Hidroxiureia/uso terapêutico , Dose Máxima Tolerável , Fases de Leitura Aberta/genética , Fenótipo , Estudos Prospectivos , Fatores de Transcrição/genética , Resultado do Tratamento
11.
PLoS One ; 8(3): e55429, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23469164

RESUMO

BACKGROUND: Mice harbouring gene mutations that cause phenotypic abnormalities during organogenesis are invaluable tools for linking gene function to normal development and human disorders. To generate mouse models harbouring novel alleles that are involved in organogenesis we conducted a phenotype-driven, genome-wide mutagenesis screen in mice using the mutagen N-ethyl-N-nitrosourea (ENU). METHODOLOGY/PRINCIPAL FINDINGS: ENU was injected into male C57BL/6 mice and the mutations transmitted through the germ-line. ENU-induced mutations were bred to homozygosity and G3 embryos screened at embryonic day (E) 13.5 and E18.5 for abnormalities in limb and craniofacial structures, skin, blood, vasculature, lungs, gut, kidneys, ureters and gonads. From 52 pedigrees screened 15 were detected with anomalies in one or more of the structures/organs screened. Using single nucleotide polymorphism (SNP)-based linkage analysis in conjunction with candidate gene or next-generation sequencing (NGS) we identified novel recessive alleles for Fras1, Ift140 and Lig1. CONCLUSIONS/SIGNIFICANCE: In this study we have generated mouse models in which the anomalies closely mimic those seen in human disorders. The association between novel mutant alleles and phenotypes will lead to a better understanding of gene function in normal development and establish how their dysfunction causes human anomalies and disease.


Assuntos
Anormalidades Congênitas/genética , Modelos Animais de Doenças , Etilnitrosoureia/toxicidade , Exoma/efeitos dos fármacos , Camundongos Endogâmicos C57BL/genética , Mutagênicos/toxicidade , Polimorfismo de Nucleotídeo Único , Alelos , Animais , Anormalidades Congênitas/embriologia , Anormalidades Congênitas/etiologia , DNA Ligase Dependente de ATP , DNA Ligases/genética , Proteínas da Matriz Extracelular/genética , Feminino , Estudo de Associação Genômica Ampla , Genótipo , Mutação em Linhagem Germinativa/efeitos dos fármacos , Sequenciamento de Nucleotídeos em Larga Escala , Homozigoto , Fatores de Determinação Direita-Esquerda/genética , Masculino , Camundongos , Mutagênese , Fenótipo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA