Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nat Med ; 2024 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-38760585

RESUMO

Neural-tumor interactions drive glioma growth as evidenced in preclinical models, but clinical validation is limited. We present an epigenetically defined neural signature of glioblastoma that independently predicts patients' survival. We use reference signatures of neural cells to deconvolve tumor DNA and classify samples into low- or high-neural tumors. High-neural glioblastomas exhibit hypomethylated CpG sites and upregulation of genes associated with synaptic integration. Single-cell transcriptomic analysis reveals a high abundance of malignant stemcell-like cells in high-neural glioblastoma, primarily of the neural lineage. These cells are further classified as neural-progenitor-cell-like, astrocyte-like and oligodendrocyte-progenitor-like, alongside oligodendrocytes and excitatory neurons. In line with these findings, high-neural glioblastoma cells engender neuron-to-glioma synapse formation in vitro and in vivo and show an unfavorable survival after xenografting. In patients, a high-neural signature is associated with decreased overall and progression-free survival. High-neural tumors also exhibit increased functional connectivity in magnetencephalography and resting-state magnet resonance imaging and can be detected via DNA analytes and brain-derived neurotrophic factor in patients' plasma. The prognostic importance of the neural signature was further validated in patients diagnosed with diffuse midline glioma. Our study presents an epigenetically defined malignant neural signature in high-grade gliomas that is prognostically relevant. High-neural gliomas likely require a maximized surgical resection approach for improved outcomes.

2.
Acta Neuropathol ; 147(1): 21, 2024 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-38244080

RESUMO

The longitudinal transition of phenotypes is pivotal in glioblastoma treatment resistance and DNA methylation emerged as an important tool for classifying glioblastoma phenotypes. We aimed to characterize DNA methylation subclass heterogeneity during progression and assess its clinical impact. Matched tissues from 47 glioblastoma patients were subjected to DNA methylation profiling, including CpG-site alterations, tissue and serum deconvolution, mass spectrometry, and immunoassay. Effects of clinical characteristics on temporal changes and outcomes were studied. Among 47 patients, 8 (17.0%) had non-matching classifications at recurrence. In the remaining 39 cases, 28.2% showed dominant DNA methylation subclass transitions, with 72.7% being a mesenchymal subclass. In general, glioblastomas with a subclass transition showed upregulated metabolic processes. Newly diagnosed glioblastomas with mesenchymal transition displayed increased stem cell-like states and decreased immune components at diagnosis and exhibited elevated immune signatures and cytokine levels in serum. In contrast, tissue of recurrent glioblastomas with mesenchymal transition showed increased immune components but decreased stem cell-like states. Survival analyses revealed comparable outcomes for patients with and without subclass transitions. This study demonstrates a temporal heterogeneity of DNA methylation subclasses in 28.2% of glioblastomas, not impacting patient survival. Changes in cell state composition associated with subclass transition may be crucial for recurrent glioblastoma targeted therapies.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Humanos , Glioblastoma/genética , Glioblastoma/terapia , Metilação de DNA , Recidiva Local de Neoplasia/genética , Análise de Sobrevida
3.
bioRxiv ; 2023 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-37609137

RESUMO

Neural-tumor interactions drive glioma growth as evidenced in preclinical models, but clinical validation is nascent. We present an epigenetically defined neural signature of glioblastoma that independently affects patients' survival. We use reference signatures of neural cells to deconvolve tumor DNA and classify samples into low- or high-neural tumors. High-neural glioblastomas exhibit hypomethylated CpG sites and upregulation of genes associated with synaptic integration. Single-cell transcriptomic analysis reveals high abundance of stem cell-like malignant cells classified as oligodendrocyte precursor and neural precursor cell-like in high-neural glioblastoma. High-neural glioblastoma cells engender neuron-to-glioma synapse formation in vitro and in vivo and show an unfavorable survival after xenografting. In patients, a high-neural signature associates with decreased survival as well as increased functional connectivity and can be detected via DNA analytes and brain-derived neurotrophic factor in plasma. Our study presents an epigenetically defined malignant neural signature in high-grade gliomas that is prognostically relevant.

4.
Nat Genet ; 53(10): 1469-1479, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34594037

RESUMO

Single-cell RNA sequencing has revealed extensive transcriptional cell state diversity in cancer, often observed independently of genetic heterogeneity, raising the central question of how malignant cell states are encoded epigenetically. To address this, here we performed multiomics single-cell profiling-integrating DNA methylation, transcriptome and genotype within the same cells-of diffuse gliomas, tumors characterized by defined transcriptional cell state diversity. Direct comparison of the epigenetic profiles of distinct cell states revealed key switches for state transitions recapitulating neurodevelopmental trajectories and highlighted dysregulated epigenetic mechanisms underlying gliomagenesis. We further developed a quantitative framework to directly measure cell state heritability and transition dynamics based on high-resolution lineage trees in human samples. We demonstrated heritability of malignant cell states, with key differences in hierarchal and plastic cell state architectures in IDH-mutant glioma versus IDH-wild-type glioblastoma, respectively. This work provides a framework anchoring transcriptional cancer cell states in their epigenetic encoding, inheritance and transition dynamics.


Assuntos
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Plasticidade Celular/genética , Epigênese Genética , Glioma/genética , Glioma/patologia , Padrões de Herança/genética , Transcrição Gênica , Linhagem Celular Tumoral , Ilhas de CpG/genética , Variações do Número de Cópias de DNA/genética , Metilação de DNA/genética , Humanos , Isocitrato Desidrogenase/genética , Filogenia , Complexo Repressor Polycomb 2/metabolismo , Regiões Promotoras Genéticas/genética , Análise de Célula Única , Transcriptoma/genética
5.
Elife ; 102021 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-34169837

RESUMO

Mortality from breast cancer is almost exclusively a result of tumor metastasis, and lungs are one of the main metastatic sites. Cancer-associated fibroblasts are prominent players in the microenvironment of breast cancer. However, their role in the metastatic niche is largely unknown. In this study, we profiled the transcriptional co-evolution of lung fibroblasts isolated from transgenic mice at defined stage-specific time points of metastases formation. Employing multiple knowledge-based platforms of data analysis provided powerful insights on functional and temporal regulation of the transcriptome of fibroblasts. We demonstrate that fibroblasts in lung metastases are transcriptionally dynamic and plastic, and reveal stage-specific gene signatures that imply functional tasks, including extracellular matrix remodeling, stress response, and shaping the inflammatory microenvironment. Furthermore, we identified Myc as a central regulator of fibroblast rewiring and found that stromal upregulation of Myc transcriptional networks is associated with disease progression in human breast cancer.


Assuntos
Fibroblastos/patologia , Neoplasias Pulmonares/secundário , Pulmão/patologia , Transcriptoma , Microambiente Tumoral/genética , Animais , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Feminino , Camundongos , Camundongos Transgênicos
6.
Bioinformatics ; 37(3): 326-333, 2021 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-32805010

RESUMO

MOTIVATION: In recent years, the well-known Infinite Sites Assumption has been a fundamental feature of computational methods devised for reconstructing tumor phylogenies and inferring cancer progressions. However, recent studies leveraging single-cell sequencing (SCS) techniques have shown evidence of the widespread recurrence and, especially, loss of mutations in several tumor samples. While there exist established computational methods that infer phylogenies with mutation losses, there remain some advancements to be made. RESULTS: We present Simulated Annealing Single-Cell inference (SASC): a new and robust approach based on simulated annealing for the inference of cancer progression from SCS datasets. In particular, we introduce an extension of the model of evolution where mutations are only accumulated, by allowing also a limited amount of mutation loss in the evolutionary history of the tumor: the Dollo-k model. We demonstrate that SASC achieves high levels of accuracy when tested on both simulated and real datasets and in comparison with some other available methods. AVAILABILITY AND IMPLEMENTATION: The SASC tool is open source and available at https://github.com/sciccolella/sasc. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Assuntos
Algoritmos , Neoplasias , Análise de Célula Única , Humanos , Mutação , Neoplasias/genética , Filogenia , Análise de Sequência , Software
7.
Nature ; 573(7775): 539-545, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31534222

RESUMO

High-grade gliomas are lethal brain cancers whose progression is robustly regulated by neuronal activity. Activity-regulated release of growth factors promotes glioma growth, but this alone is insufficient to explain the effect that neuronal activity exerts on glioma progression. Here we show that neuron and glioma interactions include electrochemical communication through bona fide AMPA receptor-dependent neuron-glioma synapses. Neuronal activity also evokes non-synaptic activity-dependent potassium currents that are amplified by gap junction-mediated tumour interconnections, forming an electrically coupled network. Depolarization of glioma membranes assessed by in vivo optogenetics promotes proliferation, whereas pharmacologically or genetically blocking electrochemical signalling inhibits the growth of glioma xenografts and extends mouse survival. Emphasizing the positive feedback mechanisms by which gliomas increase neuronal excitability and thus activity-regulated glioma growth, human intraoperative electrocorticography demonstrates increased cortical excitability in the glioma-infiltrated brain. Together, these findings indicate that synaptic and electrical integration into neural circuits promotes glioma progression.


Assuntos
Encéfalo/fisiopatologia , Sinapses Elétricas/patologia , Fenômenos Eletrofisiológicos , Glioma/fisiopatologia , Animais , Encéfalo/citologia , Membrana Celular/patologia , Proliferação de Células , Junções Comunicantes/patologia , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Xenoenxertos , Humanos , Camundongos , Camundongos Endogâmicos NOD , Neurônios/patologia , Optogenética , Potássio/metabolismo , Transmissão Sináptica , Células Tumorais Cultivadas
8.
Cell ; 178(4): 835-849.e21, 2019 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-31327527

RESUMO

Diverse genetic, epigenetic, and developmental programs drive glioblastoma, an incurable and poorly understood tumor, but their precise characterization remains challenging. Here, we use an integrative approach spanning single-cell RNA-sequencing of 28 tumors, bulk genetic and expression analysis of 401 specimens from the The Cancer Genome Atlas (TCGA), functional approaches, and single-cell lineage tracing to derive a unified model of cellular states and genetic diversity in glioblastoma. We find that malignant cells in glioblastoma exist in four main cellular states that recapitulate distinct neural cell types, are influenced by the tumor microenvironment, and exhibit plasticity. The relative frequency of cells in each state varies between glioblastoma samples and is influenced by copy number amplifications of the CDK4, EGFR, and PDGFRA loci and by mutations in the NF1 locus, which each favor a defined state. Our work provides a blueprint for glioblastoma, integrating the malignant cell programs, their plasticity, and their modulation by genetic drivers.


Assuntos
Neoplasias Encefálicas/genética , Plasticidade Celular/genética , Glioblastoma/genética , Adolescente , Idoso , Animais , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Linhagem da Célula/genética , Criança , Estudos de Coortes , Modelos Animais de Doenças , Feminino , Heterogeneidade Genética , Glioblastoma/patologia , Xenoenxertos , Humanos , Lactente , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Pessoa de Meia-Idade , Mutação , RNA-Seq , Análise de Célula Única/métodos , Microambiente Tumoral/genética
9.
Nat Commun ; 10(1): 3015, 2019 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-31289271

RESUMO

The protein-protein interaction (PPI) network of an organism serves as a skeleton for its signaling circuitry, which mediates cellular response to environmental and genetic cues. Understanding this circuitry could improve the prediction of gene function and cellular behavior in response to diverse signals. To realize this potential, one has to comprehensively map PPIs and their directions of signal flow. While the quality and the volume of identified human PPIs improved dramatically over the last decade, the directions of these interactions are still mostly unknown, thus precluding subsequent prediction and modeling efforts. Here we present a systematic approach to orient the human PPI network using drug response and cancer genomic data. We provide a diffusion-based method for the orientation task that significantly outperforms existing methods. The oriented network leads to improved prioritization of cancer driver genes and drug targets compared to the state-of-the-art unoriented network.


Assuntos
Biologia Computacional/métodos , Neoplasias/genética , Mapeamento de Interação de Proteínas/métodos , Mapas de Interação de Proteínas/efeitos dos fármacos , Análise de Dados , Bases de Dados Genéticas/estatística & dados numéricos , Bases de Dados de Produtos Farmacêuticos/estatística & dados numéricos , Conjuntos de Dados como Assunto , Humanos , Mapas de Interação de Proteínas/genética , Software
10.
Cell Syst ; 8(5): 456-466.e5, 2019 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-31103572

RESUMO

The identification of molecular pathways driving cancer progression is a fundamental challenge in cancer research. Most approaches to address it are limited in the number of data types they employ and perform data integration in a sequential manner. Here, we describe ModulOmics, a method to de novo identify cancer driver pathways, or modules, by integrating protein-protein interactions, mutual exclusivity of mutations and copy number alterations, transcriptional coregulation, and RNA coexpression into a single probabilistic model. To efficiently search and score the large space of candidate modules, ModulOmics employs a two-step optimization procedure that combines integer linear programming with stochastic search. Applied across several cancer types, ModulOmics identifies highly functionally connected modules enriched with cancer driver genes, outperforming state-of-the-art methods and demonstrating the power of using multiple omics data types simultaneously. On breast cancer subtypes, ModulOmics proposes unexplored connections supported by an independent patient cohort and independent proteomic and phosphoproteomic datasets.


Assuntos
Biologia Computacional/métodos , Neoplasias/genética , Neoplasias/metabolismo , Algoritmos , Neoplasias da Mama/genética , Variações do Número de Cópias de DNA , Perfilação da Expressão Gênica/métodos , Redes Reguladoras de Genes , Genômica/métodos , Humanos , Modelos Estatísticos , Mutação , Proteômica/métodos , Transdução de Sinais/genética , Software
11.
Cancer Res ; 77(4): 827-838, 2017 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-27965317

RESUMO

Personalized therapy is a major goal of modern oncology, as patient responses vary greatly even within a histologically defined cancer subtype. This is especially true in acute myeloid leukemia (AML), which exhibits striking heterogeneity in molecular segmentation. When calibrated to cell-specific data, executable network models can reveal subtle differences in signaling that help explain differences in drug response. Furthermore, they can suggest drug combinations to increase efficacy and combat acquired resistance. Here, we experimentally tested dynamic proteomic changes and phenotypic responses in diverse AML cell lines treated with pan-PIM kinase inhibitor and fms-related tyrosine kinase 3 (FLT3) inhibitor as single agents and in combination. We constructed cell-specific executable models of the signaling axis, connecting genetic aberrations in FLT3, tyrosine kinase 2 (TYK2), platelet-derived growth factor receptor alpha (PDGFRA), and fibroblast growth factor receptor 1 (FGFR1) to cell proliferation and apoptosis via the PIM and PI3K kinases. The models capture key differences in signaling that later enabled them to accurately predict the unique proteomic changes and phenotypic responses of each cell line. Furthermore, using cell-specific models, we tailored combination therapies to individual cell lines and successfully validated their efficacy experimentally. Specifically, we showed that cells mildly responsive to PIM inhibition exhibited increased sensitivity in combination with PIK3CA inhibition. We also used the model to infer the origin of PIM resistance engineered through prolonged drug treatment of MOLM16 cell lines and successfully validated experimentally our prediction that this resistance can be overcome with AKT1/2 inhibition. Cancer Res; 77(4); 827-38. ©2016 AACR.


Assuntos
Leucemia Mieloide Aguda/tratamento farmacológico , Proteínas Proto-Oncogênicas c-pim-1/antagonistas & inibidores , Transdução de Sinais/fisiologia , Tirosina Quinase 3 Semelhante a fms/antagonistas & inibidores , Compostos de Bifenilo/uso terapêutico , Linhagem Celular Tumoral , Classe I de Fosfatidilinositol 3-Quinases , Simulação por Computador , Resistencia a Medicamentos Antineoplásicos , Quimioterapia Combinada , Humanos , Leucemia Mieloide Aguda/fisiopatologia , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Proto-Oncogênicas c-pim-1/fisiologia , Transdução de Sinais/efeitos dos fármacos , Tiazolidinas/uso terapêutico
12.
Pac Symp Biocomput ; 21: 156-67, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26776182

RESUMO

We present a computational strategy to simulate drug treatment in a personalized setting. The method is based on integrating patient mutation and differential expression data with a protein-protein interaction network. We test the impact of in-silico deletions of different proteins on the flow of information in the network and use the results to infer potential drug targets. We apply our method to AML data from TCGA and validate the predicted drug targets using known targets. To benchmark our patient-specific approach, we compare the personalized setting predictions to those of the conventional setting. Our predicted drug targets are highly enriched with known targets from DrugBank and COSMIC (p < 10(-5) outperforming the non-personalized predictions. Finally, we focus on the largest AML patient subgroup (~30%) which is characterized by an FLT3 mutation, and utilize our prediction score to rank patient sensitivity to inhibition of each predicted target, reproducing previous findings of in-vitro experiments.


Assuntos
Descoberta de Drogas/métodos , Medicina de Precisão/métodos , Algoritmos , Antineoplásicos/farmacologia , Biologia Computacional/métodos , Biologia Computacional/estatística & dados numéricos , Simulação por Computador , Bases de Dados de Produtos Farmacêuticos/estatística & dados numéricos , Descoberta de Drogas/estatística & dados numéricos , Redes Reguladoras de Genes , Humanos , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Mutação , Medicina de Precisão/estatística & dados numéricos , Mapas de Interação de Proteínas/efeitos dos fármacos , Tirosina Quinase 3 Semelhante a fms/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA