Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros











Intervalo de año de publicación
1.
Cell Rep Methods ; 4(2): 100708, 2024 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-38412834

RESUMEN

Tumor deconvolution enables the identification of diverse cell types that comprise solid tumors. To date, however, both the algorithms developed to deconvolve tumor samples, and the gold-standard datasets used to assess the algorithms are geared toward the analysis of gene expression (e.g., RNA sequencing) rather than protein levels. Despite the popularity of gene expression datasets, protein levels often provide a more accurate view of rare cell types. To facilitate the use, development, and reproducibility of multiomic deconvolution algorithms, we introduce Decomprolute, a Common Workflow Language framework that leverages containerization to compare tumor deconvolution algorithms across multiomic datasets. Decomprolute incorporates the large-scale multiomic datasets produced by the Clinical Proteomic Tumor Analysis Consortium (CPTAC), which include matched mRNA expression and proteomic data from thousands of tumors across multiple cancer types to build a fully open-source, containerized proteogenomic tumor deconvolution benchmarking platform. http://pnnl-compbio.github.io/decomprolute.


Asunto(s)
Neoplasias , Proteómica , Humanos , Multiómica , Benchmarking , Reproducibilidad de los Resultados , Neoplasias/genética
2.
Cancer Cell ; 41(8): 1397-1406, 2023 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-37582339

RESUMEN

The National Cancer Institute's Clinical Proteomic Tumor Analysis Consortium (CPTAC) investigates tumors from a proteogenomic perspective, creating rich multi-omics datasets connecting genomic aberrations to cancer phenotypes. To facilitate pan-cancer investigations, we have generated harmonized genomic, transcriptomic, proteomic, and clinical data for >1000 tumors in 10 cohorts to create a cohesive and powerful dataset for scientific discovery. We outline efforts by the CPTAC pan-cancer working group in data harmonization, data dissemination, and computational resources for aiding biological discoveries. We also discuss challenges for multi-omics data integration and analysis, specifically the unique challenges of working with both nucleotide sequencing and mass spectrometry proteomics data.


Asunto(s)
Neoplasias , Proteogenómica , Humanos , Proteómica , Genómica , Neoplasias/genética , Perfilación de la Expresión Génica
3.
Cell ; 186(18): 3921-3944.e25, 2023 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-37582357

RESUMEN

Cancer driver events refer to key genetic aberrations that drive oncogenesis; however, their exact molecular mechanisms remain insufficiently understood. Here, our multi-omics pan-cancer analysis uncovers insights into the impacts of cancer drivers by identifying their significant cis-effects and distal trans-effects quantified at the RNA, protein, and phosphoprotein levels. Salient observations include the association of point mutations and copy-number alterations with the rewiring of protein interaction networks, and notably, most cancer genes converge toward similar molecular states denoted by sequence-based kinase activity profiles. A correlation between predicted neoantigen burden and measured T cell infiltration suggests potential vulnerabilities for immunotherapies. Patterns of cancer hallmarks vary by polygenic protein abundance ranging from uniform to heterogeneous. Overall, our work demonstrates the value of comprehensive proteogenomics in understanding the functional states of oncogenic drivers and their links to cancer development, surpassing the limitations of studying individual cancer types.


Asunto(s)
Neoplasias , Proteogenómica , Humanos , Neoplasias/genética , Oncogenes , Transformación Celular Neoplásica/genética , Variaciones en el Número de Copia de ADN
4.
Cancer Cell ; 39(4): 509-528.e20, 2021 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-33577785

RESUMEN

Glioblastoma (GBM) is the most aggressive nervous system cancer. Understanding its molecular pathogenesis is crucial to improving diagnosis and treatment. Integrated analysis of genomic, proteomic, post-translational modification and metabolomic data on 99 treatment-naive GBMs provides insights to GBM biology. We identify key phosphorylation events (e.g., phosphorylated PTPN11 and PLCG1) as potential switches mediating oncogenic pathway activation, as well as potential targets for EGFR-, TP53-, and RB1-altered tumors. Immune subtypes with distinct immune cell types are discovered using bulk omics methodologies, validated by snRNA-seq, and correlated with specific expression and histone acetylation patterns. Histone H2B acetylation in classical-like and immune-low GBM is driven largely by BRDs, CREBBP, and EP300. Integrated metabolomic and proteomic data identify specific lipid distributions across subtypes and distinct global metabolic changes in IDH-mutated tumors. This work highlights biological relationships that could contribute to stratification of GBM patients for more effective treatment.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Glioblastoma/genética , Glioblastoma/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Proteogenómica , Neoplasias Encefálicas/patología , Biología Computacional/métodos , Glioblastoma/patología , Humanos , Metabolómica/métodos , Mutación/genética , Fosfolipasa C gamma/genética , Fosfolipasa C gamma/metabolismo , Fosforilación/fisiología , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Proteogenómica/métodos , Proteómica/métodos
5.
Glia ; 68(12): 2486-2502, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32621641

RESUMEN

Radiation therapy is part of the standard of care for gliomas and kills a subset of tumor cells, while also altering the tumor microenvironment. Tumor cells with stem-like properties preferentially survive radiation and give rise to glioma recurrence. Various techniques for enriching and quantifying cells with stem-like properties have been used, including the fluorescence activated cell sorting (FACS)-based side population (SP) assay, which is a functional assay that enriches for stem-like tumor cells. In these analyses, mouse models of glioma have been used to understand the biology of this disease and therapeutic responses, including the radiation response. We present combined SP analysis and single-cell RNA sequencing of genetically-engineered mouse models of glioma to show a time course of cellular response to radiation. We identify and characterize two distinct tumor cell populations that are inherently radioresistant and also distinct effects of radiation on immune cell populations within the tumor microenvironment.


Asunto(s)
Neoplasias Encefálicas , Glioma , Células Madre , Animales , Neoplasias Encefálicas/radioterapia , Ratones , Células Madre Neoplásicas , Análisis de la Célula Individual , Microambiente Tumoral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA