Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Hematol Oncol ; 16(1): 120, 2023 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-38102665

RESUMO

Global proteomic data generated by advanced mass spectrometry (MS) technologies can help bridge the gap between genome/transcriptome and functions and hold great potential in elucidating unbiased functional models of pro-tumorigenic pathways. To this end, we collected the high-throughput, whole-genome MS data and conducted integrative proteomic network analyses of 687 cases across 7 cancer types including breast carcinoma (115 tumor samples; 10,438 genes), clear cell renal carcinoma (100 tumor samples; 9,910 genes), colorectal cancer (91 tumor samples; 7,362 genes), hepatocellular carcinoma (101 tumor samples; 6,478 genes), lung adenocarcinoma (104 tumor samples; 10,967 genes), stomach adenocarcinoma (80 tumor samples; 9,268 genes), and uterine corpus endometrial carcinoma UCEC (96 tumor samples; 10,768 genes). Through the protein co-expression network analysis, we identified co-expressed protein modules enriched for differentially expressed proteins in tumor as disease-associated pathways. Comparison with the respective transcriptome network models revealed proteome-specific cancer subnetworks associated with heme metabolism, DNA repair, spliceosome, oxidative phosphorylation and several oncogenic signaling pathways. Cross-cancer comparison identified highly preserved protein modules showing robust pan-cancer interactions and identified endoplasmic reticulum-associated degradation (ERAD) and N-acetyltransferase activity as the central functional axes. We further utilized these network models to predict pan-cancer protein regulators of disease-associated pathways. The top predicted pan-cancer regulators including RSL1D1, DDX21 and SMC2, were experimentally validated in lung, colon, breast cancer and fetal kidney cells. In summary, this study has developed interpretable network models of cancer proteomes, showcasing their potential in unveiling novel oncogenic regulators, elucidating underlying mechanisms, and identifying new therapeutic targets.


Assuntos
Adenocarcinoma , Neoplasias Renais , Neoplasias Hepáticas , Neoplasias Pulmonares , Proteínas da Gravidez , Humanos , Proteômica , Degradação Associada com o Retículo Endoplasmático , Perfilação da Expressão Gênica/métodos , Adenocarcinoma/genética , Neoplasias Pulmonares/genética , Proteínas da Gravidez/genética , Proteínas Ribossômicas/genética , RNA Helicases DEAD-box/genética
2.
bioRxiv ; 2023 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-38014015

RESUMO

Cancer mutations are often assumed to alter proteins, thus promoting tumorigenesis. However, how mutations affect protein expression has rarely been systematically investigated. We conduct a comprehensive analysis of mutation impacts on mRNA- and protein-level expressions of 953 cancer cases with paired genomics and global proteomic profiling across six cancer types. Protein-level impacts are validated for 47.2% of the somatic expression quantitative trait loci (seQTLs), including mutations from likely "long-tail" driver genes. Devising a statistical pipeline for identifying somatic protein-specific QTLs (spsQTLs), we reveal several gene mutations, including NF1 and MAP2K4 truncations and TP53 missenses showing disproportional influence on protein abundance not readily explained by transcriptomics. Cross-validating with data from massively parallel assays of variant effects (MAVE), TP53 missenses associated with high tumor TP53 proteins were experimentally confirmed as functional. Our study demonstrates the importance of considering protein-level expression to validate mutation impacts and identify functional genes and mutations.

3.
bioRxiv ; 2023 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-37502974

RESUMO

Tumor mutations can influence the surrounding microenvironment leading to suppression of anti-tumor immune responses and thereby contributing to tumor progression and failure of cancer therapies. Here we use genetically engineered lung cancer mouse models and patient samples to dissect how LKB1 mutations accelerate tumor growth by reshaping the immune microenvironment. Comprehensive immune profiling of LKB1 -mutant vs wildtype tumors revealed dramatic changes in myeloid cells, specifically enrichment of Arg1 + interstitial macrophages and SiglecF Hi neutrophils. We discovered a novel mechanism whereby autocrine LIF signaling in Lkb1 -mutant tumors drives tumorigenesis by reprogramming myeloid cells in the immune microenvironment. Inhibiting LIF signaling in Lkb1 -mutant tumors, via gene targeting or with a neutralizing antibody, resulted in a striking reduction in Arg1 + interstitial macrophages and SiglecF Hi neutrophils, expansion of antigen specific T cells, and inhibition of tumor progression. Thus, targeting LIF signaling provides a new therapeutic approach to reverse the immunosuppressive microenvironment of LKB1 -mutant tumors.

4.
bioRxiv ; 2023 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-36865220

RESUMO

Structural features of proteins capture underlying information about protein evolution and function, which enhances the analysis of proteomic and transcriptomic data. Here we develop Structural Analysis of Gene and protein Expression Signatures (SAGES), a method that describes expression data using features calculated from sequence-based prediction methods and 3D structural models. We used SAGES, along with machine learning, to characterize tissues from healthy individuals and those with breast cancer. We analyzed gene expression data from 23 breast cancer patients and genetic mutation data from the COSMIC database as well as 17 breast tumor protein expression profiles. We identified prominent expression of intrinsically disordered regions in breast cancer proteins as well as relationships between drug perturbation signatures and breast cancer disease signatures. Our results suggest that SAGES is generally applicable to describe diverse biological phenomena including disease states and drug effects.

5.
Cancer Immunol Immunother ; 72(6): 1893-1901, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36707424

RESUMO

PURPOSE: While immune checkpoint inhibitors (ICI) have had success with various malignancies, their efficacy in brain cancer is still unclear. Retrospective and prospective studies using PD-1 inhibitors for recurrent glioblastoma (GBM) have not established survival benefit. This study evaluated if ICI may be effective for select patients with recurrent GBM. METHODS: This was a single-center retrospective study of adult patients diagnosed with first recurrence GBM and received pembrolizumab or nivolumab with or without concurrent bevacizumab. Archival tissue was used for immunohistochemistry (IHC) and targeted DNA next-generation sequencing (NGS) analysis. RESULTS: Median overall survival (mOS) from initial diagnosis was 24.5 months (range 10-42). mOS from onset of ICI was 10 months (range 1-31) with 75% surviving > 6 months and 46% > 12 months. Additional IHC analysis on tumors from eight patients demonstrated a trend of longer survival after ICI for those with elevated PD-L1 expression. NGS of samples from 15 patients identified EGFR amplification at initial diagnosis and at any time point to be associated with worse survival after ICI (HR 12.2, 95% CI 1.37-108, p = 0.025 and HR 3.92, 95% CI 1.03-14.9, p = 0.045, respectively). This significance was corroborated with previously tested EGFR amplification via in situ hybridization. CONCLUSION: ICI did not extend overall survival for recurrent GBM. However, molecular sequencing identified EGFR amplification as associated with worse survival. Prospective studies can validate if EGFR amplification is a biomarker of ICI resistance and determine if its use can stratify responders from non-responders.


Assuntos
Antineoplásicos Imunológicos , Glioblastoma , Adulto , Humanos , Glioblastoma/tratamento farmacológico , Glioblastoma/genética , Glioblastoma/metabolismo , Inibidores de Checkpoint Imunológico/uso terapêutico , Estudos Retrospectivos , Antineoplásicos Imunológicos/uso terapêutico , Estudos Prospectivos , Recidiva Local de Neoplasia/tratamento farmacológico , Receptores ErbB/genética
6.
Front Oncol ; 12: 814120, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35433463

RESUMO

Hepatocellular carcinoma (HCC) is the fourth cause of cancer-related mortality worldwide. While many targeted therapies have been developed, the majority of HCC tumors do not harbor clinically actionable mutations. Protein-level aberrations, especially those not evident at the genomic level, present therapeutic opportunities but have rarely been systematically characterized in HCC. In this study, we performed proteogenomic analyses of 260 primary tumors from two HBV-related HCC patient cohorts with global mass-spectrometry (MS) proteomics data. Combining tumor-normal and inter-tumor analyses, we identified overexpressed targets including PDGFRB, FGFR4, ERBB2/3, CDK6 kinases and MFAP5, HMCN1, and Hsp proteins in HCC, many of which showed low frequencies of genomic and/or transcriptomic aberrations. Protein expression of FGFR4 kinase and Hsp proteins were significantly associated with response to their corresponding inhibitors. Our results provide a catalog of protein targets in HCC and demonstrate the potential of proteomics approaches in advancing precision medicine in cancer types lacking druggable mutations.

7.
Cell Rep ; 37(7): 110005, 2021 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-34788626

RESUMO

Young adult cancer has increased in incidence worldwide, but its molecular etiologies remain unclear. We systematically characterize genomic profiles of young adult tumors with ages of onset ≤50 years and compare them to later-onset tumors using over 6,000 cases across 14 cancer types. While young adult tumors generally show lower mutation burdens and comparable copy-number variation rates compared to later-onset cases, they are enriched for multiple driver mutations and copy-number alterations in subtype-specific contexts. Characterization of tumor immune microenvironments reveals pan-cancer patterns of elevated TGF-ß response/dendritic cells and lower IFN-γ response/macrophages relative to later-onset tumors, corresponding to age-related responses to immunotherapy in several cancer types. Finally, we identify prevalent clinically actionable events that disproportionally affect young adult or later-onset cases. The resulting catalog of age-related molecular drivers can guide precision diagnostics and treatments for young adult cancer.


Assuntos
Fatores Etários , Neoplasias/diagnóstico , Neoplasias/genética , Adulto , Idoso , Variações do Número de Cópias de DNA/genética , Metilação de DNA , Bases de Dados Genéticas , Epigênese Genética/genética , Expressão Gênica/genética , Regulação Neoplásica da Expressão Gênica/genética , Genômica/métodos , Humanos , Imunoterapia/métodos , Pessoa de Meia-Idade , Mutação/genética , Neoplasias/fisiopatologia , Microambiente Tumoral/genética , Microambiente Tumoral/imunologia , Adulto Jovem
8.
Cell Rep ; 36(12): 109729, 2021 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-34551295

RESUMO

Human ubiquinol-cytochrome c reductase core protein 1 (UQCRC1) is an evolutionarily conserved core subunit of mitochondrial respiratory chain complex III. We recently identified the disease-associated variants of UQCRC1 from patients with familial parkinsonism, but its function remains unclear. Here we investigate the endogenous function of UQCRC1 in the human neuronal cell line and the Drosophila nervous system. Flies with neuronal knockdown of uqcrc1 exhibit age-dependent parkinsonism-resembling defects, including dopaminergic neuron reduction and locomotor decline, and are ameliorated by UQCRC1 expression. Lethality of uqcrc1-KO is also rescued by neuronally expressing UQCRC1, but not the disease-causing variant, providing a platform to discern the pathogenicity of this mutation. Furthermore, UQCRC1 associates with the apoptosis trigger cytochrome c (cyt-c), and uqcrc1 deficiency increases cyt-c in the cytoplasmic fraction and activates the caspase cascade. Depleting cyt-c or expression of the anti-apoptotic p35 ameliorates uqcrc1-mediated neurodegeneration. Our findings identify a role for UQCRC1 in regulating cyt-c-induced apoptosis.


Assuntos
Neurônios Dopaminérgicos/metabolismo , Proteínas de Drosophila/metabolismo , Complexo III da Cadeia de Transporte de Elétrons/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Apoptose , Linhagem Celular Tumoral , Citocromos c/metabolismo , Citoplasma/metabolismo , Neurônios Dopaminérgicos/citologia , Drosophila/crescimento & desenvolvimento , Drosophila/metabolismo , Proteínas de Drosophila/genética , Complexo III da Cadeia de Transporte de Elétrons/deficiência , Complexo III da Cadeia de Transporte de Elétrons/genética , Edição de Genes , Humanos , Larva/metabolismo , Locomoção , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Transtornos Parkinsonianos/metabolismo , Transtornos Parkinsonianos/patologia , Ligação Proteica , Interferência de RNA , Espécies Reativas de Oxigênio/metabolismo
9.
Commun Biol ; 4(1): 1112, 2021 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-34552204

RESUMO

Identifying genomic alterations of cancer proteins has guided the development of targeted therapies, but proteomic analyses are required to validate and reveal new treatment opportunities. Herein, we develop a new algorithm, OPPTI, to discover overexpressed kinase proteins across 10 cancer types using global mass spectrometry proteomics data of 1,071 cases. OPPTI outperforms existing methods by leveraging multiple co-expressed markers to identify targets overexpressed in a subset of tumors. OPPTI-identified overexpression of ERBB2 and EGFR proteins correlates with genomic amplifications, while CDK4/6, PDK1, and MET protein overexpression frequently occur without corresponding DNA- and RNA-level alterations. Analyzing CRISPR screen data, we confirm expression-driven dependencies of multiple currently-druggable and new target kinases whose expressions are validated by immunochemistry. Identified kinases are further associated with up-regulated phosphorylation levels of corresponding signaling pathways. Collectively, our results reveal protein-level aberrations-sometimes not observed by genomics-represent cancer vulnerabilities that may be targeted in precision oncology.


Assuntos
Regulação Neoplásica da Expressão Gênica , Neoplasias/genética , Proteínas Quinases/genética , Proteogenômica/métodos , Regulação para Cima , Adulto , Idoso , Algoritmos , Criança , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias/fisiopatologia , Fosforilação , Proteínas Quinases/metabolismo , Transdução de Sinais
10.
Cancers (Basel) ; 13(18)2021 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-34572800

RESUMO

Germline BRCA1/2 mutations associated with HRD are clinical biomarkers for sensitivity to poly-ADP ribose polymerase inhibitors (PARPi) treatment in breast, ovarian, pancreatic, and prostate cancers. However, it remains unclear whether other mutations may also lead to HRD and PARPi sensitivity across a broader range of cancer types. Our goal was to determine the germline or somatic alterations associated with the HRD phenotype that might therefore confer PARPi sensitivity. Using germline and somatic genomic data from over 9000 tumors representing 32 cancer types, we examined associations between HRD scores and pathogenic germline variants, somatic driver mutations, and copy number deletions in 30 candidate genes involved in homologous recombination. We identified several germline and somatic mutations (e.g., BRCA1/2, PALB2, ATM, and ATR mutations) associated with HRD phenotype in ovarian, breast, pancreatic, stomach, bladder, and lung cancer. The co-occurrence of germline BRCA1 variants and somatic TP53 mutations was significantly associated with increasing HRD in breast cancer. Notably, we also identified multiple somatic copy number deletions associated with HRD. Our study suggests that multiple cancer types include tumor subsets that show HRD phenotype and should be considered in the future clinical studies of PARPi and synthetic lethality strategies exploiting HRD, which can be caused by a large number of genomic alterations.

11.
Genome Med ; 13(1): 147, 2021 09 09.
Artigo em Inglês | MEDLINE | ID: mdl-34503567

RESUMO

BACKGROUND: DNA sequencing is increasingly incorporated into the routine care of cancer patients, many of whom also carry inherited, moderate/high-penetrance variants associated with other diseases. Yet, the prevalence and consequence of such variants remain unclear. METHODS: We analyzed the germline genomes of 10,389 adult cancer cases in the TCGA cohort, identifying pathogenic/likely pathogenic variants in autosomal-dominant genes, autosomal-recessive genes, and 59 medically actionable genes curated by the American College of Molecular Genetics (i.e., the ACMG 59 genes). We also analyzed variant- and gene-level expression consequences in carriers. RESULTS: The affected genes exhibited varying pan-ancestry and population-specific patterns, and overall, the European population showed the highest frequency of pathogenic/likely pathogenic variants. We further identified genes showing expression consequence supporting variant functionality, including altered gene expression, allelic specific expression, and mis-splicing determined by a massively parallel splicing assay. CONCLUSIONS: Our results demonstrate that expression-altering variants are found in a substantial fraction of cases and illustrate the yield of genomic risk assessments for a wide range of diseases across diverse populations.


Assuntos
Células Germinativas , Neoplasias , Humanos , Alelos , Regulação Neoplásica da Expressão Gênica , Genômica , Heterozigoto , Padrões de Herança , Neoplasias/genética , Medição de Risco , Análise de Sequência de DNA
12.
Sci Rep ; 11(1): 13913, 2021 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-34230510

RESUMO

The global surge in COVID-19 cases underscores the need for fast, scalable, and reliable testing. Current COVID-19 diagnostic tests are limited by turnaround time, limited availability, or occasional false findings. Here, we developed a machine learning-based framework for predicting individual COVID-19 positive diagnosis relying only on readily-available baseline data, including patient demographics, comorbidities, and common lab values. Leveraging a cohort of 31,739 adults within an academic health system, we trained and tested multiple types of machine learning models, achieving an area under the curve of 0.75. Feature importance analyses highlighted serum calcium levels, temperature, age, lymphocyte count, smoking, hemoglobin levels, aspartate aminotransferase levels, and oxygen saturation as key predictors. Additionally, we developed a single decision tree model that provided an operable method for stratifying sub-populations. Overall, this study provides a proof-of-concept that COVID-19 diagnosis prediction models can be developed using only baseline data. The resulting prediction can complement existing tests to enhance screening and pandemic containment workflows.


Assuntos
Teste para COVID-19 , COVID-19/diagnóstico , Demografia , SARS-CoV-2/patogenicidade , Adulto , COVID-19/epidemiologia , Teste para COVID-19/métodos , Estudos de Coortes , Demografia/métodos , Humanos , Aprendizado de Máquina , Prognóstico , Curva ROC
13.
Sci Rep ; 11(1): 12107, 2021 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-34103633

RESUMO

Effective treatments targeting disease etiology are urgently needed for Alzheimer's disease (AD). Although candidate AD genes have been identified and altering their levels may serve as therapeutic strategies, the consequence of such alterations remain largely unknown. Herein, we analyzed CRISPR knockout/RNAi knockdown screen data for over 700 cell lines and evaluated cellular dependencies of 104 AD-associated genes previously identified by genome-wide association studies (GWAS) and gene expression network studies. Multiple genes showed widespread cell dependencies across tissue lineages, suggesting their inhibition may yield off-target effects. Meanwhile, several genes including SPI1, MEF2C, GAB2, ABCC11, ATCG1 were identified as genes of interest since their genetic knockouts specifically affected high-expressing cells whose tissue lineages are relevant to cell types found in AD. Overall, analyses of genetic screen data identified AD-associated genes whose knockout or knockdown selectively affected cell lines of relevant tissue lineages, prioritizing targets for potential AD treatments.


Assuntos
Doença de Alzheimer/genética , Doença de Alzheimer/fisiopatologia , Sistemas CRISPR-Cas , Predisposição Genética para Doença , Transportadores de Cassetes de Ligação de ATP/genética , Actinas/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Linhagem da Célula , Perfilação da Expressão Gênica , Redes Reguladoras de Genes , Estudo de Associação Genômica Ampla , Humanos , Fatores de Transcrição MEF2/genética , Microglia/metabolismo , Doenças do Sistema Nervoso/genética , Polimorfismo de Nucleotídeo Único , Proteínas Proto-Oncogênicas/genética , Interferência de RNA , Risco , Transativadores/genética
14.
Cancer Manag Res ; 13: 4351-4357, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34103990

RESUMO

The canonical model for hereditary cancer predisposition is a cancer predisposition gene (CPG) that drives either one or both of two fundamental hallmarks of cancer, defective genomic integrity and deregulated cell proliferation, ultimately resulting in the accumulation of mutations within cells. Thus, the genes most commonly associated with cancer-predisposing genetic syndromes are tumor suppressor genes that regulate DNA repair (eg, BRCA1, BRCA2, MMR genes) and/or cell cycle (eg, APC, RB1). In recent years, however, the spectrum of high-penetrance CPGs has expanded considerably to include genes in non-canonical pathways such as oncogenic signaling, metabolism, and protein translation. We propose here that, given the variety of pathways that may ultimately affect genome integrity and cell proliferation, the model of cancer genetic predisposition needs to be expanded to account for diverse mechanisms. This synthesis calls for modeling and multi-omic studies applying novel experimental and computational approaches to understand cancer genetic predisposition.

15.
Cell Rep Med ; 2(5): 100276, 2021 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-34095878

RESUMO

Tumors with DNA damage repair (DDR) deficiency accumulate genomic alterations that may serve as neoantigens and increase sensitivity to immune checkpoint inhibitor. However, over half of DDR-deficient tumors are refractory to immunotherapy, and it remains unclear which mutations may promote immunogenicity in which cancer types. We integrate deleterious somatic and germline mutations and methylation data of DDR genes in 10,080 cancers representing 32 cancer types and evaluate the associations of these alterations with tumor neoantigens and immune infiltrates. Our analyses identify DDR pathway mutations that are associated with higher neoantigen loads, adaptive immune markers, and survival outcomes of immune checkpoint inhibitor-treated animal models and patients. Different immune phenotypes are associated with distinct types of DDR deficiency, depending on the cancer type context. The comprehensive catalog of immune response-associated DDR deficiency may explain variations in immunotherapy outcomes across DDR-deficient cancers and facilitate the development of genomic biomarkers for immunotherapy.


Assuntos
Biomarcadores Tumorais/imunologia , Dano ao DNA/imunologia , Distúrbios no Reparo do DNA/imunologia , Reparo do DNA/imunologia , Neoplasias/genética , Biomarcadores Tumorais/genética , Dano ao DNA/genética , Reparo do DNA/genética , Distúrbios no Reparo do DNA/genética , Genômica/métodos , Humanos , Imunidade/genética , Imunoterapia/métodos , Mutação/genética
16.
Nat Commun ; 12(1): 2313, 2021 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-33875650

RESUMO

Advances in mass-spectrometry have generated increasingly large-scale proteomics datasets containing tens of thousands of phosphorylation sites (phosphosites) that require prioritization. We develop a bioinformatics tool called HotPho and systematically discover 3D co-clustering of phosphosites and cancer mutations on protein structures. HotPho identifies 474 such hybrid clusters containing 1255 co-clustering phosphosites, including RET p.S904/Y928, the conserved HRAS/KRAS p.Y96, and IDH1 p.Y139/IDH2 p.Y179 that are adjacent to recurrent mutations on protein structures not found by linear proximity approaches. Hybrid clusters, enriched in histone and kinase domains, frequently include expression-associated mutations experimentally shown as activating and conferring genetic dependency. Approximately 300 co-clustering phosphosites are verified in patient samples of 5 cancer types or previously implicated in cancer, including CTNNB1 p.S29/Y30, EGFR p.S720, MAPK1 p.S142, and PTPN12 p.S275. In summary, systematic 3D clustering analysis highlights nearly 3,000 likely functional mutations and over 1000 cancer phosphosites for downstream investigation and evaluation of potential clinical relevance.


Assuntos
Biologia Computacional/métodos , Mutação , Neoplasias/genética , Proteômica/métodos , Sítios de Ligação/genética , Análise por Conglomerados , Receptores ErbB/metabolismo , Humanos , Espectrometria de Massas/métodos , Neoplasias/metabolismo , Fosforilação , Proteína Tirosina Fosfatase não Receptora Tipo 12/metabolismo , beta Catenina/metabolismo
17.
Pac Symp Biocomput ; 26: 172-183, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33691015

RESUMO

Concurrently available genomic and transcriptomic data from large cohorts provide opportunities to discover expression quantitative trait loci (eQTLs)-genetic variants associated with gene expression changes. However, the statistical power of detecting rare variant eQTLs is often limited and most existing eQTL tools are not compatible with sequence variant file formats. We have developed AeQTL (Aggregated eQTL), a software tool that performs eQTL analysis on variants aggregated according to user-specified regions and is designed to accommodate standard genomic files. AeQTL consistently yielded similar or higher powers for identifying rare variant eQTLs than single-variant tests. Using AeQTL, we discovered that aggregated rare germline truncations in cis exomic regions are significantly associated with the expression of BRCA1 and SLC25A39 in breast tumors. In a somatic mutation pan-cancer analysis, aggregated mutations of those predicted to be missense versus truncations were differentially associated with gene expressions of cancer drivers, and somatic truncation eQTLs were further identified as a new multi-omic classifier of oncogenes versus tumor-suppressor genes. AeQTL is easy to use and customize, allowing a broad application for discovering rare variants, including coding and noncoding variants, associated with gene expression. AeQTL is implemented in Python and the source code is freely available at https://github.com/Huan-glab/AeQTL under the MIT license.


Assuntos
Polimorfismo de Nucleotídeo Único , Locos de Características Quantitativas , Biologia Computacional , Estudo de Associação Genômica Ampla , Genômica , Humanos , Oncogenes
18.
Cell ; 183(6): 1634-1649.e17, 2020 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-33259803

RESUMO

Microsatellite instability-high (MSI-H) tumors are characterized by high tumor mutation burden and responsiveness to checkpoint blockade. We identified tumor-specific frameshifts encoding multiple epitopes that originated from indel mutations shared among patients with MSI-H endometrial, colorectal, and stomach cancers. Epitopes derived from these shared frameshifts have high population occurrence rates, wide presence in many tumor subclones, and are predicted to bind to the most frequent MHC alleles in MSI-H patient cohorts. Neoantigens arising from these mutations are distinctly unlike self and viral antigens, signifying novel groups of potentially highly immunogenic tumor antigens. We further confirmed the immunogenicity of frameshift peptides in T cell stimulation experiments using blood mononuclear cells isolated from both healthy donors and MSI-H cancer patients. Our study uncovers the widespread occurrence and strong immunogenicity of tumor-specific antigens derived from shared frameshift mutations in MSI-H cancer and Lynch syndrome patients, suitable for the design of common "off-the-shelf" cancer vaccines.


Assuntos
Epitopos/genética , Epitopos/imunologia , Mutação da Fase de Leitura/genética , Instabilidade de Microssatélites , Neoplasias/genética , Neoplasias/imunologia , Sequência de Aminoácidos , Antígenos de Neoplasias/imunologia , Antígenos Virais/imunologia , Linhagem Celular Tumoral , Análise Mutacional de DNA , Regulação Neoplásica da Expressão Gênica , Genoma Humano , Humanos , Imunoterapia , Mutação de Sentido Incorreto/genética , Neoplasias/terapia , Peptídeos/química , Peptídeos/imunologia , Análise de Sobrevida , Linfócitos T/imunologia
19.
Gastroenterology ; 159(6): 2203-2220.e14, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32814112

RESUMO

BACKGROUND AND AIMS: The pattern of genetic alterations in cancer driver genes in patients with hepatocellular carcinoma (HCC) is highly diverse, which partially explains the low efficacy of available therapies. In spite of this, the existing mouse models only recapitulate a small portion of HCC inter-tumor heterogeneity, limiting the understanding of the disease and the nomination of personalized therapies. Here, we aimed at establishing a novel collection of HCC mouse models that captured human HCC diversity. METHODS: By performing hydrodynamic tail-vein injections, we tested the impact of altering a well-established HCC oncogene (either MYC or ß-catenin) in combination with an additional alteration in one of eleven other genes frequently mutated in HCC. Of the 23 unique pairs of genetic alterations that we interrogated, 9 were able to induce HCC. The established HCC mouse models were characterized at histopathological, immune, and transcriptomic level to identify the unique features of each model. Murine HCC cell lines were generated from each tumor model, characterized transcriptionally, and used to identify specific therapies that were validated in vivo. RESULTS: Cooperation between pairs of driver genes produced HCCs with diverse histopathology, immune microenvironments, transcriptomes, and drug responses. Interestingly, MYC expression levels strongly influenced ß-catenin activity, indicating that inter-tumor heterogeneity emerges not only from specific combinations of genetic alterations but also from the acquisition of expression-dependent phenotypes. CONCLUSIONS: This novel collection of murine HCC models and corresponding cell lines establishes the role of driver genes in diverse contexts and enables mechanistic and translational studies.


Assuntos
Carcinoma Hepatocelular/genética , Heterogeneidade Genética , Proto-Oncogenes/genética , Animais , Carcinoma Hepatocelular/imunologia , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Biologia Computacional , Modelos Animais de Doenças , Resistencia a Medicamentos Antineoplásicos/genética , Feminino , Regulação Neoplásica da Expressão Gênica/imunologia , Humanos , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/patologia , Masculino , Camundongos , Camundongos Transgênicos , Evasão Tumoral/genética , Microambiente Tumoral/genética , Microambiente Tumoral/imunologia
20.
Cancer Cell ; 37(5): 639-654.e6, 2020 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-32396860

RESUMO

We evaluated ancestry effects on mutation rates, DNA methylation, and mRNA and miRNA expression among 10,678 patients across 33 cancer types from The Cancer Genome Atlas. We demonstrated that cancer subtypes and ancestry-related technical artifacts are important confounders that have been insufficiently accounted for. Once accounted for, ancestry-associated differences spanned all molecular features and hundreds of genes. Biologically significant differences were usually tissue specific but not specific to cancer. However, admixture and pathway analyses suggested some of these differences are causally related to cancer. Specific findings included increased FBXW7 mutations in patients of African origin, decreased VHL and PBRM1 mutations in renal cancer patients of African origin, and decreased immune activity in bladder cancer patients of East Asian origin.


Assuntos
Metilação de DNA , Etnicidade/genética , Predisposição Genética para Doença , MicroRNAs/genética , Mutação , Proteínas de Neoplasias/genética , Neoplasias/genética , Proteínas de Ligação a DNA/genética , Proteína 7 com Repetições F-Box-WD/genética , Regulação Neoplásica da Expressão Gênica , Genética Populacional , Genoma Humano , Genômica , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Neoplasias/etnologia , Neoplasias/patologia , Fatores de Transcrição/genética , Proteína Supressora de Tumor Von Hippel-Lindau/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA