Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cancer Cell ; 41(9): 1586-1605.e15, 2023 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-37567170

RESUMEN

We characterized a prospective endometrial carcinoma (EC) cohort containing 138 tumors and 20 enriched normal tissues using 10 different omics platforms. Targeted quantitation of two peptides can predict antigen processing and presentation machinery activity, and may inform patient selection for immunotherapy. Association analysis between MYC activity and metformin treatment in both patients and cell lines suggests a potential role for metformin treatment in non-diabetic patients with elevated MYC activity. PIK3R1 in-frame indels are associated with elevated AKT phosphorylation and increased sensitivity to AKT inhibitors. CTNNB1 hotspot mutations are concentrated near phosphorylation sites mediating pS45-induced degradation of ß-catenin, which may render Wnt-FZD antagonists ineffective. Deep learning accurately predicts EC subtypes and mutations from histopathology images, which may be useful for rapid diagnosis. Overall, this study identified molecular and imaging markers that can be further investigated to guide patient stratification for more precise treatment of EC.


Asunto(s)
Neoplasias Endometriales , Metformina , Proteogenómica , Femenino , Humanos , Proteínas Proto-Oncogénicas c-akt/genética , Estudios Prospectivos , Neoplasias Endometriales/tratamiento farmacológico , Neoplasias Endometriales/genética , Neoplasias Endometriales/metabolismo , beta Catenina/genética , beta Catenina/metabolismo , Metformina/farmacología
2.
Cell ; 186(9): 1985-2001.e19, 2023 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-37075754

RESUMEN

Aneuploidy, the presence of chromosome gains or losses, is a hallmark of cancer. Here, we describe KaryoCreate (karyotype CRISPR-engineered aneuploidy technology), a system that enables the generation of chromosome-specific aneuploidies by co-expression of an sgRNA targeting chromosome-specific CENPA-binding ɑ-satellite repeats together with dCas9 fused to mutant KNL1. We design unique and highly specific sgRNAs for 19 of the 24 chromosomes. Expression of these constructs leads to missegregation and induction of gains or losses of the targeted chromosome in cellular progeny, with an average efficiency of 8% for gains and 12% for losses (up to 20%) validated across 10 chromosomes. Using KaryoCreate in colon epithelial cells, we show that chromosome 18q loss, frequent in gastrointestinal cancers, promotes resistance to TGF-ß, likely due to synergistic hemizygous deletion of multiple genes. Altogether, we describe an innovative technology to create and study chromosome missegregation and aneuploidy in the context of cancer and beyond.


Asunto(s)
Centrómero , Técnicas Genéticas , Humanos , Aneuploidia , Centrómero/genética , Deleción Cromosómica , Neoplasias/genética , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas
3.
Cancer ; 129(5): 714-727, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36597662

RESUMEN

BACKGROUND: Studies of the immune landscape led to breakthrough trials of programmed death-1 (PD-1) inhibitors for recurrent/metastatic head and neck squamous cell carcinoma therapy. This study investigated the timing, influence of somatic copy-number alterations (SCNAs), and clinical implications of PD-L1 and immune-cell patterns in oral precancer (OPC). METHODS: The authors evaluated spatial CD3, CD3/8, and CD68 density (cells/mm2 ) and PD-L1 (membranous expression in cytokeratin-positive intraepithelial neoplastic cells and CD68) patterns by multiplex immunofluorescence in a 188-patient prospective OPC cohort, characterized by clinical, histologic, and SCNA risk factors and protocol-specified primary end point of invasive cancer. The authors used Wilcoxon rank-sum and Fisher exact tests, linear mixed effect models, mediation, and Cox regression and recursive-partitioning analyses. RESULTS: Epithelial, but not CD68 immune-cell, PD-L1 expression was detected in 28% of OPCs, correlated with immune-cell infiltration, 9p21.3 loss of heterozygosity (LOH), and inferior oral cancer-free survival (OCFS), notably in OPCs with low CD3/8 cell density, dysplasia, and/or 9p21.3 LOH. High CD3/8 cell density in dysplastic lesions predicted better OCFS and eliminated the excess risk associated with prior oral cancer and dysplasia. PD-L1 and CD3/8 patterns revealed inferior OCFS in PD-L1 high intrinsic induction and dysplastic immune-cold subgroups. CONCLUSION: This report provides spatial insight into the immune landscape and drivers of OPCs, and a publicly available immunogenomic data set for future precancer interrogation. The data suggest that 9p21.3 LOH triggers an immune-hot inflammatory phenotype; whereas increased 9p deletion size encompassing CD274 at 9p24.1 may contribute to CD3/8 and PD-L1 depletion during invasive transition. The inferior OCFS in PD-L1-high, immune-cold OPCs support the development of T-cell recruitment strategies.


Asunto(s)
Neoplasias de Cabeza y Cuello , Neoplasias de la Boca , Humanos , Antígeno B7-H1 , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Genómica , Neoplasias de Cabeza y Cuello/metabolismo , Linfocitos Infiltrantes de Tumor , Neoplasias de la Boca/genética , Neoplasias de la Boca/metabolismo , Recurrencia Local de Neoplasia/metabolismo , Estudios Prospectivos , Carcinoma de Células Escamosas de Cabeza y Cuello/metabolismo , Microambiente Tumoral/genética
4.
Proc Natl Acad Sci U S A ; 119(47): e2213835119, 2022 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-36395141

RESUMEN

Somatic copy number alterations (SCNAs), generally (1) losses containing interferons and interferon-pathway genes, many on chromosome 9p, predict immune-cold, immune checkpoint therapy (ICT)-resistant tumors (2); however, genomic regions mediating these effects are unclear and probably tissue specific. Previously, 9p21.3 loss was found to be an early genetic driver of human papillomavirus-negative (HPV-) head and neck squamous cancer (HNSC), associated with an immune-cold tumor microenvironment (TME) signal, and recent evidence suggested that this TME-cold phenotype was greatly enhanced with 9p21 deletion size, notably encompassing band 9p24.1 (3). Here, we report multi-omic, -threshold and continuous-variable dissection of 9p21 and 9p24 loci (including depth and degree of somatic alteration of each band at each locus, and each gene at each band) and TME of four HPV- HNSC cohorts. Preferential 9p24 deletion, CD8 T-cell immune-cold associations were observed, driven by 9p24.1 loss, and in turn by an essential telomeric regulatory gene element, JAK2-CD274. Surprisingly, same genetic region gains were immune hot. Related 9p21-TME analyses were less evident. Inherent 9p-band-level influences on anti-PD1 ICT survival rates, coincident with TME patterns, were also observed. At a 9p24.1 whole-transcriptome expression threshold of 60th percentile, ICT survival rate exceeded that of lower expression percentiles and of chemotherapy; below this transcript threshold, ICT survival was inferior to chemotherapy, the latter unaffected by 9p24.1 expression level (P-values < 0.01, including in a PD-L1 immunohistochemistry-positive patient subgroup). Whole-exome analyses of 10 solid-tumor types suggest that these 9p-related ICT findings could be relevant to squamous cancers, in which 9p24.1 gain/immune-hot associations exist.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias de Cabeza y Cuello , Infecciones por Papillomavirus , Humanos , Microambiente Tumoral/genética , Inhibidores de Puntos de Control Inmunológico , Infecciones por Papillomavirus/genética , Carcinoma de Células Escamosas de Cabeza y Cuello/tratamiento farmacológico , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Neoplasias de Cabeza y Cuello/genética
5.
Elife ; 112022 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-36129397

RESUMEN

How cells control gene expression is a fundamental question. The relative contribution of protein-level and RNA-level regulation to this process remains unclear. Here, we perform a proteogenomic analysis of tumors and untransformed cells containing somatic copy number alterations (SCNAs). By revealing how cells regulate RNA and protein abundances of genes with SCNAs, we provide insights into the rules of gene regulation. Protein complex genes have a strong protein-level regulation while non-complex genes have a strong RNA-level regulation. Notable exceptions are plasma membrane protein complex genes, which show a weak protein-level regulation and a stronger RNA-level regulation. Strikingly, we find a strong negative association between the degree of RNA-level and protein-level regulation across genes and cellular pathways. Moreover, genes participating in the same pathway show a similar degree of RNA- and protein-level regulation. Pathways including translation, splicing, RNA processing, and mitochondrial function show a stronger protein-level regulation while cell adhesion and migration pathways show a stronger RNA-level regulation. These results suggest that the evolution of gene regulation is shaped by functional constraints and that many cellular pathways tend to evolve one predominant mechanism of gene regulation at the protein level or at the RNA level.


Asunto(s)
Neoplasias , Proteogenómica , Aneuploidia , Humanos , Proteínas de la Membrana , Neoplasias/genética , ARN
6.
Nature ; 603(7902): 728-735, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35296855

RESUMEN

The engineering of autologous patient T cells for adoptive cell therapies has revolutionized the treatment of several types of cancer1. However, further improvements are needed to increase response and cure rates. CRISPR-based loss-of-function screens have been limited to negative regulators of T cell functions2-4 and raise safety concerns owing to the permanent modification of the genome. Here we identify positive regulators of T cell functions through overexpression of around 12,000 barcoded human open reading frames (ORFs). The top-ranked genes increased the proliferation and activation of primary human CD4+ and CD8+ T cells and their secretion of key cytokines such as interleukin-2 and interferon-γ. In addition, we developed the single-cell genomics method OverCITE-seq for high-throughput quantification of the transcriptome and surface antigens in ORF-engineered T cells. The top-ranked ORF-lymphotoxin-ß receptor (LTBR)-is typically expressed in myeloid cells but absent in lymphocytes. When overexpressed in T cells, LTBR induced profound transcriptional and epigenomic remodelling, leading to increased T cell effector functions and resistance to exhaustion in chronic stimulation settings through constitutive activation of the canonical NF-κB pathway. LTBR and other highly ranked genes improved the antigen-specific responses of chimeric antigen receptor T cells and γδ T cells, highlighting their potential for future cancer-agnostic therapies5. Our results provide several strategies for improving next-generation T cell therapies by the induction of synthetic cell programmes.


Asunto(s)
Linfocitos T CD8-positivos , Neoplasias , Linfocitos T CD4-Positivos , Proliferación Celular , Humanos , Inmunoterapia Adoptiva , Activación de Linfocitos/genética
7.
Proc Natl Acad Sci U S A ; 118(19)2021 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-33952700

RESUMEN

An aneuploid-immune paradox encompasses somatic copy-number alterations (SCNAs), unleashing a cytotoxic response in experimental precancer systems, while conversely being associated with immune suppression and cytotoxic-cell depletion in human tumors, especially head and neck cancer (HNSC). We present evidence from patient samples and cell lines that alterations in chromosome dosage contribute to an immune hot-to-cold switch during human papillomavirus-negative (HPV-) head and neck tumorigenesis. Overall SCNA (aneuploidy) level was associated with increased CD3+ and CD8+ T cell microenvironments in precancer (mostly CD3+, linked to trisomy and aneuploidy), but with T cell-deficient tumors. Early lesions with 9p21.3 loss were associated with depletion of cytotoxic T cell infiltration in TP53 mutant tumors; and with aneuploidy were associated with increased NK-cell infiltration. The strongest driver of cytotoxic T cell and Immune Score depletion in oral cancer was 9p-arm level loss, promoting profound decreases of pivotal IFN-γ-related chemokines (e.g., CXCL9) and pathway genes. Chromosome 9p21.3 deletion contributed mainly to cell-intrinsic senescence suppression, but deletion of the entire arm was necessary to diminish levels of cytokine, JAK-STAT, and Hallmark NF-κB pathways. Finally, 9p arm-level loss and JAK2-PD-L1 codeletion (at 9p24) were predictive markers of poor survival in recurrent HPV- HNSC after anti-PD-1 therapy; likely amplified by independent aneuploidy-induced immune-cold microenvironments observed here. We hypothesize that 9p21.3 arm-loss expansion and epistatic interactions allow oral precancer cells to acquire properties to overcome a proimmunogenic aneuploid checkpoint, transform and invade. These findings enable distinct HNSC interception and precision-therapeutic approaches, concepts that may apply to other CN-driven neoplastic, immune or aneuploid diseases, and immunotherapies.


Asunto(s)
Aneuploidia , Deleción Cromosómica , Neoplasias de Cabeza y Cuello/genética , Evasión Inmune , Infecciones por Papillomavirus , Adulto , Anciano , Anciano de 80 o más Años , Antígeno B7-H1 , Complejo CD3 , Linfocitos T CD8-positivos , Línea Celular Tumoral , Cromosomas , Citocinas , Variaciones en el Número de Copia de ADN , Regulación Neoplásica de la Expresión Génica , Genes p53/genética , Humanos , Evasión Inmune/genética , Inmunoterapia , Janus Quinasa 2 , Persona de Mediana Edad , Infecciones por Papillomavirus/genética , Linfocitos T Citotóxicos , Microambiente Tumoral , Adulto Joven
8.
Genetics ; 218(1)2021 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-33742653

RESUMEN

Design and large-scale synthesis of DNA has been applied to the functional study of viral and microbial genomes. New and expanded technology development is required to unlock the transformative potential of such bottom-up approaches to the study of larger mammalian genomes. Two major challenges include assembling and delivering long DNA sequences. Here, we describe a workflow for de novo DNA assembly and delivery that enables functional evaluation of mammalian genes on the length scale of 100 kilobase pairs (kb). The DNA assembly step is supported by an integrated robotic workcell. We demonstrate assembly of the 101 kb human HPRT1 gene in yeast from 3 kb building blocks, precision delivery of the resulting construct to mouse embryonic stem cells, and subsequent expression of the human protein from its full-length human gene in mouse cells. This workflow provides a framework for mammalian genome writing. We envision utility in producing designer variants of human genes linked to disease and their delivery and functional analysis in cell culture or animal models.


Asunto(s)
Clonación Molecular/métodos , Ingeniería Genética/métodos , Animales , ADN/genética , Técnicas de Transferencia de Gen/veterinaria , Técnicas Genéticas/veterinaria , Genoma/genética , Genómica/métodos , Humanos , Hipoxantina Fosforribosiltransferasa/genética , Ratones , Análisis de Secuencia de ADN/métodos , Flujo de Trabajo
9.
Curr Opin Cell Biol ; 63: 135-143, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32092639

RESUMEN

Tumors arise through waves of genetic alterations and clonal expansion that allow tumor cells to acquire cancer hallmarks, such as genome instability and immune evasion. Recent genomic analyses showed that the vast majority of cancer driver genes are mutated in a tissue-dependent manner, that is, are altered in some cancers but not others. Often the tumor type also affects the likelihood of therapy response. What is the origin of tissue specificity in cancer? Recent studies suggest that both cell-intrinsic and cell-extrinsic factors play a role. On one hand, cell type-specific wiring of the cell signaling network determines the outcome of cancer driver gene mutations. On the other hand, the tumor cells' exposure to tissue-specific microenvironments (e.g. immune cells) also contributes to shape the tissue specificity of driver genes and of therapy response. In the future, a more complete understanding of tissue specificity in cancer may inform methods to better predict and improve therapeutic outcomes.


Asunto(s)
Neoplasias/clasificación , Neoplasias/genética , Neoplasias/patología , Animales , Regulación Neoplásica de la Expresión Génica , Inestabilidad Genómica/fisiología , Humanos , Mutación/fisiología , Neoplasias/metabolismo , Oncogenes/fisiología , Especificidad de Órganos/genética , Transducción de Señal/genética , Microambiente Tumoral/genética
10.
Cell ; 173(2): 499-514.e23, 2018 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-29576454

RESUMEN

Genomics has provided a detailed structural description of the cancer genome. Identifying oncogenic drivers that work primarily through dosage changes is a current challenge. Unrestrained proliferation is a critical hallmark of cancer. We constructed modular, barcoded libraries of human open reading frames (ORFs) and performed screens for proliferation regulators in multiple cell types. Approximately 10% of genes regulate proliferation, with most performing in an unexpectedly highly tissue-specific manner. Proliferation drivers in a given cell type showed specific enrichment in somatic copy number changes (SCNAs) from cognate tumors and helped predict aneuploidy patterns in those tumors, implying that tissue-type-specific genetic network architectures underlie SCNA and driver selection in different cancers. In vivo screening confirmed these results. We report a substantial contribution to the catalog of SCNA-associated cancer drivers, identifying 147 amplified and 107 deleted genes as potential drivers, and derive insights about the genetic network architecture of aneuploidy in tumors.


Asunto(s)
Aneuploidia , Neoplasias/patología , Animales , Línea Celular Tumoral , Proliferación Celular , Mapeo Cromosómico , Cromosomas/genética , Factor de Transcripción E2F1/antagonistas & inhibidores , Factor de Transcripción E2F1/genética , Factor de Transcripción E2F1/metabolismo , Femenino , Biblioteca de Genes , Genómica , Humanos , Queratinas/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Oncogenes , Sistemas de Lectura Abierta/genética , Interferencia de ARN , ARN Interferente Pequeño/metabolismo
11.
Genes Dev ; 31(2): 184-196, 2017 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-28167502

RESUMEN

A large number of cancer drivers have been identified through tumor sequencing efforts, but how they interact and the degree to which they can substitute for each other have not been systematically explored. To comprehensively investigate how cancer drivers genetically interact, we searched for modifiers of epidermal growth factor receptor (EGFR) dependency by performing CRISPR, shRNA, and expression screens in a non-small cell lung cancer (NSCLC) model. We elucidated a broad spectrum of tumor suppressor genes (TSGs) and oncogenes (OGs) that can genetically modify proliferation and survival of cancer cells when EGFR signaling is altered. These include genes already known to mediate EGFR inhibitor resistance as well as many TSGs not previously connected to EGFR and whose biological functions in tumorigenesis are not well understood. We show that mutation of PBRM1, a subunit of the SWI/SNF complex, attenuates the effects of EGFR inhibition in part by sustaining AKT signaling. We also show that mutation of Capicua (CIC), a transcriptional repressor, suppresses the effects of EGFR inhibition by partially restoring the EGFR-promoted gene expression program, including the sustained expression of Ets transcription factors such as ETV1 Together, our data provide strong support for the hypothesis that many cancer drivers can substitute for each other in certain contexts and broaden our understanding of EGFR regulation.


Asunto(s)
Adenocarcinoma/genética , Adenocarcinoma/fisiopatología , Receptores ErbB/genética , Receptores ErbB/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/fisiopatología , Adenocarcinoma del Pulmón , Antineoplásicos/farmacología , Línea Celular Tumoral , Proteínas de Unión al ADN , Resistencia a Antineoplásicos/genética , Activación Enzimática/efectos de los fármacos , Gefitinib , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Proteínas Nucleares/genética , Proteína Oncogénica v-akt/metabolismo , Quinazolinas/farmacología , Proteínas Represoras/genética , Eliminación de Secuencia , Transducción de Señal/genética , Factores de Transcripción/genética , Transcriptoma
12.
Science ; 355(6322)2017 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-28104840

RESUMEN

Immunotherapies based on immune checkpoint blockade are highly effective in a subset of patients. An ongoing challenge is the identification of biomarkers that predict which patients will benefit from these therapies. Aneuploidy, also known as somatic copy number alterations (SCNAs), is widespread in cancer and is posited to drive tumorigenesis. Analyzing 12 human cancer types, we find that, for most, highly aneuploid tumors show reduced expression of markers of cytotoxic infiltrating immune cells, especially CD8+ T cells, and increased expression of cell proliferation markers. Different types of SCNAs predict the proliferation and immune signatures, implying distinct underlying mechanisms. Using published data from two clinical trials of immune checkpoint blockade therapy for metastatic melanoma, we found that tumor aneuploidy inversely correlates with patient survival. Together with other tumor characteristics such as tumor mutational load, aneuploidy may thus help identify patients most likely to respond to immunotherapy.


Asunto(s)
Variaciones en el Número de Copia de ADN , Linfocitos Infiltrantes de Tumor/inmunología , Neoplasias/genética , Neoplasias/terapia , Linfocitos T Citotóxicos/inmunología , Escape del Tumor/genética , Biomarcadores de Tumor/inmunología , Antígeno CTLA-4/antagonistas & inhibidores , Ciclo Celular/inmunología , Proliferación Celular , Citotoxicidad Inmunológica , Humanos , Inmunoterapia , Modelos Biológicos , Neoplasias/inmunología , Neoplasias/mortalidad , Mutación Puntual , Pronóstico , Transcriptoma
13.
G3 (Bethesda) ; 6(9): 2781-90, 2016 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-27402361

RESUMEN

Genetic screens are invaluable tools for dissection of biological phenomena. Optimization of such screens to enhance discovery of candidate genes and minimize false positives is thus a critical aim. Here, we report several sources of error common to pooled genetic screening techniques used in mammalian cell culture systems, and demonstrate methods to eliminate these errors. We find that reverse transcriptase-mediated recombination during retroviral replication can lead to uncoupling of molecular tags, such as DNA barcodes (BCs), from their associated library elements, leading to chimeric proviral genomes in which BCs are paired to incorrect ORFs, shRNAs, etc This effect depends on the length of homologous sequence between unique elements, and can be minimized with careful vector design. Furthermore, we report that residual plasmid DNA from viral packaging procedures can contaminate transduced cells. These plasmids serve as additional copies of the PCR template during library amplification, resulting in substantial inaccuracies in measurement of initial reference populations for screen normalization. The overabundance of template in some samples causes an imbalance between PCR cycles of contaminated and uncontaminated samples, which results in a systematic artifactual depletion of GC-rich library elements. Elimination of contaminating plasmid DNA using the bacterial endonuclease Benzonase can restore faithful measurements of template abundance and minimize GC bias.


Asunto(s)
Código de Barras del ADN Taxonómico/normas , Pruebas Genéticas/normas , Mamíferos/genética , Animales , Técnicas de Cultivo de Célula/normas , Vectores Genéticos , Genoma , Plásmidos/genética , Reacción en Cadena de la Polimerasa/normas , ARN Interferente Pequeño/genética
14.
Genes Dev ; 30(24): 2684-2695, 2016 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-28087713

RESUMEN

Activating mutations in the phosphoinositide 3-kinase (PI3K) signaling pathway are frequently identified in cancer. To identify pathways that support PI3K oncogenesis, we performed a genome-wide RNAi screen in isogenic cell lines harboring wild-type or mutant PIK3CA to search for PI3K synthetic-lethal (SL) genes. A combined analysis of these results with a meta-analysis of two other large-scale RNAi screening data sets in PI3K mutant cancer cell lines converged on ribosomal protein translation and proteasomal protein degradation as critical nononcogene dependencies for PI3K-driven tumors. Genetic or pharmacologic inhibition of either pathway alone, but not together, selectively killed PI3K mutant tumor cells in an mTOR-dependent manner. The expression of ribosomal and proteasomal components was significantly up-regulated in primary human colorectal tumors harboring PI3K pathway activation. Importantly, a PI3K SL gene signature containing the top hits of the SL genes identified in our meta-analysis robustly predicted overall patient survival in colorectal cancer, especially among patients with tumors with an activated PI3K pathway. These results suggest that disruption of protein turnover homeostasis via ribosome or proteasome inhibition may be a novel treatment strategy for PI3K mutant human tumors.


Asunto(s)
Neoplasias Colorrectales/enzimología , Neoplasias Colorrectales/genética , Regulación Neoplásica de la Expresión Génica , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal/genética , Animales , Neoplasias Colorrectales/diagnóstico , Neoplasias Colorrectales/fisiopatología , Genómica , Células HCT116 , Células HEK293 , Humanos , Ratones , Mutación , Complejo de la Endopetidasa Proteasomal/genética , Ribosomas/genética
15.
J Cutan Pathol ; 42(2): 108-17, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25407517

RESUMEN

BACKGROUND: Alterations in pathways including BRAF, CDKN2A, and TERT contribute to the development of melanoma, but the sequence in which the genetic alterations occur and their prognostic significance remains unclear. To clarify the role of these pathways, we analyzed a primary melanoma and its metastasis. METHODS: Immunohistochemistry for BRAF-V600E, Sanger sequencing of BRAF and the TERT promoter, fluorescence in-situ hybridization, and telomere analyses were performed on a primary melanoma and its asynchronous cerebellar metastasis. Using the log-rank test and Cox-proportional model, the cancer genome atlas (TCGA) cohort of melanomas was analyzed for the effect of BRAF mutation and CDKN2A loss on survival. RESULTS: The primary melanoma expressed mutant BRAF-V600E and possessed a homozygous deletion of CDKN2A. In addition to these early defects, the metastatic lesion also possessed evidence of aneuploidy and an activating mutation of the TERT promoter. In the TCGA melanoma cohort, there was a non-significant trend toward poor prognosis in early stage cutaneous melanoma patients with concomitant BRAF mutation and CDKN2A loss. CONCLUSION: BRAF mutation and CDKN2A loss occurred early and TERT promoter mutation later in a case of lethal metastatic melanoma. The effects of these pathways on survival warrant further investigation in early stage cutaneous melanoma patients.


Asunto(s)
Genes p16 , Melanoma/genética , Neoplasias Meníngeas/genética , Neoplasias Meníngeas/secundario , Proteínas Proto-Oncogénicas B-raf/genética , Telomerasa/genética , Adulto , Secuencia de Bases , Femenino , Estudios de Seguimiento , Humanos , Melanoma/patología , Mutación , Neoplasias Cutáneas , Adulto Joven , Melanoma Cutáneo Maligno
16.
Cell Rep ; 9(2): 752-66, 2014 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-25373910

RESUMEN

RNAi screens have implicated hundreds of host proteins as HIV-1 dependency factors (HDFs). While informative, these early studies overlap poorly due to false positives and false negatives. To ameliorate these issues, we combined information from the existing HDF screens together with new screens performed with multiple orthologous RNAi reagents (MORR). In addition to being traditionally validated, the MORR screens and the historical HDF screens were quantitatively integrated by the adaptation of an established analysis program, RIGER, for the collective interpretation of each gene's phenotypic significance. False positives were addressed by the removal of poorly expressed candidates through gene expression filtering, as well as with GESS, which identifies off-target effects. This workflow produced a quantitatively integrated network of genes that modulate HIV-1 replication. We further investigated the roles of GOLGI49, SEC13, and COG in HIV-1 replication. Collectively, the MORR-RIGER method minimized the caveats of RNAi screening and improved our understanding of HIV-1-host cell interactions.


Asunto(s)
VIH-1/fisiología , Ensayos Analíticos de Alto Rendimiento/métodos , Interacciones Huésped-Patógeno , Interferencia de ARN , Replicación Viral , Proteínas Adaptadoras del Transporte Vesicular/genética , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Algoritmos , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Unión al ADN , Células HEK293 , Células HeLa , Humanos , Células Jurkat , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas de Unión al ARN
17.
Cell ; 155(4): 948-62, 2013 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-24183448

RESUMEN

Aneuploidy has been recognized as a hallmark of cancer for more than 100 years, yet no general theory to explain the recurring patterns of aneuploidy in cancer has emerged. Here, we develop Tumor Suppressor and Oncogene (TUSON) Explorer, a computational method that analyzes the patterns of mutational signatures in tumors and predicts the likelihood that any individual gene functions as a tumor suppressor (TSG) or oncogene (OG). By analyzing >8,200 tumor-normal pairs, we provide statistical evidence suggesting that many more genes possess cancer driver properties than anticipated, forming a continuum of oncogenic potential. Integrating our driver predictions with information on somatic copy number alterations, we find that the distribution and potency of TSGs (STOP genes), OGs, and essential genes (GO genes) on chromosomes can predict the complex patterns of aneuploidy and copy number variation characteristic of cancer genomes. We propose that the cancer genome is shaped through a process of cumulative haploinsufficiency and triplosensitivity.


Asunto(s)
Algoritmos , Aneuploidia , Genes Supresores de Tumor , Neoplasias/genética , Oncogenes , Dosificación de Gen , Humanos
18.
Proc Natl Acad Sci U S A ; 110(5): E407-14, 2013 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-23319639

RESUMEN

Cancer develops through genetic and epigenetic alterations that allow unrestrained proliferation and increased survival. Using a genetic RNAi screen, we previously identified hundreds of suppressors of tumorigenesis and/or proliferation (STOP) genes that restrain normal cell proliferation. Our STOP gene set was significantly enriched for known and putative tumor suppressor genes. Here, we report a tumor-suppressive role for one STOP gene, phosphatase and actin regulator 4 (PHACTR4). Phactr4 is one of four members of the largely uncharacterized Phactr family of protein phosphatase 1 (PP1)-and actin-binding proteins. Our work suggests that Phactr4 restrains normal cell proliferation and transformation. Depletion of Phactr4 with multiple shRNAs leads to increased proliferation and soft agar colony formation. Phactr4 acts, in part, through an Rb-dependent pathway, because Rb phosphorylation is maintained upon growth factor withdrawal in Phactr4-depleted cells. Examination of tumor copy number analysis and sequencing revealed that PHACTR4 is significantly deleted and mutant in many tumor subtypes. Furthermore,cancer cell lines with reduced Phactr4 expression exhibit tumor suppressor hypersensitivity upon Phactr4 complementation,leading to reduced proliferation, transformation, and tumor formation. Thus, Phactr4 acts as a tumor suppressor that is deleted and mutant in several cancers.


Asunto(s)
Neoplasias de la Mama/genética , Proliferación Celular , Mutación , Proteínas Supresoras de Tumor/genética , Animales , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Transformada , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Células Cultivadas , Doxiciclina/farmacología , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Immunoblotting , Células MCF-7 , Glándulas Mamarias Humanas/citología , Glándulas Mamarias Humanas/metabolismo , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Interferencia de ARN , Transfección , Trasplante Heterólogo , Proteínas Supresoras de Tumor/metabolismo
19.
Cancer Cell ; 21(6): 765-76, 2012 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-22698402

RESUMEN

Human cancers with a subtetraploid karyotype are thought to originate from tetraploid precursors, but the cause of tetraploidization is unknown. We previously documented endoreduplication in mouse cells with persistent telomere dysfunction or genome-wide DNA damage. We now report that endoreduplication and mitotic failure occur during telomere crisis in human fibroblasts and mammary epithelial cells and document the role of p53 and Rb in repressing tetraploidization. Using an inducible system to generate transient telomere damage, we show that telomere-driven tetraploidization enhances the tumorigenic transformation of mouse cells. Similar to human solid cancers, the resulting tumors evolved subtetraploid karyotypes. These data establish that telomere-driven tetraploidization is induced by critically short telomeres and has the potential to promote tumorigenesis in early cancerous lesions.


Asunto(s)
Transformación Celular Neoplásica/genética , Daño del ADN , Telómero/genética , Tetraploidía , Animales , Puntos de Control del Ciclo Celular/genética , Línea Celular , Células Cultivadas , Células Epiteliales/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Citometría de Flujo , Humanos , Immunoblotting , Hibridación Fluorescente in Situ , Cariotipo , Glándulas Mamarias Humanas/citología , Ratones , Mitosis/genética , Interferencia de ARN , Proteína de Retinoblastoma/genética , Proteína de Retinoblastoma/metabolismo , Factores de Tiempo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
20.
Annu Rev Cell Dev Biol ; 27: 585-610, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21801013

RESUMEN

Although nearly all mammalian species are diploid, whole-genome duplications occur in select mammalian tissues as part of normal development. Such programmed polyploidization involves changes in the regulatory pathways that normally maintain the diploid state of the mammalian genome. Unscheduled whole-genome duplications, which lead primarily to tetraploid cells, also take place in a substantial fraction of human tumors and have been proposed to constitute an important step in the development of cancer aneuploidy. The origins of these polyploidization events and their consequences for tumor progression are explored in this review.


Asunto(s)
Morfogénesis/genética , Neoplasias/genética , Poliploidía , Envejecimiento/genética , Aneuploidia , Animales , Ciclo Celular/fisiología , Transformación Celular Neoplásica/genética , Daño del ADN , Diploidia , Genoma , Humanos , Cariotipificación , Estrés Fisiológico/genética , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...