Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 148
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 24(3): 487-500, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36759711

RESUMEN

The T cell repertoire of healthy mice and humans harbors self-reactive CD4+ conventional T (Tconv) cells capable of inducing autoimmunity. Using T cell receptor profiling paired with in vivo clonal analysis of T cell differentiation, we identified Tconv cell clones that are recurrently enriched in non-lymphoid organs following ablation of Foxp3+ regulatory T (Treg) cells. A subset of these clones was highly proliferative in the lymphoid organs at steady state and exhibited overt reactivity to self-ligands displayed by dendritic cells, yet were not purged by clonal deletion. These clones spontaneously adopted numerous hallmarks of follicular helper T (TFH) cells, including expression of Bcl6 and PD-1, exhibited an elevated propensity to localize within B cell follicles at steady state, and produced interferon-γ in non-lymphoid organs following sustained Treg cell depletion. Our work identifies a naturally occurring population of self-reactive TFH-like cells and delineates a previously unappreciated fate for self-specific Tconv cells.


Asunto(s)
Linfocitos T CD4-Positivos , Células T Auxiliares Foliculares , Linfocitos T Reguladores , Animales , Humanos , Ratones , Autoinmunidad , Diferenciación Celular , Células Clonales , Fenotipo , Linfocitos T Colaboradores-Inductores , Linfocitos T CD4-Positivos/inmunología
2.
Nat Immunol ; 21(5): 567-577, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32284593

RESUMEN

Unprimed mice harbor a substantial population of 'memory-phenotype' CD8+ T cells (CD8-MP cells) that exhibit hallmarks of activation and innate-like functional properties. Due to the lack of faithful markers to distinguish CD8-MP cells from bona fide CD8+ memory T cells, the developmental origins and antigen specificities of CD8-MP cells remain incompletely defined. Using deep T cell antigen receptor (TCR) sequencing, we found that the TCRs expressed by CD8-MP cells are highly recurrent and distinct from the TCRs expressed by naive-phenotype CD8+ T cells. CD8-MP clones exhibited reactivity to widely expressed self-ligands. T cell precursors expressing CD8-MP TCRs showed upregulation of the transcription factor Eomes during maturation in the thymus, prior to induction of the full memory phenotype, which is suggestive of a unique program triggered by recognition of self-ligands. Moreover, CD8-MP cells infiltrate oncogene-driven prostate tumors and express high densities of PD-1, which suggests potential roles in antitumor immunity and the response to immunotherapy.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Neoplasias de la Próstata/inmunología , Receptores de Antígenos de Linfocitos T/genética , Proteínas de Dominio T Box/metabolismo , Timo/fisiología , Animales , Autoantígenos/inmunología , Diferenciación Celular , Selección Clonal Mediada por Antígenos , Células Clonales , Memoria Inmunológica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Receptor de Muerte Celular Programada 1/metabolismo , Proteínas de Dominio T Box/genética , Regulación hacia Arriba
3.
Cell ; 167(3): 695-708.e16, 2016 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-27745971

RESUMEN

Heritability and genome stability are shaped by meiotic recombination, which is initiated via hundreds of DNA double-strand breaks (DSBs). The distribution of DSBs throughout the genome is not random, but mechanisms molding this landscape remain poorly understood. Here, we exploit genome-wide maps of mouse DSBs at unprecedented nucleotide resolution to uncover previously invisible spatial features of recombination. At fine scale, we reveal a stereotyped hotspot structure-DSBs occur within narrow zones between methylated nucleosomes-and identify relationships between SPO11, chromatin, and the histone methyltransferase PRDM9. At large scale, DSB formation is suppressed on non-homologous portions of the sex chromosomes via the DSB-responsive kinase ATM, which also shapes the autosomal DSB landscape at multiple size scales. We also provide a genome-wide analysis of exonucleolytic DSB resection lengths and elucidate spatial relationships between DSBs and recombination products. Our results paint a comprehensive picture of features governing successive steps in mammalian meiotic recombination.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN , Inestabilidad Genómica/genética , Recombinación Homóloga , Meiosis/genética , Animales , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Cromatina/genética , Cromatina/metabolismo , Metilación de ADN , Endodesoxirribonucleasas/genética , Endodesoxirribonucleasas/metabolismo , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Ratones , Ratones Endogámicos C57BL , Nucleosomas/enzimología , Nucleosomas/genética , Cromosoma X/genética , Cromosoma Y/genética
4.
Nature ; 629(8013): 927-936, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38588697

RESUMEN

Broad-spectrum RAS inhibition has the potential to benefit roughly a quarter of human patients with cancer whose tumours are driven by RAS mutations1,2. RMC-7977 is a highly selective inhibitor of the active GTP-bound forms of KRAS, HRAS and NRAS, with affinity for both mutant and wild-type variants3. More than 90% of cases of human pancreatic ductal adenocarcinoma (PDAC) are driven by activating mutations in KRAS4. Here we assessed the therapeutic potential of RMC-7977 in a comprehensive range of PDAC models. We observed broad and pronounced anti-tumour activity across models following direct RAS inhibition at exposures that were well-tolerated in vivo. Pharmacological analyses revealed divergent responses to RMC-7977 in tumour versus normal tissues. Treated tumours exhibited waves of apoptosis along with sustained proliferative arrest, whereas normal tissues underwent only transient decreases in proliferation, with no evidence of apoptosis. In the autochthonous KPC mouse model, RMC-7977 treatment resulted in a profound extension of survival followed by on-treatment relapse. Analysis of relapsed tumours identified Myc copy number gain as a prevalent candidate resistance mechanism, which could be overcome by combinatorial TEAD inhibition in vitro. Together, these data establish a strong preclinical rationale for the use of broad-spectrum RAS-GTP inhibition in the setting of PDAC and identify a promising candidate combination therapeutic regimen to overcome monotherapy resistance.


Asunto(s)
Antineoplásicos , Carcinoma Ductal Pancreático , Guanosina Trifosfato , Neoplasias Pancreáticas , Proteínas Proto-Oncogénicas p21(ras) , Animales , Femenino , Humanos , Ratones , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/patología , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Variaciones en el Número de Copia de ADN , Resistencia a Antineoplásicos/efectos de los fármacos , Genes myc , Guanosina Trifosfato/metabolismo , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Recurrencia Local de Neoplasia/tratamiento farmacológico , Recurrencia Local de Neoplasia/genética , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/antagonistas & inhibidores , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto , Mutación
5.
Nature ; 602(7895): 156-161, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34847567

RESUMEN

CD8 T cell-mediated autoimmune diseases result from the breakdown of self-tolerance mechanisms in autoreactive CD8 T cells1. How autoimmune T cell populations arise and are sustained, and the molecular programmes defining the autoimmune T cell state, are unknown. In type 1 diabetes, ß-cell-specific CD8 T cells destroy insulin-producing ß-cells. Here we followed the fate of ß-cell-specific CD8 T cells in non-obese diabetic mice throughout the course of type 1 diabetes. We identified a stem-like autoimmune progenitor population in the pancreatic draining lymph node (pLN), which self-renews and gives rise to pLN autoimmune mediators. pLN autoimmune mediators migrate to the pancreas, where they differentiate further and destroy ß-cells. Whereas transplantation of as few as 20 autoimmune progenitors induced type 1 diabetes, as many as 100,000 pancreatic autoimmune mediators did not. Pancreatic autoimmune mediators are short-lived, and stem-like autoimmune progenitors must continuously seed the pancreas to sustain ß-cell destruction. Single-cell RNA sequencing and clonal analysis revealed that autoimmune CD8 T cells represent unique T cell differentiation states and identified features driving the transition from autoimmune progenitor to autoimmune mediator. Strategies aimed at targeting the stem-like autoimmune progenitor pool could emerge as novel and powerful immunotherapeutic interventions for type 1 diabetes.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/patología , Células Secretoras de Insulina/inmunología , Células Madre/patología , Animales , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/trasplante , Autorrenovación de las Células , Células Clonales/inmunología , Células Clonales/metabolismo , Células Clonales/patología , Modelos Animales de Enfermedad , Femenino , Glucosa-6-Fosfatasa/inmunología , Factor Nuclear 1-alfa del Hepatocito/metabolismo , Células Secretoras de Insulina/patología , Ganglios Linfáticos/inmunología , Masculino , Ratones , Receptores de Antígenos de Linfocitos T/metabolismo , Análisis de la Célula Individual , Trasplante de Células Madre , Células Madre/inmunología , Células Madre/metabolismo , Transcriptoma
6.
Nature ; 608(7924): 795-802, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35978189

RESUMEN

Although p53 inactivation promotes genomic instability1 and presents a route to malignancy for more than half of all human cancers2,3, the patterns through which heterogenous TP53 (encoding human p53) mutant genomes emerge and influence tumorigenesis remain poorly understood. Here, in a mouse model of pancreatic ductal adenocarcinoma that reports sporadic p53 loss of heterozygosity before cancer onset, we find that malignant properties enabled by p53 inactivation are acquired through a predictable pattern of genome evolution. Single-cell sequencing and in situ genotyping of cells from the point of p53 inactivation through progression to frank cancer reveal that this deterministic behaviour involves four sequential phases-Trp53 (encoding mouse p53) loss of heterozygosity, accumulation of deletions, genome doubling, and the emergence of gains and amplifications-each associated with specific histological stages across the premalignant and malignant spectrum. Despite rampant heterogeneity, the deletion events that follow p53 inactivation target functionally relevant pathways that can shape genomic evolution and remain fixed as homogenous events in diverse malignant populations. Thus, loss of p53-the 'guardian of the genome'-is not merely a gateway to genetic chaos but, rather, can enable deterministic patterns of genome evolution that may point to new strategies for the treatment of TP53-mutant tumours.


Asunto(s)
Carcinogénesis , Progresión de la Enfermedad , Genes p53 , Genoma , Pérdida de Heterocigocidad , Neoplasias Pancreáticas , Proteína p53 Supresora de Tumor , Adenocarcinoma/genética , Adenocarcinoma/patología , Animales , Carcinogénesis/genética , Carcinogénesis/patología , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Evolución Molecular , Eliminación de Gen , Genes p53/genética , Genoma/genética , Ratones , Modelos Genéticos , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Proteína p53 Supresora de Tumor/genética
7.
Blood ; 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38728378

RESUMEN

B-cell maturation antigen (BCMA)-targeting therapeutics have dramatically improved outcomes in relapsed/refractory multiple myeloma (RRMM). However, whether the mechanisms of resistance between these therapies are shared and how the identification of such mechanisms before therapy initiation could refine clinical decision-making remains undefined. We analyzed outcomes for 72 RRMM patients treated with teclistamab, a CD3 x BCMA bispecific antibody (BsAb), 42% (30/72) of whom had prior BCMA-directed therapy exposure. Malignant plasma cell BCMA expression was present in all BCMA therapy-naïve patients. Prior therapy-mediated loss of plasma cell BCMA expression before teclistamab treatment, measured by immunohistochemistry, was observed in 3 patients, none of whom responded to teclistamab, and one of whom also did not respond to ciltacabtagene autoleucel. Whole exome sequencing of tumor DNA from one patient revealed biallelic loss of TNFRSF17 following treatment with belantamab mafodotin. Low-to-undetectable peripheral blood soluble BCMA levels correlated with the absence of BCMA expression by bone marrow plasma cells. Thus, although rare, loss of BCMA expression following TNFRSF17 gene deletions can occur following any BCMA-directed therapy and prevents response to subsequent anti-BCMA-directed treatments, underscoring the importance of verifying the presence of a target antigen.

8.
Cell ; 144(5): 719-31, 2011 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-21376234

RESUMEN

The nonrandom distribution of meiotic recombination influences patterns of inheritance and genome evolution, but chromosomal features governing this distribution are poorly understood. Formation of the DNA double-strand breaks (DSBs) that initiate recombination results in the accumulation of Spo11 protein covalently bound to small DNA fragments. By sequencing these fragments, we uncover a genome-wide DSB map of unprecedented resolution and sensitivity. We use this map to explore how DSB distribution is influenced by large-scale chromosome structures, chromatin, transcription factors, and local sequence composition. Our analysis offers mechanistic insight into DSB formation and early processing steps, supporting the view that the recombination terrain is molded by combinatorial and hierarchical interaction of factors that work on widely different size scales. This map illuminates the occurrence of DSBs in repetitive DNA elements, repair of which can lead to chromosomal rearrangements. We also discuss implications for evolutionary dynamics of recombination hot spots.


Asunto(s)
Genoma Fúngico , Saccharomyces cerevisiae/genética , Roturas del ADN de Doble Cadena , Endodesoxirribonucleasas/metabolismo , Estudio de Asociación del Genoma Completo , Recombinación Genética , Proteínas de Saccharomyces cerevisiae/metabolismo
9.
Cell ; 147(3): 554-64, 2011 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-22036564

RESUMEN

Insights into cancer genetics can lead to therapeutic opportunities. By cross-referencing chromosomal changes with an unbiased genetic screen we identify the ephrin receptor A7 (EPHA7) as a tumor suppressor in follicular lymphoma (FL). EPHA7 is a target of 6q deletions and inactivated in 72% of FLs. Knockdown of EPHA7 drives lymphoma development in a murine FL model. In analogy to its physiological function in brain development, a soluble splice variant of EPHA7 (EPHA7(TR)) interferes with another Eph-receptor and blocks oncogenic signals in lymphoma cells. Consistent with this drug-like activity, administration of the purified EPHA7(TR) protein produces antitumor effects against xenografted human lymphomas. Further, by fusing EPHA7(TR) to the anti-CD20 antibody (rituximab) we can directly target this tumor suppressor to lymphomas in vivo. Our study attests to the power of combining descriptive tumor genomics with functional screens and reveals EPHA7(TR) as tumor suppressor with immediate therapeutic potential.


Asunto(s)
Genes Supresores de Tumor , Linfoma Folicular/metabolismo , Receptor EphA7/metabolismo , Animales , Anticuerpos Monoclonales de Origen Murino/uso terapéutico , Línea Celular Tumoral , Cromosomas Humanos Par 6 , Genómica , Humanos , Linfoma Folicular/tratamiento farmacológico , Linfoma Folicular/genética , Masculino , Ratones , Trasplante de Neoplasias , Interferencia de ARN , Rituximab , Trasplante Heterólogo
11.
Immunity ; 44(5): 1102-13, 2016 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-27130899

RESUMEN

The promiscuous expression of tissue-restricted antigens in the thymus, driven in part by autoimmune regulator (Aire), is critical for the protection of peripheral tissues from autoimmune attack. Aire-dependent processes are thought to promote both clonal deletion and the development of Foxp3(+) regulatory T (Treg) cells, suggesting that autoimmunity associated with Aire deficiency results from two failed tolerance mechanisms. Here, examination of autoimmune lesions in Aire(-/-) mice revealed an unexpected third possibility. We found that the predominant conventional T cell clonotypes infiltrating target lesions express antigen receptors that were preferentially expressed by Foxp3(+) Treg cells in Aire(+/+) mice. Thus, Aire enforces immune tolerance by ensuring that distinct autoreactive T cell specificities differentiate into the Treg cell lineage; dysregulation of this process results in the diversion of Treg cell-biased clonotypes into pathogenic conventional T cells.


Asunto(s)
Autoinmunidad , Tolerancia Inmunológica , Próstata/inmunología , Subgrupos de Linfocitos T/fisiología , Linfocitos T Reguladores/fisiología , Timo/inmunología , Factores de Transcripción/metabolismo , Animales , Autoantígenos/inmunología , Autoinmunidad/genética , Diferenciación Celular , Linaje de la Célula , Supresión Clonal , Selección Clonal Mediada por Antígenos , Células Clonales , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Tolerancia Inmunológica/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Especificidad del Receptor de Antígeno de Linfocitos T , Factores de Transcripción/genética , Proteína AIRE
12.
Immunity ; 44(4): 847-59, 2016 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-27037189

RESUMEN

Although antigen recognition mediated by the T cell receptor (TCR) influences many facets of Foxp3(+) regulatory T (Treg) cell biology, including development and function, the cell types that present antigen to Treg cells in vivo remain largely undefined. By tracking a clonal population of Aire-dependent, prostate-specific Treg cells in mice, we demonstrated an essential role for dendritic cells (DCs) in regulating organ-specific Treg cell biology. We have shown that the thymic development of prostate-specific Treg cells required antigen presentation by DCs. Moreover, Batf3-dependent CD8α(+) DCs were dispensable for the development of this clonotype and had negligible impact on the polyclonal Treg cell repertoire. In the periphery, CCR7-dependent migratory DCs coordinated the activation of organ-specific Treg cells in the prostate-draining lymph nodes. Our results demonstrate that the development and peripheral regulation of organ-specific Treg cells are dependent on antigen presentation by DCs, implicating DCs as key mediators of organ-specific immune tolerance.


Asunto(s)
Presentación de Antígeno/inmunología , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Células Dendríticas/inmunología , Próstata/inmunología , Proteínas Represoras/genética , Autotolerancia/inmunología , Linfocitos T Reguladores/inmunología , Animales , Antígeno B7-1/biosíntesis , Antígeno B7-1/genética , Antígeno B7-2/biosíntesis , Antígeno B7-2/genética , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/inmunología , Antígenos CD8/metabolismo , Diferenciación Celular/inmunología , Movimiento Celular/inmunología , Activación de Linfocitos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Próstata/citología , Receptores de Antígenos de Linfocitos T/inmunología , Receptores CCR7/metabolismo , Proteínas Represoras/inmunología , Linfocitos T Reguladores/citología , Factores de Transcripción/metabolismo , Proteína AIRE
13.
Nature ; 571(7766): 576-579, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31292550

RESUMEN

Mutations in BRCA1 and BRCA2 predispose individuals to certain cancers1-3, and disease-specific screening and preventative strategies have reduced cancer mortality in affected patients4,5. These classical tumour-suppressor genes have tumorigenic effects associated with somatic biallelic inactivation, although haploinsufficiency may also promote the formation and progression of tumours6,7. Moreover, BRCA1/2-mutant tumours are often deficient in the repair of double-stranded DNA breaks by homologous recombination8-13, and consequently exhibit increased therapeutic sensitivity to platinum-containing therapy and inhibitors of poly-(ADP-ribose)-polymerase (PARP)14,15. However, the phenotypic and therapeutic relevance of mutations in BRCA1 or BRCA2 remains poorly defined in most cancer types. Here we show that in the 2.7% and 1.8% of patients with advanced-stage cancer and germline pathogenic or somatic loss-of-function alterations in BRCA1/2, respectively, selective pressure for biallelic inactivation, zygosity-dependent phenotype penetrance, and sensitivity to PARP inhibition were observed only in tumour types associated with increased heritable cancer risk in BRCA1/2 carriers (BRCA-associated cancer types). Conversely, among patients with non-BRCA-associated cancer types, most carriers of these BRCA1/2 mutation types had evidence for tumour pathogenesis that was independent of mutant BRCA1/2. Overall, mutant BRCA is an indispensable founding event for some tumours, but in a considerable proportion of other cancers, it appears to be biologically neutral-a difference predominantly conditioned by tumour lineage-with implications for disease pathogenesis, screening, design of clinical trials and therapeutic decision-making.


Asunto(s)
Linaje de la Célula , Genes BRCA1 , Genes BRCA2 , Mutación , Neoplasias/genética , Neoplasias/patología , Fenotipo , Alelos , Estudios de Cohortes , Heterocigoto , Humanos , Neoplasias/tratamiento farmacológico , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Cigoto
14.
Cancer Immunol Immunother ; 70(5): 1189-1202, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33123756

RESUMEN

Identification of neoepitopes as tumor-specific targets remains challenging, especially for cancers with low mutational burden, such as ovarian cancer. To identify mutated human leukocyte antigen (HLA) ligands as potential targets for immunotherapy in ovarian cancer, we combined mass spectrometry analysis of the major histocompatibility complex (MHC) class I peptidomes of ovarian cancer cells with parallel sequencing of whole exome and RNA in a patient with high-grade serous ovarian cancer. Four of six predicted mutated epitopes capable of binding to HLA-A*02:01 induced peptide-specific T cell responses in blood from healthy donors. In contrast, all six peptides failed to induce autologous peptide-specific response by T cells in peripheral blood or tumor-infiltrating lymphocytes from ascites of the patient. Surprisingly, T cell responses against a low-affinity p53-mutant Y220C epitope were consistently detected in the patient with either unprimed or in vitro peptide-stimulated T cells even though the patient's primary tumor did not bear this mutation. Our results demonstrated that tumor heterogeneity and distinct immune microenvironments within a patient should be taken into consideration for identification of immunogenic neoantigens. T cell responses to a driver gene-derived p53 Y220C mutation in ovarian cancer warrant further study.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Epítopos de Linfocito T/metabolismo , Antígeno HLA-A2/metabolismo , Inmunoterapia Adoptiva/métodos , Mutación/genética , Neoplasias Ováricas/inmunología , Linfocitos T/inmunología , Proteína p53 Supresora de Tumor/metabolismo , Antígenos de Neoplasias/genética , Células Cultivadas , Epítopos de Linfocito T/genética , Femenino , Antígeno HLA-A2/genética , Humanos , Persona de Mediana Edad , Estadificación de Neoplasias , Microambiente Tumoral , Proteína p53 Supresora de Tumor/genética , Secuenciación del Exoma
15.
Proc Natl Acad Sci U S A ; 113(42): E6409-E6417, 2016 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-27702896

RESUMEN

Genetically engineered mouse models (GEMMs) of cancer are increasingly being used to assess putative driver mutations identified by large-scale sequencing of human cancer genomes. To accurately interpret experiments that introduce additional mutations, an understanding of the somatic genetic profile and evolution of GEMM tumors is necessary. Here, we performed whole-exome sequencing of tumors from three GEMMs of lung adenocarcinoma driven by mutant epidermal growth factor receptor (EGFR), mutant Kirsten rat sarcoma viral oncogene homolog (Kras), or overexpression of MYC proto-oncogene. Tumors from EGFR- and Kras-driven models exhibited, respectively, 0.02 and 0.07 nonsynonymous mutations per megabase, a dramatically lower average mutational frequency than observed in human lung adenocarcinomas. Tumors from models driven by strong cancer drivers (mutant EGFR and Kras) harbored few mutations in known cancer genes, whereas tumors driven by MYC, a weaker initiating oncogene in the murine lung, acquired recurrent clonal oncogenic Kras mutations. In addition, although EGFR- and Kras-driven models both exhibited recurrent whole-chromosome DNA copy number alterations, the specific chromosomes altered by gain or loss were different in each model. These data demonstrate that GEMM tumors exhibit relatively simple somatic genotypes compared with human cancers of a similar type, making these autochthonous model systems useful for additive engineering approaches to assess the potential of novel mutations on tumorigenesis, cancer progression, and drug sensitivity.


Asunto(s)
Adenocarcinoma/genética , Transformación Celular Neoplásica/genética , Receptores ErbB/genética , Genes myc , Genes ras , Neoplasias Pulmonares/genética , Mutación , Adenocarcinoma/patología , Adenocarcinoma del Pulmón , Animales , Carcinógenos , Variaciones en el Número de Copia de ADN , Análisis Mutacional de ADN , Modelos Animales de Enfermedad , Dosificación de Gen , Estudio de Asociación del Genoma Completo , Neoplasias Pulmonares/patología , Ratones , Ratones Transgénicos , Mutación Puntual , Proto-Oncogenes Mas , Curva ROC , Secuenciación del Exoma
16.
J Immunol ; 194(6): 2635-42, 2015 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-25672759

RESUMEN

MicroRNA (miR)-mediated regulation of protein abundance is a pervasive mechanism of directing cellular processes. The well-studied and abundant miR-182 has previously been implicated in many aspects of T cell function, DNA repair, and cancer. In this study, we show that miR-182 is the most highly induced miR in B cells undergoing class-switch recombination. To elucidate the requirement of miR-182 in lymphocyte function, we extensively characterized mice with a targeted deletion of Mir182. We show that despite its dramatic induction, loss of miR-182 has minimal impact on B cell development, the ability of B cells to undergo class-switch recombination ex vivo and to undergo Ag-driven affinity maturation in vivo. Furthermore, in striking contrast to knockdown studies that demonstrated the requirement of miR-182 in T cell function, miR-182-deficient mice display no defect in T cell development and activation. Finally, we show that T cell-dependent immune response to experimental Listeria monocytogenes infection is intact in miR-182-deficient mice. We conclude that, contrary to previous studies, miR-182 does not play a significant role in all measured aspects of mouse adaptive immunity. This striking absence of a phenotype highlights the lack of correlation between expression pattern and functional requirement, underscores the limitations of using knockdown approaches to assess miR requirements, and suggests that miR networks may compensate for the chronic loss of specific miRs.


Asunto(s)
Inmunidad Adaptativa/inmunología , Linfocitos B/inmunología , Cambio de Clase de Inmunoglobulina/inmunología , MicroARNs/inmunología , Inmunidad Adaptativa/genética , Animales , Linfocitos B/metabolismo , Citometría de Flujo , Expresión Génica/inmunología , Interacciones Huésped-Patógeno/inmunología , Cambio de Clase de Inmunoglobulina/genética , Listeria monocytogenes/inmunología , Listeria monocytogenes/fisiología , Listeriosis/genética , Listeriosis/inmunología , Listeriosis/microbiología , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/genética , MicroARNs/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T/inmunología , Linfocitos T/metabolismo
17.
BMC Genomics ; 17: 548, 2016 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-27491780

RESUMEN

BACKGROUND: Numerous human genes encode potentially active DNA transposases or recombinases, but our understanding of their functions remains limited due to shortage of methods to profile their activities on endogenous genomic substrates. RESULTS: To enable functional analysis of human transposase-derived genes, we combined forward chemical genetic hypoxanthine-guanine phosphoribosyltransferase 1 (HPRT1) screening with massively parallel paired-end DNA sequencing and structural variant genome assembly and analysis. Here, we report the HPRT1 mutational spectrum induced by the human transposase PGBD5, including PGBD5-specific signal sequences (PSS) that serve as potential genomic rearrangement substrates. CONCLUSIONS: The discovered PSS motifs and high-throughput forward chemical genomic screening approach should prove useful for the elucidation of endogenous genome remodeling activities of PGBD5 and other domesticated human DNA transposases and recombinases.


Asunto(s)
Reordenamiento Génico , Pruebas Genéticas , Genoma Humano , Genómica , Transposasas/genética , Secuencia de Bases , Línea Celular , Expresión Génica , Genómica/métodos , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Hipoxantina Fosforribosiltransferasa/química , Hipoxantina Fosforribosiltransferasa/genética , Mutación , Señales de Clasificación de Proteína/genética , Análisis de Secuencia de ADN , Transposasas/química
18.
Nature ; 464(7287): 436-40, 2010 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-20173739

RESUMEN

Inhibition of an initiating oncogene often leads to extensive tumour cell death, a phenomenon known as oncogene addiction. This has led to the search for compounds that specifically target and inhibit oncogenes as anticancer agents. However, there has been no systematic exploration of whether chromosomal instability generated as a result of deregulation of the mitotic checkpoint pathway, a frequent characteristic of solid tumours, has any effect on oncogene addiction. Here we show that induction of chromosome instability by overexpression of the mitotic checkpoint gene Mad2 in mice does not affect the regression of Kras-driven lung tumours when Kras is inhibited. However, tumours that experience transient Mad2 overexpression and consequent chromosome instability recur at markedly elevated rates. The recurrent tumours are highly aneuploid and have varied activation of pro-proliferative pathways. Thus, early chromosomal instability may be responsible for tumour relapse after seemingly effective anticancer treatments.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Inestabilidad Cromosómica , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Recurrencia Local de Neoplasia/metabolismo , Oncogenes/fisiología , Proteínas Proto-Oncogénicas p21(ras)/deficiencia , Aneuploidia , Animales , Proteínas de Ciclo Celular/genética , Inestabilidad Cromosómica/genética , Neoplasias Pulmonares/genética , Proteínas Mad2 , Ratones , Ratones Transgénicos , Recurrencia Local de Neoplasia/patología , Oncogenes/genética , Proteínas Proto-Oncogénicas p21(ras)/antagonistas & inhibidores , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo
19.
Genes Chromosomes Cancer ; 54(10): 606-15, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26171757

RESUMEN

CTNNB1 mutations or APC abnormalities have been observed in ∼85% of desmoids examined by Sanger sequencing and are associated with Wnt/ß-catenin activation. We sought to identify molecular aberrations in "wild-type" tumors (those without CTNNB1 or APC alteration) and to determine their prognostic relevance. CTNNB1 was examined by Sanger sequencing in 117 desmoids; a mutation was observed in 101 (86%) and 16 were wild type. Wild-type status did not associate with tumor recurrence. Moreover, in unsupervised clustering based on U133A-derived gene expression profiles, wild-type and mutated tumors clustered together. Whole-exome sequencing of eight of the wild-type desmoids revealed that three had a CTNNB1 mutation that had been undetected by Sanger sequencing. The mutation was found in a mean 16% of reads (vs. 37% for mutations identified by Sanger). Of the other five wild-type tumors sequenced, two had APC loss, two had chromosome 6 loss, and one had mutation of BMI1. The finding of low-frequency CTNNB1 mutation or APC loss in wild-type desmoids was validated in the remaining eight wild-type desmoids; directed miSeq identified low-frequency CTNNB1 mutation in four and comparative genomic hybridization identified APC loss in one. These results demonstrate that mutations affecting CTNNB1 or APC occur more frequently in desmoids than previously recognized (111 of 117; 95%), and designation of wild-type genotype is largely determined by sensitivity of detection methods. Even true CTNNB1 wild-type tumors (determined by next-generation sequencing) may have genomic alterations associated with Wnt activation (chromosome 6 loss/BMI1 mutation), supporting Wnt/ß-catenin activation as the common pathway governing desmoid initiation.


Asunto(s)
Exoma , Fibromatosis Agresiva/genética , Proteínas Wnt/genética , beta Catenina/genética , Carcinogénesis/genética , Carcinogénesis/metabolismo , Cromosomas Humanos Par 6 , Fibromatosis Agresiva/fisiopatología , Dosificación de Gen , Genómica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Mutación
20.
N Engl J Med ; 366(12): 1079-89, 2012 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-22417203

RESUMEN

BACKGROUND: Acute myeloid leukemia (AML) is a heterogeneous disease with respect to presentation and clinical outcome. The prognostic value of recently identified somatic mutations has not been systematically evaluated in a phase 3 trial of treatment for AML. METHODS: We performed a mutational analysis of 18 genes in 398 patients younger than 60 years of age who had AML and who were randomly assigned to receive induction therapy with high-dose or standard-dose daunorubicin. We validated our prognostic findings in an independent set of 104 patients. RESULTS: We identified at least one somatic alteration in 97.3% of the patients. We found that internal tandem duplication in FLT3 (FLT3-ITD), partial tandem duplication in MLL (MLL-PTD), and mutations in ASXL1 and PHF6 were associated with reduced overall survival (P=0.001 for FLT3-ITD, P=0.009 for MLL-PTD, P=0.05 for ASXL1, and P=0.006 for PHF6); CEBPA and IDH2 mutations were associated with improved overall survival (P=0.05 for CEBPA and P=0.01 for IDH2). The favorable effect of NPM1 mutations was restricted to patients with co-occurring NPM1 and IDH1 or IDH2 mutations. We identified genetic predictors of outcome that improved risk stratification among patients with AML, independently of age, white-cell count, induction dose, and post-remission therapy, and validated the significance of these predictors in an independent cohort. High-dose daunorubicin, as compared with standard-dose daunorubicin, improved the rate of survival among patients with DNMT3A or NPM1 mutations or MLL translocations (P=0.001) but not among patients with wild-type DNMT3A, NPM1, and MLL (P=0.67). CONCLUSIONS: We found that DNMT3A and NPM1 mutations and MLL translocations predicted an improved outcome with high-dose induction chemotherapy in patients with AML. These findings suggest that mutational profiling could potentially be used for risk stratification and to inform prognostic and therapeutic decisions regarding patients with AML. (Funded by the National Cancer Institute and others.).


Asunto(s)
Análisis Mutacional de ADN , Quimioterapia de Inducción , Leucemia Mieloide Aguda/genética , Mutación , Medición de Riesgo/métodos , Adolescente , Adulto , Antibióticos Antineoplásicos/administración & dosificación , Dermatoglifia del ADN , Daunorrubicina/administración & dosificación , Duplicación de Gen , Humanos , Estimación de Kaplan-Meier , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/mortalidad , Persona de Mediana Edad , Nucleofosmina , Pronóstico , Translocación Genética , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA