Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 101
Filtrar
Más filtros

País/Región como asunto
Intervalo de año de publicación
1.
Cell ; 171(1): 14-17, 2017 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-28888324

RESUMEN

This year's Lasker-DeBakey Prize for Clinical Research to Douglas Lowy and John Schiller celebrates the science behind one of the greatest advances in the history of cancer research: the development of vaccines that prevent infection and thus prevent tumor induction by pathogenic strains of human papilloma virus (HPV).


Asunto(s)
Distinciones y Premios , Papillomavirus Humano 16/fisiología , Infecciones por Papillomavirus/prevención & control , Vacunas contra Papillomavirus/inmunología , Neoplasias del Cuello Uterino/prevención & control , Femenino , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Masculino , National Cancer Institute (U.S.) , Infecciones por Papillomavirus/inmunología , Infecciones por Papillomavirus/virología , Vacunas contra Papillomavirus/historia , Estados Unidos , Neoplasias del Cuello Uterino/inmunología , Neoplasias del Cuello Uterino/virología
2.
Cell ; 171(2): 495, 2017 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-28985568
3.
Proc Natl Acad Sci U S A ; 121(21): e2403685121, 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38743625

RESUMEN

The tumor suppressor LKB1 is a serine/threonine protein kinase that is frequently mutated in human lung adenocarcinoma (LUAD). LKB1 regulates a complex signaling network that is known to control cell polarity and metabolism; however, the pathways that mediate the tumor-suppressive activity of LKB1 are incompletely defined. To identify mechanisms of LKB1-mediated growth suppression, we developed a spheroid-based cell culture assay to study LKB1-dependent growth. We then performed genome-wide CRISPR screens in spheroidal culture and found that LKB1 suppresses growth, in part, by activating the PIKFYVE lipid kinase. Finally, we used chemical inhibitors and a pH-sensitive reporter to determine that LKB1 impairs growth by promoting the internalization of wild-type EGFR in a PIKFYVE-dependent manner.


Asunto(s)
Quinasas de la Proteína-Quinasa Activada por el AMP , Fosfatidilinositol 3-Quinasas , Proteínas Serina-Treonina Quinasas , Esferoides Celulares , Humanos , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Quinasas de la Proteína-Quinasa Activada por el AMP/metabolismo , Quinasas de la Proteína-Quinasa Activada por el AMP/genética , Esferoides Celulares/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Proliferación Celular , Línea Celular Tumoral , Sistemas CRISPR-Cas , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/metabolismo , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética
6.
Proc Natl Acad Sci U S A ; 115(44): E10437-E10446, 2018 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-30322915

RESUMEN

Mutations affecting the spliceosomal protein U2AF1 are commonly found in myelodysplastic syndromes (MDS) and secondary acute myeloid leukemia (sAML). We have generated mice that carry Cre-dependent knock-in alleles of U2af1(S34F), the murine version of the most common mutant allele of U2AF1 encountered in human cancers. Cre-mediated recombination in murine hematopoietic lineages caused changes in RNA splicing, as well as multilineage cytopenia, macrocytic anemia, decreased hematopoietic stem and progenitor cells, low-grade dysplasias, and impaired transplantability, but without lifespan shortening or leukemia development. In an attempt to identify U2af1(S34F)-cooperating changes that promote leukemogenesis, we combined U2af1(S34F) with Runx1 deficiency in mice and further treated the mice with a mutagen, N-ethyl-N-nitrosourea (ENU). Overall, 3 of 16 ENU-treated compound transgenic mice developed AML. However, AML did not arise in mice with other genotypes or without ENU treatment. Sequencing DNA from the three AMLs revealed somatic mutations homologous to those considered to be drivers of human AML, including predicted loss- or gain-of-function mutations in Tet2, Gata2, Idh1, and Ikzf1 However, the engineered U2af1(S34F) missense mutation reverted to WT in two of the three AML cases, implying that U2af1(S34F) is dispensable, or even selected against, once leukemia is established.


Asunto(s)
Hematopoyesis/genética , Leucemia/genética , Factor de Empalme U2AF/metabolismo , Alelos , Animales , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Etilnitrosourea/toxicidad , Regulación de la Expresión Génica/efectos de los fármacos , Predisposición Genética a la Enfermedad , Genotipo , Humanos , Ratones , Ratones Transgénicos , Mutación , Síndromes Mielodisplásicos/genética , Empalme del ARN , Factor de Empalme U2AF/genética
7.
Proc Natl Acad Sci U S A ; 115(26): E6030-E6038, 2018 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-29875142

RESUMEN

In ∼30% of patients with EGFR-mutant lung adenocarcinomas whose disease progresses on EGFR inhibitors, the basis for acquired resistance remains unclear. We have integrated transposon mutagenesis screening in an EGFR-mutant cell line and clinical genomic sequencing in cases of acquired resistance to identify mechanisms of resistance to EGFR inhibitors. The most prominent candidate genes identified by insertions in or near the genes during the screen were MET, a gene whose amplification is known to mediate resistance to EGFR inhibitors, and the gene encoding the Src family kinase YES1. Cell clones with transposon insertions that activated expression of YES1 exhibited resistance to all three generations of EGFR inhibitors and sensitivity to pharmacologic and siRNA-mediated inhibition of YES1 Analysis of clinical genomic sequencing data from cases of acquired resistance to EGFR inhibitors revealed amplification of YES1 in five cases, four of which lacked any other known mechanisms of resistance. Preinhibitor samples, available for two of the five patients, lacked YES1 amplification. None of 136 postinhibitor samples had detectable amplification of other Src family kinases (SRC and FYN). YES1 amplification was also found in 2 of 17 samples from ALK fusion-positive lung cancer patients who had progressed on ALK TKIs. Taken together, our findings identify acquired amplification of YES1 as a recurrent and targetable mechanism of resistance to EGFR inhibition in EGFR-mutant lung cancers and demonstrate the utility of transposon mutagenesis in discovering clinically relevant mechanisms of drug resistance.


Asunto(s)
Elementos Transponibles de ADN , Resistencia a Antineoplásicos , Inhibidores Enzimáticos/farmacología , Receptores ErbB , Amplificación de Genes , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias Pulmonares , Proteínas Proto-Oncogénicas c-yes , Línea Celular Tumoral , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Receptores ErbB/metabolismo , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Proteínas Proto-Oncogénicas c-fyn/genética , Proteínas Proto-Oncogénicas c-fyn/metabolismo , Proteínas Proto-Oncogénicas c-yes/biosíntesis , Proteínas Proto-Oncogénicas c-yes/genética , Proteínas Proto-Oncogénicas pp60(c-src)/genética , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo
8.
PLoS Genet ; 12(10): e1006384, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27776121

RESUMEN

We have asked how the common S34F mutation in the splicing factor U2AF1 regulates alternative splicing in lung cancer, and why wild-type U2AF1 is retained in cancers with this mutation. A human lung epithelial cell line was genetically modified so that U2AF1S34F is expressed from one of the two endogenous U2AF1 loci. By altering levels of mutant or wild-type U2AF1 in this cell line and by analyzing published data on human lung adenocarcinomas, we show that S34F-associated changes in alternative splicing are proportional to the ratio of S34F:wild-type gene products and not to absolute levels of either the mutant or wild-type factor. Preferential recognition of specific 3' splice sites in S34F-expressing cells is largely explained by differential in vitro RNA-binding affinities of mutant versus wild-type U2AF1 for those same 3' splice sites. Finally, we show that lung adenocarcinoma cell lines bearing U2AF1 mutations do not require the mutant protein for growth in vitro or in vivo. In contrast, wild-type U2AF1 is required for survival, regardless of whether cells carry the U2AF1S34F allele. Our results provide mechanistic explanations of the magnitude of splicing changes observed in U2AF1-mutant cells and why tumors harboring U2AF1 mutations always retain an expressed copy of the wild-type allele.


Asunto(s)
Adenocarcinoma/genética , Empalme Alternativo/genética , Proliferación Celular/genética , Neoplasias Pulmonares/genética , Factor de Empalme U2AF/genética , Adenocarcinoma/patología , Adenocarcinoma del Pulmón , Animales , Línea Celular Tumoral , Supervivencia Celular/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/patología , Ratones , Proteínas Mutantes/genética , Mutación , Motivos de Nucleótidos/genética , ARN Mensajero/biosíntesis , Factor de Empalme U2AF/biosíntesis , Transcriptoma/genética , Ensayos Antitumor por Modelo de Xenoinjerto
9.
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
10.
N Engl J Med ; 372(9): 793-5, 2015 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-25635347

RESUMEN

President Obama has announced a research initiative that aims to accelerate progress toward a new era of precision medicine, with a near-term focus on cancers and a longer-term aim to generate knowledge applicable to the whole range of health and disease.


Asunto(s)
Investigación Biomédica/legislación & jurisprudencia , Medicina de Precisión , Apoyo a la Investigación como Asunto/legislación & jurisprudencia , Investigación Biomédica/economía , Financiación Gubernamental , Salud , Humanos , Neoplasias/terapia , Estados Unidos
11.
Proc Natl Acad Sci U S A ; 111(16): 5773-7, 2014 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-24733905

RESUMEN

The long-held but erroneous assumption of never-ending rapid growth in biomedical science has created an unsustainable hypercompetitive system that is discouraging even the most outstanding prospective students from entering our profession--and making it difficult for seasoned investigators to produce their best work. This is a recipe for long-term decline, and the problems cannot be solved with simplistic approaches. Instead, it is time to confront the dangers at hand and rethink some fundamental features of the US biomedical research ecosystem.


Asunto(s)
Investigación Biomédica/organización & administración , Investigación Biomédica/economía , Directrices para la Planificación en Salud , Humanos , Investigadores/provisión & distribución , Apoyo a la Investigación como Asunto/economía , Estados Unidos , Recursos Humanos
12.
Genes Dev ; 23(14): 1677-88, 2009 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-19605689

RESUMEN

The advent of targeted therapies for cancer has provoked interest in experimental models for the systematic study of oncogene dependence. To that end, we developed a three-dimensional (3D) culture system to analyze the responses of primary mouse mammary epithelial cells to the induction and deinduction of oncogenes. Mammary cells derived from normal virgin mice, or from tritransgenic mice (TetO-MYC;TetO-Kras(G12D);MMTV-rtTA) in which MYC and mutant Kras can be regulated by doxycycline, develop from single cells into polarized acini. Lumen formation occurs without apparent apoptosis, and the hollow spheres of cells enlarge by division, with metaphase plates oriented perpendicularly to the apical surface. When MYC and Kras(G12D) are induced, the acini enlarge and form solid, depolarized spheres. Upon deinduction of MYC and Kras(G12D) the solid structures regress, leaving a repolarized monolayer of viable cells. These cells display a phenotype consistent with progenitors of mammary epithelium: They exclude Hoechst dye 33342, and reform acini in 3D cultures and repopulate mammary fat pads more efficiently than cells harvested from uninduced acini. Moreover, cells in the surviving spheres retain the ability to respond to reinduction and thus may represent the type of cells that give rise to recurrent tumors.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Supervivencia Celular/fisiología , Regulación de la Expresión Génica , Glándulas Mamarias Animales/citología , Oncogenes/genética , Transgenes/fisiología , Animales , Antibacterianos/farmacología , Apoptosis , Bencimidazoles/metabolismo , Caspasa 3/metabolismo , División Celular , Doxiciclina/farmacología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Ratones , Ratones Transgénicos , Mitocondrias/metabolismo , Células Madre/citología , Activación Transcripcional
13.
EMBO J ; 31(12): 2670-84, 2012 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-22617420

RESUMEN

Wnt genes and components of Wnt signalling pathways have been implicated in a wide spectrum of important biological phenomena, ranging from early organismal development to cell behaviours to several diseases, especially cancers. Emergence of the field of Wnt signalling can be largely traced back to the discovery of the first mammalian Wnt gene in 1982. In this essay, we mark the thirtieth anniversary of that discovery by describing some of the critical scientific developments that led to the flowering of this field of research.


Asunto(s)
Regulación de la Expresión Génica , Vía de Señalización Wnt , Animales , Investigación Biomédica/historia , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Modelos Biológicos
15.
Proc Natl Acad Sci U S A ; 109(47): 19408-13, 2012 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-23129625

RESUMEN

Bromodomain and extra terminal domain (BET) proteins function as epigenetic signaling factors that associate with acetylated histones and facilitate transcription of target genes. Inhibitors targeting the activity of BET proteins have shown potent antiproliferative effects in hematological cancers through the suppression of c-MYC and downstream target genes. However, as the epigenetic landscape of a cell varies drastically depending on lineage, transcriptional coactivators such as BETs would be expected to have different targets in cancers derived from different cells of origin, and this may influence the activity and mechanism of action of BET inhibitors. To test this hypothesis, we treated a panel of lung adenocarcinoma (LAC) cell lines with the BET inhibitor JQ1 and found that a subset is acutely susceptible to BET inhibition. In contrast to blood tumors, we show that LAC cells are inhibited by JQ1 through a mechanism independent of c-MYC down-regulation. Through gene expression profiling, we discovered that the oncogenic transcription factor FOSL1 and its targets are suppressed by JQ1 in a dose-dependant manner. Knockdown of BRD4 also decreased FOSL1 levels, and inhibition of FOSL1 phenocopied the effects of JQ1 treatment, suggesting that loss of this transcription factor may be partly responsible for the cytotoxic effects of BET inhibition in LAC cells, although ectopic expression of FOSL1 alone did not rescue the phenotype. Together, these findings suggest that BET inhibitors may be useful in solid tumors and that cell-lineage-specific differences in transcriptional targets of BETs may influence the activity of inhibitors of these proteins in different cancer types.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/genética , Azepinas/farmacología , Epigénesis Genética/efectos de los fármacos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Terapia Molecular Dirigida , Proteínas Nucleares/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Triazoles/farmacología , Adenocarcinoma/patología , Adenocarcinoma del Pulmón , Azepinas/uso terapéutico , Proteínas de Ciclo Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Ensayos de Selección de Medicamentos Antitumorales , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Neoplasias Pulmonares/patología , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Transducción de Señal/genética , Factores de Transcripción/metabolismo , Triazoles/uso terapéutico
16.
Proc Natl Acad Sci U S A ; 109(3): E119-26, 2012 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-22233804

RESUMEN

We have previously shown that all six members of the anti-apoptotic BCL2 gene family can cooperate with (myelocytomatosis oncogene) MYC in a mouse model of leukemia, but three of them are significantly less potent contributors to leukemogenicity than the other three. The protein encoded by one of these less potent genes, BCL2L10/BCLb, was recently shown to vary dramatically in many primary human cancers by immunohistochemistry, and the protein levels were inversely correlated with survival in patients with several cancer types. We examined BCLb mRNA in a panel of human cancer cell lines and did not observe the extensive variation in mRNA that would be required to explain the vast differences in protein levels. We found that the levels of BCLb protein diminish quickly after inhibition of protein synthesis with cycloheximide, so we searched for interacting proteins that might affect posttranslational stability of BCLb. Using a variety of approaches, including immunoaffinity and mass spectrometry, we identified a protein, Ubiquilin1 (Ubqln), that specifically interacts with BCLb, and not with other anti-apoptotic BCL2-like proteins. Ubqln stabilizes BCLb protein, while also promoting monoubiquitination on multiple lysine residues and relocation to the cytosol. Furthermore, primary lung adencarcinomas have more Ubqln mRNA than normal adjacent lung tissue, and higher Ubqln mRNA levels are associated with shorter survival of lung cancer patients, suggesting that potentiation of the anti-apoptotic potential of BCLb through regulation of its stability by Ubqln may be an important factor in tumor progression.


Asunto(s)
Apoptosis , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ubiquitinación , Proteínas Adaptadoras Transductoras de Señales , Adenocarcinoma/genética , Adenocarcinoma/patología , Adenocarcinoma del Pulmón , Animales , Proteínas Relacionadas con la Autofagia , Proteínas Portadoras/genética , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Citoplasma/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Lisina/metabolismo , Ratones , Modelos Biológicos , Unión Proteica , Estabilidad Proteica , Transporte de Proteínas , Proteínas Proto-Oncogénicas c-bcl-2/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Fracciones Subcelulares/metabolismo , Análisis de Supervivencia , Ubiquitinas/metabolismo
18.
Proc Natl Acad Sci U S A ; 108(40): 16753-8, 2011 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-21940500

RESUMEN

The gene encoding the receptor for hyaluronan-mediated motility (RHAMM) is overexpressed in many human cancers. However, it is unclear whether RHAMM plays a causal role in tumor initiation or progression. Using somatic gene transfer in a mouse model of islet cell tumorigenesis, we demonstrate that RHAMM isoform B (RHAMM(B)) promotes tumor growth and metastases to lymph nodes and the liver. The propensity of RHAMM(B)-expressing cells to metastasize to the liver was confirmed using an experimental metastasis assay in which cells were injected into the tail vein of immunodeficient mice. However, RHAMM(B) did not increase cell migration or proliferation in culture. In initial efforts to identify signaling pathways activated by RHAMM(B), we found that RHAMM(B) induced phosphorylation of epidermal growth factor receptor (EGFR), Erk1/2, and STAT3 and conferred susceptibility to apoptosis after treatment with an EGFR inhibitor, gefitinib. Taken together, the results indicate that RHAMM(B) promotes hepatic metastasis by islet tumor cells, perhaps through growth factor receptor-mediated signaling.


Asunto(s)
Adenoma de Células de los Islotes Pancreáticos/patología , Adenoma de Células de los Islotes Pancreáticos/fisiopatología , Proteínas de la Matriz Extracelular/metabolismo , Receptores de Hialuranos/metabolismo , Neoplasias Hepáticas/secundario , Transducción de Señal/fisiología , Animales , Western Blotting , Ensayo de Inmunoadsorción Enzimática , Receptores ErbB/metabolismo , Técnicas de Transferencia de Gen , Inmunohistoquímica , Inmunoprecipitación , Ratones , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosforilación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT3/metabolismo
19.
Proc Natl Acad Sci U S A ; 108(39): 16375-80, 2011 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-21930909

RESUMEN

We previously described four small molecules that reduced the growth of lung adenocarcinoma cell lines with either epidermal growth factor receptor (EGFR) or KRAS mutations in a high-throughout chemical screen. By combining affinity proteomics and gene expression analysis, we now propose superoxide dismutase 1 (SOD1) as the most likely target of one of these small molecules, referred to as lung cancer screen 1 (LCS-1). siRNAs against SOD1 slowed the growth of LCS-1 sensitive cell lines; conversely, expression of a SOD1 cDNA increased proliferation of H358 cells and reduced sensitivity of these cells to LCS-1. In addition, SOD1 enzymatic activity was inhibited in vitro by LCS-1 and two closely related analogs. These results suggest that SOD1 is an LCS-1-binding protein that may act in concert with mutant proteins, such as EGFR and KRAS, to promote cell growth, providing a therapeutic target for compounds like LCS-1.


Asunto(s)
Adenocarcinoma/patología , División Celular/efectos de los fármacos , Neoplasias Pulmonares/patología , Superóxido Dismutasa/metabolismo , Adenocarcinoma/enzimología , Línea Celular Tumoral , Perfilación de la Expresión Génica , Humanos , Neoplasias Pulmonares/enzimología , ARN Interferente Pequeño/genética , Superóxido Dismutasa/efectos de los fármacos , Superóxido Dismutasa/genética , Superóxido Dismutasa-1
20.
Science ; 383(6683): eadj1415, 2024 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-38330136

RESUMEN

Lung adenocarcinoma (LUAD) and small cell lung cancer (SCLC) are thought to originate from different epithelial cell types in the lung. Intriguingly, LUAD can histologically transform into SCLC after treatment with targeted therapies. In this study, we designed models to follow the conversion of LUAD to SCLC and found that the barrier to histological transformation converges on tolerance to Myc, which we implicate as a lineage-specific driver of the pulmonary neuroendocrine cell. Histological transformations are frequently accompanied by activation of the Akt pathway. Manipulating this pathway permitted tolerance to Myc as an oncogenic driver, producing rare, stem-like cells that transcriptionally resemble the pulmonary basal lineage. These findings suggest that histological transformation may require the plasticity inherent to the basal stem cell, enabling tolerance to previously incompatible oncogenic driver programs.


Asunto(s)
Adenocarcinoma del Pulmón , Neoplasias Pulmonares , Proteínas Proto-Oncogénicas c-akt , Proteínas Proto-Oncogénicas c-myc , Carcinoma Pulmonar de Células Pequeñas , Humanos , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/patología , Adenocarcinoma del Pulmón/terapia , Células Epiteliales/patología , Pulmón/patología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/terapia , Carcinoma Pulmonar de Células Pequeñas/genética , Carcinoma Pulmonar de Células Pequeñas/patología , Carcinoma Pulmonar de Células Pequeñas/terapia , Oncogenes , Linaje de la Célula , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-akt/genética , Terapia Molecular Dirigida
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA