Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Nat Biotechnol ; 42(3): 424-436, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37169967

RESUMEN

Genetically engineered mouse models only capture a small fraction of the genetic lesions that drive human cancer. Current CRISPR-Cas9 models can expand this fraction but are limited by their reliance on error-prone DNA repair. Here we develop a system for in vivo prime editing by encoding a Cre-inducible prime editor in the mouse germline. This model allows rapid, precise engineering of a wide range of mutations in cell lines and organoids derived from primary tissues, including a clinically relevant Kras mutation associated with drug resistance and Trp53 hotspot mutations commonly observed in pancreatic cancer. With this system, we demonstrate somatic prime editing in vivo using lipid nanoparticles, and we model lung and pancreatic cancer through viral delivery of prime editing guide RNAs or orthotopic transplantation of prime-edited organoids. We believe that this approach will accelerate functional studies of cancer-associated mutations and complex genetic combinations that are challenging to construct with traditional models.


Asunto(s)
Neoplasias Pancreáticas , ARN Guía de Sistemas CRISPR-Cas , Ratones , Humanos , Animales , Ratones Transgénicos , Mutación/genética , Neoplasias Pancreáticas/genética , Línea Celular , Edición Génica , Sistemas CRISPR-Cas/genética
2.
Nat Commun ; 13(1): 256, 2022 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-35017504

RESUMEN

The GATA4 transcription factor acts as a master regulator of development of multiple tissues. GATA4 also acts in a distinct capacity to control a stress-inducible pro-inflammatory secretory program that is associated with senescence, a potent tumor suppression mechanism, but also operates in non-senescent contexts such as tumorigenesis. This secretory pathway is composed of chemokines, cytokines, growth factors, and proteases. Since GATA4 is deleted or epigenetically silenced in cancer, here we examine the role of GATA4 in tumorigenesis in mouse models through both loss-of-function and overexpression experiments. We find that GATA4 promotes non-cell autonomous tumor suppression in multiple model systems. Mechanistically, we show that Gata4-dependent tumor suppression requires cytotoxic CD8 T cells and partially requires the secreted chemokine CCL2. Analysis of transcriptome data in human tumors reveals reduced lymphocyte infiltration in GATA4-deficient tumors, consistent with our murine data. Notably, activation of the GATA4-dependent secretory program combined with an anti-PD-1 antibody robustly abrogates tumor growth in vivo.


Asunto(s)
Transporte Biológico/fisiología , Factor de Transcripción GATA4/metabolismo , Neoplasias/metabolismo , Linfocitos T Citotóxicos/metabolismo , Animales , Anticuerpos Monoclonales Humanizados , Quimiocina CCL2/metabolismo , Factor de Transcripción GATA4/genética , Regulación Neoplásica de la Expresión Génica , Proteínas de Homeodominio , Humanos , Evasión Inmune , Pulmón/patología , Melanoma , Ratones , Ratones Endogámicos C57BL , Miocitos Cardíacos/metabolismo , Neoplasias/inmunología , Neoplasias/patología , Transcriptoma
3.
Cancer Cell ; 39(10): 1342-1360.e14, 2021 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-34358448

RESUMEN

The CD155/TIGIT axis can be co-opted during immune evasion in chronic viral infections and cancer. Pancreatic adenocarcinoma (PDAC) is a highly lethal malignancy, and immune-based strategies to combat this disease have been largely unsuccessful to date. We corroborate prior reports that a substantial portion of PDAC harbors predicted high-affinity MHC class I-restricted neoepitopes and extend these findings to advanced/metastatic disease. Using multiple preclinical models of neoantigen-expressing PDAC, we demonstrate that intratumoral neoantigen-specific CD8+ T cells adopt multiple states of dysfunction, resembling those in tumor-infiltrating lymphocytes of PDAC patients. Mechanistically, genetic and/or pharmacologic modulation of the CD155/TIGIT axis was sufficient to promote immune evasion in autochthonous neoantigen-expressing PDAC. Finally, we demonstrate that the CD155/TIGIT axis is critical in maintaining immune evasion in PDAC and uncover a combination immunotherapy (TIGIT/PD-1 co-blockade plus CD40 agonism) that elicits profound anti-tumor responses in preclinical models, now poised for clinical evaluation.


Asunto(s)
Evasión Inmune/inmunología , Inmunoterapia/métodos , Linfocitos Infiltrantes de Tumor/metabolismo , Neoplasias Pancreáticas/inmunología , Receptores Virales/inmunología , Animales , Humanos , Ratones , Neoplasias Pancreáticas
5.
Nat Cancer ; 1(6): 589-602, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-34414377

RESUMEN

Approximately 20-30% of human lung adenocarcinomas (LUAD) harbor loss-of-function (LOF) mutations in Kelch-like ECH Associated-Protein 1 (KEAP1), which lead to hyperactivation of the nuclear factor, erythroid 2-like 2 (NRF2) antioxidant pathway and correlate with poor prognosis1-3. We previously showed that Keap1 mutation accelerates KRAS-driven LUAD and produces a marked dependency on glutaminolysis4. To extend the investigation of genetic dependencies in the context of Keap1 mutation, we performed a druggable genome CRISPR-Cas9 screen in Keap1-mutant cells. This analysis uncovered a profound Keap1 mutant-specific dependency on solute carrier family 33 member 1 (Slc33a1), an endomembrane-associated protein with roles in autophagy regulation5, as well as a series of functionally-related genes implicated in the unfolded protein response. Targeted genetic and biochemical experiments using mouse and human Keap1-mutant tumor lines, as well as preclinical genetically-engineered mouse models (GEMMs) of LUAD, validate Slc33a1 as a robust Keap1-mutant-specific dependency. Furthermore, unbiased genome-wide CRISPR screening identified additional genes related to Slc33a1 dependency. Overall, our study provides a strong rationale for stratification of patients harboring KEAP1-mutant or NRF2-hyperactivated tumors as likely responders to targeted SLC33A1 inhibition and underscores the value of integrating functional genetic approaches with GEMMs to identify and validate genotype-specific therapeutic targets.


Asunto(s)
Adenocarcinoma del Pulmón , Proteína 1 Asociada A ECH Tipo Kelch , Neoplasias Pulmonares , Proteínas de Transporte de Membrana , Adenocarcinoma del Pulmón/genética , Animales , Humanos , Proteína 1 Asociada A ECH Tipo Kelch/genética , Neoplasias Pulmonares/genética , Proteínas de Transporte de Membrana/genética , Ratones , Mutación , Factor 2 Relacionado con NF-E2/genética
6.
Proc Natl Acad Sci U S A ; 117(1): 513-521, 2020 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-31871154

RESUMEN

Small cell lung cancer (SCLC) is a highly aggressive subtype of lung cancer that remains among the most lethal of solid tumor malignancies. Recent genomic sequencing studies have identified many recurrently mutated genes in human SCLC tumors. However, the functional roles of most of these genes remain to be validated. Here, we have adapted the CRISPR-Cas9 system to a well-established murine model of SCLC to rapidly model loss-of-function mutations in candidate genes identified from SCLC sequencing studies. We show that loss of the gene p107 significantly accelerates tumor progression. Notably, compared with loss of the closely related gene p130, loss of p107 results in fewer but larger tumors as well as earlier metastatic spread. In addition, we observe differences in proliferation and apoptosis as well as altered distribution of initiated tumors in the lung, resulting from loss of p107 or p130 Collectively, these data demonstrate the feasibility of using the CRISPR-Cas9 system to model loss of candidate tumor suppressor genes in SCLC, and we anticipate that this approach will facilitate efforts to investigate mechanisms driving tumor progression in this deadly disease.


Asunto(s)
Edición Génica/métodos , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor , Neoplasias Pulmonares/genética , Carcinoma Pulmonar de Células Pequeñas/genética , Animales , Apoptosis/genética , Sistemas CRISPR-Cas/genética , Línea Celular , Proliferación Celular/genética , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Estudios de Factibilidad , Humanos , Mutación con Pérdida de Función , Pulmón/patología , Neoplasias Pulmonares/patología , Ratones , Ratones Transgénicos , Estadificación de Neoplasias , Proteína p107 Similar a la del Retinoblastoma/genética , Proteína p130 Similar a la del Retinoblastoma/genética , Carcinoma Pulmonar de Células Pequeñas/patología , Carga Tumoral/genética , Proteína p53 Supresora de Tumor/genética
7.
Sci Transl Med ; 11(517)2019 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-31694929

RESUMEN

Small cell lung cancer (SCLC) is an aggressive lung cancer subtype with extremely poor prognosis. No targetable genetic driver events have been identified, and the treatment landscape for this disease has remained nearly unchanged for over 30 years. Here, we have taken a CRISPR-based screening approach to identify genetic vulnerabilities in SCLC that may serve as potential therapeutic targets. We used a single-guide RNA (sgRNA) library targeting ~5000 genes deemed to encode "druggable" proteins to perform loss-of-function genetic screens in a panel of cell lines derived from autochthonous genetically engineered mouse models (GEMMs) of SCLC, lung adenocarcinoma (LUAD), and pancreatic ductal adenocarcinoma (PDAC). Cross-cancer analyses allowed us to identify SCLC-selective vulnerabilities. In particular, we observed enhanced sensitivity of SCLC cells toward disruption of the pyrimidine biosynthesis pathway. Pharmacological inhibition of dihydroorotate dehydrogenase (DHODH), a key enzyme in this pathway, reduced the viability of SCLC cells in vitro and strongly suppressed SCLC tumor growth in human patient-derived xenograft (PDX) models and in an autochthonous mouse model. These results indicate that DHODH inhibition may be an approach to treat SCLC.


Asunto(s)
Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/enzimología , Terapia Molecular Dirigida , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/antagonistas & inhibidores , Carcinoma Pulmonar de Células Pequeñas/tratamiento farmacológico , Carcinoma Pulmonar de Células Pequeñas/enzimología , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/enzimología , Adenocarcinoma/patología , Animales , Compuestos de Bifenilo/farmacología , Compuestos de Bifenilo/uso terapéutico , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/enzimología , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , DCMP Desaminasa/metabolismo , Dihidroorotato Deshidrogenasa , Progresión de la Enfermedad , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Humanos , Neoplasias Pulmonares/patología , Ratones , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/metabolismo , Neoplasias Pancreáticas/metabolismo , Pirimidinas/biosíntesis , Carcinoma Pulmonar de Células Pequeñas/patología , Análisis de Supervivencia , Ensayos Antitumor por Modelo de Xenoinjerto , Neoplasias Pancreáticas
8.
Genes Dev ; 24(8): 837-52, 2010 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-20395368

RESUMEN

Chemotherapy resistance is a major obstacle in cancer treatment, yet the mechanisms of response to specific therapies have been largely unexplored in vivo. Employing genetic, genomic, and imaging approaches, we examined the dynamics of response to a mainstay chemotherapeutic, cisplatin, in multiple mouse models of human non-small-cell lung cancer (NSCLC). We show that lung tumors initially respond to cisplatin by sensing DNA damage, undergoing cell cycle arrest, and inducing apoptosis-leading to a significant reduction in tumor burden. Importantly, we demonstrate that this response does not depend on the tumor suppressor p53 or its transcriptional target, p21. Prolonged cisplatin treatment promotes the emergence of resistant tumors with enhanced repair capacity that are cross-resistant to platinum analogs, exhibit advanced histopathology, and possess an increased frequency of genomic alterations. Cisplatin-resistant tumors express elevated levels of multiple DNA damage repair and cell cycle arrest-related genes, including p53-inducible protein with a death domain (Pidd). We demonstrate a novel role for PIDD as a regulator of chemotherapy response in human lung tumor cells.


Asunto(s)
Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Cisplatino/farmacología , Cisplatino/uso terapéutico , Reparación del ADN/efectos de los fármacos , Neoplasias Pulmonares/tratamiento farmacológico , Animales , Carcinoma de Pulmón de Células no Pequeñas/patología , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , Proteínas Adaptadoras de Señalización del Receptor del Dominio de Muerte , Modelos Animales de Enfermedad , Resistencia a Antineoplásicos/fisiología , Perfilación de la Expresión Génica , Humanos , Neoplasias Pulmonares/patología , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos
9.
Science ; 327(5965): 593-6, 2010 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-20019247

RESUMEN

Acute exposure to ionizing radiation can cause lethal damage to the gastrointestinal (GI) tract, a condition called the GI syndrome. Whether the target cells affected by radiation to cause the GI syndrome are derived from the epithelium or endothelium and whether the target cells die by apoptosis or other mechanisms are controversial issues. Studying mouse models, we found that selective deletion of the proapoptotic genes Bak1 and Bax from the GI epithelium or from endothelial cells did not protect mice from developing the GI syndrome after sub-total-body gamma irradiation. In contrast, selective deletion of p53 from the GI epithelium, but not from endothelial cells, sensitized irradiated mice to the GI syndrome. Transgenic mice overexpressing p53 in all tissues were protected from the GI syndrome after irradiation. These results suggest that the GI syndrome is caused by the death of GI epithelial cells and that these epithelial cells die by a mechanism that is regulated by p53 but independent of apoptosis.


Asunto(s)
Apoptosis , Rayos gamma/efectos adversos , Enfermedades Intestinales/fisiopatología , Mucosa Intestinal/efectos de la radiación , Intestino Delgado/efectos de la radiación , Traumatismos por Radiación/fisiopatología , Proteína p53 Supresora de Tumor/fisiología , Animales , Muerte Celular , Células Epiteliales/citología , Células Epiteliales/fisiología , Células Epiteliales/efectos de la radiación , Eliminación de Gen , Genes p53 , Enfermedades Intestinales/etiología , Enfermedades Intestinales/patología , Mucosa Intestinal/patología , Mucosa Intestinal/fisiopatología , Intestino Delgado/patología , Intestino Delgado/fisiopatología , Mesodermo/citología , Ratones , Ratones Transgénicos , Modelos Biológicos , Dosis de Radiación , Traumatismos por Radiación/etiología , Traumatismos por Radiación/patología , Proteína Destructora del Antagonista Homólogo bcl-2/genética , Proteína Destructora del Antagonista Homólogo bcl-2/metabolismo , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo
10.
Carcinogenesis ; 28(12): 2589-96, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17690114

RESUMEN

Oncogenic K-ras is one of the most common genetic alterations in human lung adenocarcinomas. In addition, inactivation of clusters of tumor suppressor genes is required to bring about classical characteristics of cancer including angiogenesis as a prelude to invasion and metastasis. Transforming growth factor-beta (TGF-beta) 1 is a tumor suppressor gene that is implicated in lung cancer progression. Although in vitro studies have shown that TGF-beta1 and Ras pathways cooperate during tumorigenesis, the biology of interaction of TGF-beta1 and Ras has not been studied in in vivo tumorigenesis. We hypothesized that inactivation of TGF-beta1 in addition to oncogeneic activation of K-ras would lead to early initiation and faster progression to lung adenocarcinoma and invasion and metastasis. Heterozygous (HT) TGF-beta1 mice were mated with latent activatable (LA) mutated K-ras mice to generate TGF-beta1(+/+), K-ras LA (wild-type (WT)/LA) and TGF-beta1(+/-), K-ras LA (HT/LA) mice. Both HT/LA and WT/LA mice developed spontaneous lung tumors, but HT/LA mice progressed to adenocarcinomas significantly earlier compared with WT/LA mice. In addition, WT/LA adenocarcinomas had significantly higher angiogenic activity compared with HT/LA adenocarcinomas. Thus, while oncogenic K-ras mutation and insensitivity to the growth regulatory effects of TGF-beta1 is essential for initiation and progression of mouse lung tumors to adenocarcinoma, a full gene dosage of TGF-beta1 is required for tumor-induced angiogenesis and invasive potential. This study identifies a number of genes not previously associated with lung cancer that are involved in tumor induction and progression. In addition, we provide evidence that progression to invasive angiogenic lesions requires TGF-beta1 responsiveness in addition to Ras mutation.


Asunto(s)
Adenocarcinoma/metabolismo , Genes ras/fisiología , Neoplasias Pulmonares/metabolismo , Factor de Crecimiento Transformador beta1/fisiología , Adenocarcinoma/irrigación sanguínea , Adenocarcinoma/genética , Animales , Progresión de la Enfermedad , Heterocigoto , Neoplasias Pulmonares/irrigación sanguínea , Neoplasias Pulmonares/genética , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Mutación , Neovascularización Patológica , Análisis de Secuencia por Matrices de Oligonucleótidos , Factor de Crecimiento Transformador beta1/genética
11.
Nat Genet ; 39(5): 673-7, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17401365

RESUMEN

MicroRNAs (miRNAs) are a new class of small noncoding RNAs that post-transcriptionally regulate the expression of target mRNA transcripts. Many of these target mRNA transcripts are involved in proliferation, differentiation and apoptosis, processes commonly altered during tumorigenesis. Recent work has shown a global decrease of mature miRNA expression in human cancers. However, it is unclear whether this global repression of miRNAs reflects the undifferentiated state of tumors or causally contributes to the transformed phenotype. Here we show that global repression of miRNA maturation promotes cellular transformation and tumorigenesis. Cancer cells expressing short hairpin RNAs (shRNAs) targeting three different components of the miRNA processing machinery showed a substantial decrease in steady-state miRNA levels and a more pronounced transformed phenotype. In animals, miRNA processing-impaired cells formed tumors with accelerated kinetics. These tumors were more invasive than control tumors, suggesting that global miRNA loss enhances tumorigenesis. Furthermore, conditional deletion of Dicer1 enhanced tumor development in a K-Ras-induced mouse model of lung cancer. Overall, these studies indicate that abrogation of global miRNA processing promotes tumorigenesis.


Asunto(s)
Transformación Celular Neoplásica/genética , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor , MicroARNs/metabolismo , Neoplasias/genética , Animales , Bromodesoxiuridina , Pruebas de Carcinogenicidad , Línea Celular Tumoral , Citometría de Flujo , Humanos , Immunoblotting , Luciferasas , Ratones , MicroARNs/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ribonucleasa III/genética , Ribonucleasa III/metabolismo
12.
Cancer Cell ; 5(4): 375-87, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15093544

RESUMEN

Activating mutations in the ras oncogene are not considered sufficient to induce abnormal cellular proliferation in the absence of cooperating oncogenes. We demonstrate that the conditional expression of an endogenous K-ras(G12D) allele in murine embryonic fibroblasts causes enhanced proliferation and partial transformation in the absence of further genetic abnormalities. Interestingly, K-ras(G12D)-expressing fibroblasts demonstrate attenuation and altered regulation of canonical Ras effector signaling pathways. Widespread expression of endogenous K-ras(G12D) is not tolerated during embryonic development, and directed expression in the lung and GI tract induces preneoplastic epithelial hyperplasias. Our results suggest that endogenous oncogenic ras is sufficient to initiate transformation by stimulating proliferation, while further genetic lesions may be necessary for progression to frank malignancy.


Asunto(s)
Transformación Celular Neoplásica , Anomalías Congénitas/genética , Fibroblastos/patología , Regulación del Desarrollo de la Expresión Génica/fisiología , Genes ras/fisiología , Neoplasias/genética , Animales , Ciclo Celular , División Celular , Senescencia Celular , Anomalías Congénitas/patología , Cruzamientos Genéticos , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Embrión de Mamíferos/citología , Femenino , Fibroblastos/metabolismo , Integrasas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Neoplasias/patología , Células Madre/patología , Proteína p14ARF Supresora de Tumor/genética , Proteína p14ARF Supresora de Tumor/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Virales/metabolismo
13.
Proc Natl Acad Sci U S A ; 99(26): 16865-70, 2002 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-12486224

RESUMEN

Mice heterozygous for the retinoblastoma (Rb) tumor suppressor gene develop pituitary and thyroid tumors with high penetrance. We demonstrate here that loss of the ARF tumor suppressor strongly accelerates intermediate lobe pituitary tumorigenesis in Rb heterozygous mice. These effects in the pituitary are greater than those conferred by p53 loss in that Rb+-;ARF-- mice display significantly more early atypical lesions than Rb+-; p53-- mice. Also, Rb+-;ARF-- compound mutants do not develop many of the novel tumors or precancerous lesions seen in Rb+-;p53-- compound mutants. Although complete loss of ARF expression is not obligatory for pituitary tumorigenesis in Rb+- mice, alterations of the ARF locus are observed in tumors from Rb+-;ARF+- mice, consistent with a selective advantage of ARF inactivation in this context. We conclude that inactivation of ARF acts more broadly than that of p53 in connecting abrogation of the Rb pathway to tumorigenesis.


Asunto(s)
Genes de Retinoblastoma/fisiología , Mutación , Neoplasias Hipofisarias/genética , Proteína p14ARF Supresora de Tumor/genética , Animales , División Celular , Mapeo Cromosómico , Genes p53/fisiología , Pérdida de Heterocigocidad , Ratones , Neoplasias Hipofisarias/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA