Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Mol Cell Proteomics ; 21(7): 100247, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35594991

RESUMO

Since the discovery of oncogenes, there has been tremendous interest to understand their mechanistic basis and to develop broadly actionable therapeutics. Some of the most frequently activated oncogenes driving diverse cancers are c-MYC, EGFR, HER2, AKT, KRAS, BRAF, and MEK. Using a reductionist approach, we explored how cellular proteomes are remodeled in isogenic cell lines engineered with or without these driver oncogenes. The most striking discovery for all oncogenic models was the systematic downregulation of scores of antiviral proteins regulated by type 1 interferon. These findings extended to cancer cell lines and patient-derived xenograft models of highly refractory pancreatic cancer and osteosarcoma driven by KRAS and MYC oncogenes. The oncogenes reduced basal expression of and autocrine stimulation by type 1 interferon causing remarkable convergence on common phenotypic and functional profiles. In particular, there was dramatically lower expression of dsRNA sensors including DDX58 (RIG-I) and OAS proteins, which resulted in attenuated functional responses when the oncogenic cells were treated with the dsRNA mimetic, polyI:C, and increased susceptibility to infection with an RNA virus shown using SARS-CoV-2. Our reductionist approach provides molecular and functional insights connected to immune evasion hallmarks in cancers and suggests therapeutic opportunities.


Assuntos
COVID-19 , Interferon beta , Oncogenes , Proteômica , Animais , Fatores de Restrição Antivirais , COVID-19/imunologia , Carcinogênese , Linhagem Celular Tumoral , Humanos , Interferon beta/imunologia , Proteínas Proto-Oncogênicas p21(ras)/genética , SARS-CoV-2
2.
Genes Dev ; 24(8): 837-52, 2010 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-20395368

RESUMO

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.


Assuntos
Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Cisplatino/farmacologia , Cisplatino/uso terapêutico , Reparo do DNA/efeitos dos fármacos , Neoplasias Pulmonares/tratamento farmacológico , Animais , Carcinoma Pulmonar de Células não Pequenas/patologia , Proteínas de Transporte/metabolismo , Linhagem Celular Tumoral , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte , Modelos Animais de Doenças , Resistencia a Medicamentos Antineoplásicos/fisiologia , Perfilação da Expressão Gênica , Humanos , Neoplasias Pulmonares/patologia , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos
3.
Nat Genet ; 56(6): 1300-1309, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38724748

RESUMO

Concurrent readout of sequence and base modifications from long unamplified DNA templates by Pacific Biosciences of California (PacBio) single-molecule sequencing requires large amounts of input material. Here we adapt Tn5 transposition to introduce hairpin oligonucleotides and fragment (tagment) limiting quantities of DNA for generating PacBio-compatible circular molecules. We developed two methods that implement tagmentation and use 90-99% less input than current protocols: (1) single-molecule real-time sequencing by tagmentation (SMRT-Tag), which allows detection of genetic variation and CpG methylation; and (2) single-molecule adenine-methylated oligonucleosome sequencing assay by tagmentation (SAMOSA-Tag), which uses exogenous adenine methylation to add a third channel for probing chromatin accessibility. SMRT-Tag of 40 ng or more human DNA (approximately 7,000 cell equivalents) yielded data comparable to gold standard whole-genome and bisulfite sequencing. SAMOSA-Tag of 30,000-50,000 nuclei resolved single-fiber chromatin structure, CTCF binding and DNA methylation in patient-derived prostate cancer xenografts and uncovered metastasis-associated global epigenome disorganization. Tagmentation thus promises to enable sensitive, scalable and multimodal single-molecule genomics for diverse basic and clinical applications.


Assuntos
Metilação de DNA , Neoplasias da Próstata , Humanos , Animais , Masculino , Neoplasias da Próstata/genética , Camundongos , Análise de Sequência de DNA/métodos , Cromatina/genética , DNA/genética , Elementos de DNA Transponíveis/genética , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Ilhas de CpG/genética , Linhagem Celular Tumoral , Fator de Ligação a CCCTC/genética , Fator de Ligação a CCCTC/metabolismo , Transposases
4.
Clin Cancer Res ; 30(4): 849-864, 2024 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-37703185

RESUMO

PURPOSE: Models to study metastatic disease in rare cancers are needed to advance preclinical therapeutics and to gain insight into disease biology. Osteosarcoma is a rare cancer with a complex genomic landscape in which outcomes for patients with metastatic disease are poor. As osteosarcoma genomes are highly heterogeneous, multiple models are needed to fully elucidate key aspects of disease biology and to recapitulate clinically relevant phenotypes. EXPERIMENTAL DESIGN: Matched patient samples, patient-derived xenografts (PDX), and PDX-derived cell lines were comprehensively evaluated using whole-genome sequencing and RNA sequencing. The in vivo metastatic phenotype of the PDX-derived cell lines was characterized in both an intravenous and an orthotopic murine model. As a proof-of-concept study, we tested the preclinical effectiveness of a cyclin-dependent kinase inhibitor on the growth of metastatic tumors in an orthotopic amputation model. RESULTS: PDXs and PDX-derived cell lines largely maintained the expression profiles of the patient from which they were derived despite the emergence of whole-genome duplication in a subset of cell lines. The cell lines were heterogeneous in their metastatic capacity, and heterogeneous tissue tropism was observed in both intravenous and orthotopic models. Single-agent dinaciclib was effective at dramatically reducing the metastatic burden. CONCLUSIONS: The variation in metastasis predilection sites between osteosarcoma PDX-derived cell lines demonstrates their ability to recapitulate the spectrum of the disease observed in patients. We describe here a panel of new osteosarcoma PDX-derived cell lines that we believe will be of wide use to the osteosarcoma research community.


Assuntos
Neoplasias Ósseas , Óxidos N-Cíclicos , Indolizinas , Osteossarcoma , Compostos de Piridínio , Humanos , Animais , Camundongos , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Ensaios Antitumorais Modelo de Xenoenxerto , Osteossarcoma/tratamento farmacológico , Osteossarcoma/genética , Osteossarcoma/metabolismo , Linhagem Celular Tumoral , Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/genética , Neoplasias Ósseas/metabolismo
5.
bioRxiv ; 2023 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-36711882

RESUMO

Models to study metastatic disease in rare cancers are needed to advance preclinical therapeutics and to gain insight into disease biology, especially for highly aggressive cancers with a propensity for metastatic spread. Osteosarcoma is a rare cancer with a complex genomic landscape in which outcomes for patients with metastatic disease are poor. As osteosarcoma genomes are highly heterogeneous, a large panel of models is needed to fully elucidate key aspects of disease biology and to recapitulate clinically-relevant phenotypes. We describe the development and characterization of osteosarcoma patient-derived xenografts (PDXs) and a panel of PDX-derived cell lines. Matched patient samples, PDXs, and PDX-derived cell lines were comprehensively evaluated using whole genome sequencing and RNA sequencing. PDXs and PDX-derived cell lines largely maintained the expression profiles of the patient from which they were derived despite the emergence of whole-genome duplication (WGD) in a subset of cell lines. These cell line models were heterogeneous in their metastatic capacity and their tissue tropism as observed in both intravenous and orthotopic models. As proof-of-concept study, we used one of these models to test the preclinical effectiveness of a CDK inhibitor on the growth of metastatic tumors in an orthotopic amputation model. Single-agent dinaciclib was effective at dramatically reducing the metastatic burden in this model.

6.
Cancer Cell ; 41(4): 660-677.e7, 2023 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-37001527

RESUMO

Pediatric solid and central nervous system tumors are the leading cause of cancer-related death among children. Identifying new targeted therapies necessitates the use of pediatric cancer models that faithfully recapitulate the patient's disease. However, the generation and characterization of pediatric cancer models has significantly lagged behind adult cancers, underscoring the urgent need to develop pediatric-focused cell line resources. Herein, we establish a single-site collection of 261 cell lines, including 224 pediatric cell lines representing 18 distinct extracranial and brain childhood tumor types. We subjected 182 cell lines to multi-omics analyses (DNA sequencing, RNA sequencing, DNA methylation), and in parallel performed pharmacological and genetic CRISPR-Cas9 loss-of-function screens to identify pediatric-specific treatment opportunities and biomarkers. Our work provides insight into specific pathway vulnerabilities in molecularly defined pediatric tumor classes and uncovers biomarker-linked therapeutic opportunities of clinical relevance. Cell line data and resources are provided in an open access portal.


Assuntos
Neoplasias Encefálicas , Criança , Humanos , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral
7.
Adv Healthc Mater ; 11(17): e2200768, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35767377

RESUMO

Osteosarcoma (OS) is an aggressive bone cancer for which survival has not improved over three decades. While biomaterials have been widely used to engineer 3D soft-tissue tumor models, the potential of engineering 3D biomaterials-based OS models for comprehensive interrogation of OS pathology and drug discovery remains untapped. Bone is characterized by high mineral content, yet the role of bone mineral in OS progression and drug response remains unknown. Here, a microribbon-based OS model with bone-mimicking compositions is developed to elucidate the role of 3D culture and hydroxyapatite in OS signaling and drug response. The results reveal that hydroxyapatite in 3D is critical to support retention of OS signaling and drug resistance similar to patient tissues and mouse orthotopic tumors. The physiological relevance of this 3D model is validated using four established OS cell lines, seven patient-derived xenograft (PDX) cell lines and two animal models. Integrating 3D OS PDX models with RNA-sequencing identified 3D-specific druggable target, which predicts drug response in mouse orthotopic model. These results establish microribbon-based 3D OS models as a novel experimental tool to enable discovery of novel therapeutics that would be otherwise missed with 2D model and may serve as platforms to study patient-specific OS heterogeneity and drug resistance mechanisms.


Assuntos
Neoplasias Ósseas , Osteossarcoma , Animais , Materiais Biocompatíveis , Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/patologia , Linhagem Celular Tumoral , Sinais (Psicologia) , Descoberta de Drogas , Humanos , Hidroxiapatitas , Camundongos , Minerais , Osteossarcoma/patologia
8.
J Clin Invest ; 130(4): 1879-1895, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-31874105

RESUMO

Few therapies are currently available for patients with KRAS-driven cancers, highlighting the need to identify new molecular targets that modulate central downstream effector pathways. Here we found that the microRNA (miRNA) cluster including miR181ab1 is a key modulator of KRAS-driven oncogenesis. Ablation of Mir181ab1 in genetically engineered mouse models of Kras-driven lung and pancreatic cancer was deleterious to tumor initiation and progression. Expression of both resident miRNAs in the Mir181ab1 cluster, miR181a1 and miR181b1, was necessary to rescue the Mir181ab1-loss phenotype, underscoring their nonredundant role. In human cancer cells, depletion of miR181ab1 impaired proliferation and 3D growth, whereas overexpression provided a proliferative advantage. Lastly, we unveiled miR181ab1-regulated genes responsible for this phenotype. These studies identified what we believe to be a previously unknown role for miR181ab1 as a potential therapeutic target in 2 highly aggressive and difficult to treat KRAS-mutated cancers.


Assuntos
Carcinogênese/metabolismo , Neoplasias Pulmonares/metabolismo , MicroRNAs/metabolismo , Família Multigênica , Neoplasias Experimentais/metabolismo , Neoplasias Pancreáticas/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , RNA Neoplásico/metabolismo , Animais , Carcinogênese/genética , Linhagem Celular Tumoral , Proliferação de Células , Humanos , Neoplasias Pulmonares/genética , Camundongos , Camundongos Knockout , MicroRNAs/genética , Neoplasias Experimentais/genética , Neoplasias Pancreáticas/genética , Proteínas Proto-Oncogênicas p21(ras)/genética , RNA Neoplásico/genética
9.
Nat Med ; 25(11): 1783-1795, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31700175

RESUMO

Proinflammatory cytokines in the tumor microenvironment can promote tumor growth, yet their value as therapeutic targets remains underexploited. We validated the functional significance of the cardiotrophin-like cytokine factor 1 (CLCF1)-ciliary neurotrophic factor receptor (CNTFR) signaling axis in lung adenocarcinoma (LUAD) and generated a high-affinity soluble receptor (eCNTFR-Fc) that sequesters CLCF1, thereby inhibiting its oncogenic effects. eCNTFR-Fc inhibits tumor growth in multiple xenograft models and in an autochthonous, highly aggressive genetically engineered mouse model of LUAD, driven by activation of oncogenic Kras and loss of Trp53. Abrogation of CLCF1 through eCNTFR-Fc appears most effective in tumors driven by oncogenic KRAS. We observed a correlation between the effectiveness of eCNTFR-Fc and the presence of KRAS mutations that retain the intrinsic capacity to hydrolyze guanosine triphosphate, suggesting that the mechanism of action may be related to altered guanosine triphosphate loading. Overall, we nominate blockade of CLCF1-CNTFR signaling as a novel therapeutic opportunity for LUAD and potentially for other tumor types in which CLCF1 is present in the tumor microenvironment.


Assuntos
Adenocarcinoma de Pulmão/tratamento farmacológico , Proliferação de Células/genética , Subunidade alfa do Receptor do Fator Neutrófico Ciliar/genética , Citocinas/genética , Adenocarcinoma de Pulmão/genética , Adenocarcinoma de Pulmão/patologia , Animais , Linhagem Celular Tumoral , Subunidade alfa do Receptor do Fator Neutrófico Ciliar/química , Citocinas/química , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Interleucinas/genética , Camundongos , Mutação/genética , Ligação Proteica , Proteínas Proto-Oncogênicas p21(ras)/genética , Transdução de Sinais/efeitos dos fármacos , Microambiente Tumoral/efeitos dos fármacos , Proteína Supressora de Tumor p53/genética , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Cancer Discov ; 9(1): 46-63, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30266815

RESUMO

Osteosarcoma is a highly aggressive cancer for which treatment has remained essentially unchanged for more than 30 years. Osteosarcoma is characterized by widespread and recurrent somatic copy-number alterations (SCNA) and structural rearrangements. In contrast, few recurrent point mutations in protein-coding genes have been identified, suggesting that genes within SCNAs are key oncogenic drivers in this disease. SCNAs and structural rearrangements are highly heterogeneous across osteosarcoma cases, suggesting the need for a genome-informed approach to targeted therapy. To identify patient-specific candidate drivers, we used a simple heuristic based on degree and rank order of copy-number amplification (identified by whole-genome sequencing) and changes in gene expression as identified by RNA sequencing. Using patient-derived tumor xenografts, we demonstrate that targeting of patient-specific SCNAs leads to significant decrease in tumor burden, providing a road map for genome-informed treatment of osteosarcoma. SIGNIFICANCE: Osteosarcoma is treated with a chemotherapy regimen established 30 years ago. Although osteosarcoma is genomically complex, we hypothesized that tumor-specific dependencies could be identified within SCNAs. Using patient-derived tumor xenografts, we found a high degree of response for "genome-matched" therapies, demonstrating the utility of a targeted genome-informed approach.This article is highlighted in the In This Issue feature, p. 1.


Assuntos
Neoplasias Ósseas/terapia , Variação Estrutural do Genoma , Terapia de Alvo Molecular , Osteossarcoma/terapia , Animais , Neoplasias Ósseas/genética , Variações do Número de Cópias de DNA , Genômica , Humanos , Camundongos , Osteossarcoma/genética , Análise de Sequência de RNA , Sequenciamento Completo do Genoma , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Biochim Biophys Acta ; 1767(9): 1157-63, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17706937

RESUMO

The NADH:ubiquinone oxidoreductase or complex I of the mitochondrial respiratory chain is an intricate enzyme with a vital role in energy metabolism. Mutations affecting complex I can affect at least three processes; they can impair the oxidation of NADH, reduce the enzyme's ability to pump protons for the generation of a mitochondrial membrane potential and increase the production of damaging reactive oxygen species. We have previously developed a nematode model of complex I-associated mitochondrial dysfunction that features hallmark characteristics of mitochondrial disease, such as lactic acidosis and decreased respiration. We have expressed the Saccharomyces cerevisiae NDI1 gene, which encodes a single subunit NADH dehydrogenase, in a strain of Caenorhabditis elegans with an impaired complex I. Expression of Ndi1p produces marked improvements in animal fitness and reproduction, increases respiration rates and restores mitochondrial membrane potential to wild type levels. Ndi1p functionally integrates into the nematode respiratory chain and mitigates the deleterious effects of a complex I deficit. However, we have also shown that Ndi1p cannot substitute for the absence of complex I. Nevertheless, the yeast Ndi1p should be considered as a candidate for gene therapy in human diseases involving complex I.


Assuntos
Potencial da Membrana Mitocondrial , Doenças Mitocondriais/metabolismo , NADH Desidrogenase/fisiologia , Proteínas de Saccharomyces cerevisiae/fisiologia , Animais , Animais Geneticamente Modificados , Caenorhabditis elegans , Complexo I de Transporte de Elétrons , Regulação da Expressão Gênica , Potenciais da Membrana , Modelos Biológicos , NADH Desidrogenase/química , Estresse Oxidativo , Consumo de Oxigênio , Fenótipo , Plasmídeos/metabolismo , Proteínas de Saccharomyces cerevisiae/química
13.
Methods Mol Biol ; 372: 51-66, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-18314717

RESUMO

Mitochondria are essential organelles with central roles in diverse cellular processes such as apoptosis, energy production via oxidative phosphorylation, ion homeostasis, and the synthesis of heme, lipid, amino acids, and iron-sulfur clusters. Defects in the mitochondrial respiratory chain lead to or are associated with a wide variety of diseases in humans. The nematode Caenorhabditis elegans provides a powerful genetic and developmental model system for reproducing deleterious mutations causing mitochondrial dysfunction and investigating their metabolic consequences and their mechanisms of pathology. In this chapter, we describe the growth of C. elegans in liquid culture, the isolation of crude and purified mitochondria, and polarographic and histochemical approaches for measuring mitochondrial respiratory chain function.


Assuntos
Caenorhabditis elegans/metabolismo , Fracionamento Celular/métodos , Mitocôndrias/metabolismo , Animais , Bactérias/efeitos dos fármacos , Bactérias/crescimento & desenvolvimento , Caenorhabditis elegans/citologia , Caenorhabditis elegans/efeitos dos fármacos , Caenorhabditis elegans/crescimento & desenvolvimento , Proteínas de Caenorhabditis elegans/metabolismo , Respiração Celular/efeitos dos fármacos , Histocitoquímica , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/enzimologia , NADP/metabolismo , Permeabilidade/efeitos dos fármacos , Polarografia , Prostaglandina-Endoperóxido Sintases/metabolismo , Rotenona/farmacologia , Succinato Desidrogenase/metabolismo , Sacarose , Fixação de Tecidos
14.
Nat Commun ; 8: 14294, 2017 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-28220783

RESUMO

KRAS mutated tumours represent a large fraction of human cancers, but the vast majority remains refractory to current clinical therapies. Thus, a deeper understanding of the molecular mechanisms triggered by KRAS oncogene may yield alternative therapeutic strategies. Here we report the identification of a common transcriptional signature across mutant KRAS cancers of distinct tissue origin that includes the transcription factor FOSL1. High FOSL1 expression identifies mutant KRAS lung and pancreatic cancer patients with the worst survival outcome. Furthermore, FOSL1 genetic inhibition is detrimental to both KRAS-driven tumour types. Mechanistically, FOSL1 links the KRAS oncogene to components of the mitotic machinery, a pathway previously postulated to function orthogonally to oncogenic KRAS. FOSL1 targets include AURKA, whose inhibition impairs viability of mutant KRAS cells. Lastly, combination of AURKA and MEK inhibitors induces a deleterious effect on mutant KRAS cells. Our findings unveil KRAS downstream effectors that provide opportunities to treat KRAS-driven cancers.


Assuntos
Neoplasias Pulmonares/metabolismo , Neoplasias Pancreáticas/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células/genética , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Humanos , Estimativa de Kaplan-Meier , Neoplasias Pulmonares/genética , Camundongos Endogâmicos BALB C , Camundongos Knockout , Camundongos Nus , Mutação , Oncogenes/genética , Neoplasias Pancreáticas/genética , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas p21(ras)/genética , Interferência de RNA , Terapêutica com RNAi/métodos , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
15.
Nat Med ; 23(3): 291-300, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28191885

RESUMO

Lung cancer is the leading cause of cancer deaths worldwide, with the majority of mortality resulting from metastatic spread. However, the molecular mechanism by which cancer cells acquire the ability to disseminate from primary tumors, seed distant organs, and grow into tissue-destructive metastases remains incompletely understood. We combined tumor barcoding in a mouse model of human lung adenocarcinoma with unbiased genomic approaches to identify a transcriptional program that confers metastatic ability and predicts patient survival. Small-scale in vivo screening identified several genes, including Cd109, that encode novel pro-metastatic factors. We uncovered signaling mediated by Janus kinases (Jaks) and the transcription factor Stat3 as a critical, pharmacologically targetable effector of CD109-driven lung cancer metastasis. In summary, by coupling the systematic genomic analysis of purified cancer cells in distinct malignant states from mouse models with extensive human validation, we uncovered several key regulators of metastatic ability, including an actionable pro-metastatic CD109-Jak-Stat3 axis.


Assuntos
Adenocarcinoma/genética , Antígenos CD/genética , Regulação Neoplásica da Expressão Gênica/genética , Janus Quinases/genética , Neoplasias Pulmonares/genética , Proteínas de Neoplasias/genética , Fator de Transcrição STAT3/genética , Adenocarcinoma/metabolismo , Animais , Western Blotting , Linhagem Celular Tumoral , Modelos Animais de Doenças , Técnicas de Silenciamento de Genes , Janus Quinase 1/genética , Janus Quinase 3/genética , Neoplasias Pulmonares/metabolismo , Camundongos , Terapia de Alvo Molecular , Metástase Neoplásica/genética , Reação em Cadeia da Polimerase , Inibidores de Proteínas Quinases , Proteínas Proto-Oncogênicas p21(ras)/genética , Transdução de Sinais , Proteína Supressora de Tumor p53/genética
16.
Nat Med ; 21(10): 1163-71, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26390243

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal human cancers and shows resistance to any therapeutic strategy used. Here we tested small-molecule inhibitors targeting chromatin regulators as possible therapeutic agents in PDAC. We show that JQ1, an inhibitor of the bromodomain and extraterminal (BET) family of proteins, suppresses PDAC development in mice by inhibiting both MYC activity and inflammatory signals. The histone deacetylase (HDAC) inhibitor SAHA synergizes with JQ1 to augment cell death and more potently suppress advanced PDAC. Finally, using a CRISPR-Cas9-based method for gene editing directly in the mouse adult pancreas, we show that de-repression of p57 (also known as KIP2 or CDKN1C) upon combined BET and HDAC inhibition is required for the induction of combination therapy-induced cell death in PDAC. SAHA is approved for human use, and molecules similar to JQ1 are being tested in clinical trials. Thus, these studies identify a promising epigenetic-based therapeutic strategy that may be rapidly implemented in fatal human tumors.


Assuntos
Adenocarcinoma/tratamento farmacológico , Carcinoma Ductal Pancreático/tratamento farmacológico , Epigênese Genética , Inibidores de Histona Desacetilases/uso terapêutico , Proteínas/antagonistas & inibidores , Adenocarcinoma/terapia , Animais , Carcinoma Ductal Pancreático/terapia , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Humanos , Camundongos
17.
Cancer Cell ; 24(1): 59-74, 2013 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-23845442

RESUMO

Sustained tumor progression has been attributed to a distinct population of tumor-propagating cells (TPCs). To identify TPCs relevant to lung cancer pathogenesis, we investigated functional heterogeneity in tumor cells isolated from Kras-driven mouse models of non-small-cell lung cancer (NSCLC). CD24(+)ITGB4(+)Notch(hi) cells are capable of propagating tumor growth in both a clonogenic and an orthotopic serial transplantation assay. While all four Notch receptors mark TPCs, Notch3 plays a nonredundant role in tumor cell propagation in two mouse models and in human NSCLC. The TPC population is enriched after chemotherapy, and the gene signature of mouse TPCs correlates with poor prognosis in human NSCLC. The role of Notch3 in tumor propagation may provide a therapeutic target for NSCLC.


Assuntos
Antígeno CD24/análise , Carcinoma Pulmonar de Células não Pequenas/etiologia , Integrina beta4/análise , Neoplasias Pulmonares/etiologia , Receptores Notch/fisiologia , Animais , Carcinoma Pulmonar de Células não Pequenas/patologia , Humanos , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Endogâmicos C57BL , Receptor Notch3 , Esferoides Celulares
18.
Cancer Res ; 72(22): 5744-56, 2012 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-22962265

RESUMO

Cancer-associated fibroblasts (CAF) have been reported to support tumor progression by a variety of mechanisms. However, their role in the progression of non-small cell lung cancer (NSCLC) remains poorly defined. In addition, the extent to which specific proteins secreted by CAFs contribute directly to tumor growth is unclear. To study the role of CAFs in NSCLCs, a cross-species functional characterization of mouse and human lung CAFs was conducted. CAFs supported the growth of lung cancer cells in vivo by secretion of soluble factors that directly stimulate the growth of tumor cells. Gene expression analysis comparing normal mouse lung fibroblasts and mouse lung CAFs identified multiple genes that correlate with the CAF phenotype. A gene signature of secreted genes upregulated in CAFs was an independent marker of poor survival in patients with NSCLC. This secreted gene signature was upregulated in normal lung fibroblasts after long-term exposure to tumor cells, showing that lung fibroblasts are "educated" by tumor cells to acquire a CAF-like phenotype. Functional studies identified important roles for CLCF1-CNTFR and interleukin (IL)-6-IL-6R signaling in promoting growth of NSCLCs. This study identifies novel soluble factors contributing to the CAF protumorigenic phenotype in NSCLCs and suggests new avenues for the development of therapeutic strategies.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/imunologia , Carcinoma Pulmonar de Células não Pequenas/patologia , Fibroblastos/imunologia , Fibroblastos/patologia , Interleucina-6/imunologia , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/patologia , Adenocarcinoma/imunologia , Adenocarcinoma/patologia , Animais , Processos de Crescimento Celular/fisiologia , Linhagem Celular Tumoral , Humanos , Camundongos , Especificidade da Espécie , Células Estromais/imunologia , Células Estromais/patologia , Transplante Heterólogo
19.
J Clin Invest ; 120(11): 3940-52, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20972333

RESUMO

KRAS is one of the most frequently mutated human oncogenes. In some settings, oncogenic KRAS can trigger cellular senescence, whereas in others it produces hyperproliferation. Elucidating the mechanisms regulating these 2 drastically distinct outcomes would help identify novel therapeutic approaches in RAS-driven cancers. Using a combination of functional genomics and mouse genetics, we identified a role for the transcription factor Wilms tumor 1 (WT1) as a critical regulator of senescence and proliferation downstream of oncogenic KRAS signaling. Deletion or suppression of Wt1 led to senescence of mouse primary cells expressing physiological levels of oncogenic Kras but had no effect on wild-type cells, and Wt1 loss decreased tumor burden in a mouse model of Kras-driven lung cancer. In human lung cancer cell lines dependent on oncogenic KRAS, WT1 loss decreased proliferation and induced senescence. Furthermore, WT1 inactivation defined a gene expression signature that was prognostic of survival only in lung cancer patients exhibiting evidence of oncogenic KRAS activation. These findings reveal an unexpected role for WT1 as a key regulator of the genetic network of oncogenic KRAS and provide important insight into the mechanisms that regulate proliferation or senescence in response to oncogenic signals.


Assuntos
Transformação Celular Neoplásica , Neoplasias Pulmonares , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas WT1/metabolismo , Proteínas ras/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células , Senescência Celular/genética , Fibroblastos/citologia , Fibroblastos/fisiologia , Perfilação da Expressão Gênica , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Camundongos , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas p21(ras)/genética , Interferência de RNA , Transdução de Sinais/fisiologia , Proteínas WT1/genética , Proteínas ras/genética
20.
Proc Natl Acad Sci U S A ; 102(51): 18367-72, 2005 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-16344465

RESUMO

Mitochondrial dysfunction, with an estimated incidence of 1 in 5,000 births, is associated with a wide variety of multisystem degenerative diseases. Among the most prevalent forms of dysfunction are defects in the NADH:ubiquinone oxidoreductase (complex I). Caenorhabditis elegans strains with complex I mutations exhibit characteristic features of human mitochondrial disease including decreased rates of respiration and lactic acidosis. We hypothesized that introducing an additional pathway for the direct oxidation of lactate would be beneficial for energy metabolism. The yeast CYB2 gene encodes an L-lactate:cytochrome c oxidoreductase that oxidizes lactate, donates electrons directly into the mitochondrial respiratory chain, and supports lactate-dependent respiration. Cyb2p expression markedly increases lifespan, fertility, respiration rates, and ATP content in complex I-deficient animals. Our results indicate that metabolic imbalance leading to lactic acidosis and energy depletion are central mechanisms of pathogenesis in mitochondrial dysfunction and that introduction of an additional pathway for lactate oxidation should be considered as a treatment.


Assuntos
Acidose Láctica/metabolismo , Caenorhabditis elegans/metabolismo , Ácido Láctico/metabolismo , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Animais , Animais Geneticamente Modificados , Caenorhabditis elegans/citologia , Caenorhabditis elegans/genética , Metabolismo Energético , Regulação da Expressão Gênica , NADH Desidrogenase/genética , NADH Desidrogenase/metabolismo , Oxirredução , Estresse Oxidativo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA