Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 171(6): 1301-1315.e14, 2017 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-29195074

RESUMO

The two oncogenes KRas and Myc cooperate to drive tumorigenesis, but the mechanism underlying this remains unclear. In a mouse lung model of KRasG12D-driven adenomas, we find that co-activation of Myc drives the immediate transition to highly proliferative and invasive adenocarcinomas marked by highly inflammatory, angiogenic, and immune-suppressed stroma. We identify epithelial-derived signaling molecules CCL9 and IL-23 as the principal instructing signals for stromal reprogramming. CCL9 mediates recruitment of macrophages, angiogenesis, and PD-L1-dependent expulsion of T and B cells. IL-23 orchestrates exclusion of adaptive T and B cells and innate immune NK cells. Co-blockade of both CCL9 and IL-23 abrogates Myc-induced tumor progression. Subsequent deactivation of Myc in established adenocarcinomas triggers immediate reversal of all stromal changes and tumor regression, which are independent of CD4+CD8+ T cells but substantially dependent on returning NK cells. We show that Myc extensively programs an immune suppressive stroma that is obligatory for tumor progression.


Assuntos
Adenocarcinoma/imunologia , Adenoma/imunologia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/imunologia , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Adenoma/genética , Adenoma/patologia , Animais , Carcinogênese , Quimiocinas CC/imunologia , Modelos Animais de Doenças , Feminino , Inflamação/imunologia , Inflamação/metabolismo , Interleucina-23/imunologia , Neoplasias Pulmonares/patologia , Proteínas Inflamatórias de Macrófagos/imunologia , Macrófagos/imunologia , Masculino , Camundongos , Microambiente Tumoral
2.
Nature ; 616(7957): 553-562, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-37055640

RESUMO

Circulating tumour DNA (ctDNA) can be used to detect and profile residual tumour cells persisting after curative intent therapy1. The study of large patient cohorts incorporating longitudinal plasma sampling and extended follow-up is required to determine the role of ctDNA as a phylogenetic biomarker of relapse in early-stage non-small-cell lung cancer (NSCLC). Here we developed ctDNA methods tracking a median of 200 mutations identified in resected NSCLC tissue across 1,069 plasma samples collected from 197 patients enrolled in the TRACERx study2. A lack of preoperative ctDNA detection distinguished biologically indolent lung adenocarcinoma with good clinical outcome. Postoperative plasma analyses were interpreted within the context of standard-of-care radiological surveillance and administration of cytotoxic adjuvant therapy. Landmark analyses of plasma samples collected within 120 days after surgery revealed ctDNA detection in 25% of patients, including 49% of all patients who experienced clinical relapse; 3 to 6 monthly ctDNA surveillance identified impending disease relapse in an additional 20% of landmark-negative patients. We developed a bioinformatic tool (ECLIPSE) for non-invasive tracking of subclonal architecture at low ctDNA levels. ECLIPSE identified patients with polyclonal metastatic dissemination, which was associated with a poor clinical outcome. By measuring subclone cancer cell fractions in preoperative plasma, we found that subclones seeding future metastases were significantly more expanded compared with non-metastatic subclones. Our findings will support (neo)adjuvant trial advances and provide insights into the process of metastatic dissemination using low-ctDNA-level liquid biopsy.


Assuntos
Biomarcadores Tumorais , Carcinoma Pulmonar de Células não Pequenas , DNA Tumoral Circulante , Neoplasias Pulmonares , Mutação , Metástase Neoplásica , Carcinoma de Pequenas Células do Pulmão , Humanos , Biomarcadores Tumorais/sangue , Biomarcadores Tumorais/genética , Carcinoma Pulmonar de Células não Pequenas/sangue , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , DNA Tumoral Circulante/sangue , DNA Tumoral Circulante/genética , Estudos de Coortes , Neoplasias Pulmonares/sangue , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Metástase Neoplásica/diagnóstico , Metástase Neoplásica/genética , Metástase Neoplásica/patologia , Recidiva Local de Neoplasia/diagnóstico , Recidiva Local de Neoplasia/genética , Recidiva Local de Neoplasia/patologia , Filogenia , Carcinoma de Pequenas Células do Pulmão/patologia , Biópsia Líquida
3.
Cell ; 151(1): 11-3, 2012 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-23021211

RESUMO

Myc is an enigma wrapped in a mystery. Attempts to identify Myc target genes, particularly in cancer, have been fraught with dead ends and context-specific functions. Lin et al. and Nie et al. address this conundrum by showing that Myc acts to amplify the output of existing transcriptionally active genes.

4.
Hepatology ; 73(3): 1028-1044, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32460431

RESUMO

BACKGROUND AND AIMS: Hepatocytes undergo profound metabolic rewiring when primed to proliferate during compensatory regeneration and in hepatocellular carcinoma (HCC). However, the metabolic control of these processes is not fully understood. In order to capture the metabolic signature of proliferating hepatocytes, we applied state-of-the-art systems biology approaches to models of liver regeneration, pharmacologically and genetically activated cell proliferation, and HCC. APPROACH AND RESULTS: Integrating metabolomics, lipidomics, and transcriptomics, we link changes in the lipidome of proliferating hepatocytes to altered metabolic pathways including lipogenesis, fatty acid desaturation, and generation of phosphatidylcholine (PC). We confirm this altered lipid signature in human HCC and show a positive correlation of monounsaturated PC with hallmarks of cell proliferation and hepatic carcinogenesis. CONCLUSIONS: Overall, we demonstrate that specific lipid metabolic pathways are coherently altered when hepatocytes switch to proliferation. These represent a source of targets for the development of therapeutic strategies and prognostic biomarkers of HCC.


Assuntos
Carcinoma Hepatocelular/metabolismo , Proliferação de Células , Hepatócitos/metabolismo , Metabolismo dos Lipídeos , Neoplasias Hepáticas/metabolismo , Animais , Perfilação da Expressão Gênica , Hepatócitos/fisiologia , Humanos , Lipidômica , Lipogênese , Masculino , Redes e Vias Metabólicas , Metabolômica , Camundongos , Camundongos Endogâmicos C57BL
5.
Proc Natl Acad Sci U S A ; 116(44): 22399-22408, 2019 10 29.
Artigo em Inglês | MEDLINE | ID: mdl-31611367

RESUMO

Cells with higher levels of Myc proliferate more rapidly and supercompetitively eliminate neighboring cells. Nonetheless, tumor cells in aggressive breast cancers typically exhibit significant and stable heterogeneity in their Myc levels, which correlates with refractoriness to therapy and poor prognosis. This suggests that Myc heterogeneity confers some selective advantage on breast tumor growth and progression. To investigate this, we created a traceable MMTV-Wnt1-driven in vivo chimeric mammary tumor model comprising an admixture of low-Myc- and reversibly switchable high-Myc-expressing clones. We show that such tumors exhibit interclonal mutualism wherein cells with high-Myc expression facilitate tumor growth by promoting protumorigenic stroma yet concomitantly suppress Wnt expression, which renders them dependent for survival on paracrine Wnt provided by low-Myc-expressing clones. To identify any therapeutic vulnerabilities arising from such interdependency, we modeled Myc/Ras/p53/Wnt signaling cross talk as an executable network for low-Myc, for high-Myc clones, and for the 2 together. This executable mechanistic model replicated the observed interdependence of high-Myc and low-Myc clones and predicted a pharmacological vulnerability to coinhibition of COX2 and MEK. This was confirmed experimentally. Our study illustrates the power of executable models in elucidating mechanisms driving tumor heterogeneity and offers an innovative strategy for identifying combination therapies tailored to the oligoclonal landscape of heterogenous tumors.


Assuntos
Heterogeneidade Genética , Neoplasias Mamárias Experimentais/genética , Modelos Teóricos , Proteínas Proto-Oncogênicas c-myc/genética , Animais , Resistencia a Medicamentos Antineoplásicos , Feminino , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/metabolismo , Camundongos , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Via de Sinalização Wnt , Proteínas ras/genética , Proteínas ras/metabolismo
6.
Genes Dev ; 27(5): 504-13, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23475959

RESUMO

The principal reason for failure of targeted cancer therapies is the emergence of resistant clones that regenerate the tumor. Therapeutic efficacy therefore depends on not only how effectively a drug inhibits its target, but also the innate or adaptive functional redundancy of that target and its attendant pathway. In this regard, the Myc transcription factors are intriguing therapeutic targets because they serve the unique and irreplaceable role of coordinating expression of the many diverse genes that, together, are required for somatic cell proliferation. Furthermore, Myc expression is deregulated in most-perhaps all-cancers, underscoring its irreplaceable role in proliferation. We previously showed in a preclinical mouse model of non-small-cell lung cancer that systemic Myc inhibition using the dominant-negative Myc mutant Omomyc exerts a dramatic therapeutic impact, triggering rapid regression of tumors with only mild and fully reversible side effects. Using protracted episodic expression of Omomyc, we now demonstrate that metronomic Myc inhibition not only contains Ras-driven lung tumors indefinitely, but also leads to their progressive eradication. Hence, Myc does indeed serve a unique and nondegenerate role in lung tumor maintenance that cannot be complemented by any adaptive mechanism, even in the most aggressive p53-deficient tumors. These data endorse Myc as a compelling cancer drug target.


Assuntos
Neoplasias Pulmonares/metabolismo , Fragmentos de Peptídeos/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Animais , Animais Geneticamente Modificados , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Camundongos , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/farmacologia , Fragmentos de Peptídeos/uso terapêutico , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/farmacologia , Proteínas Proto-Oncogênicas c-myc/uso terapêutico , Análise de Sobrevida , Proteína Supressora de Tumor p53/metabolismo
7.
J Lipid Res ; 61(11): 1390-1399, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32753459

RESUMO

Nonsmall cell lung cancer (NSCLC) is a leading cause of cancer-related deaths. While mutations in Kras and overexpression of Myc are commonly found in patients, the role of altered lipid metabolism in lung cancer and its interplay with oncogenic Myc is poorly understood. Here we use a transgenic mouse model of Kras-driven lung adenocarcinoma with reversible activation of Myc combined with surface analysis lipid profiling of lung tumors and transcriptomics to study the effect of Myc activity on cholesterol homeostasis. Our findings reveal that the activation of Myc leads to the accumulation of cholesteryl esters (CEs) stored in lipid droplets. Subsequent Myc deactivation leads to further increases in CEs, in contrast to tumors in which Myc was never activated. Gene expression analysis linked cholesterol transport and storage pathways to Myc activity. Our results suggest that increased Myc activity is associated with increased cholesterol influx, reduced efflux, and accumulation of CE-rich lipid droplets in lung tumors. Targeting cholesterol homeostasis is proposed as a promising avenue to explore for novel treatments of lung cancer, with diagnostic and stratification potential in human NSCLC.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/metabolismo , Colesterol/metabolismo , Neoplasias Pulmonares/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Animais , Transporte Biológico , Carcinoma Pulmonar de Células não Pequenas/patologia , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Transgênicos
8.
Genes Dev ; 25(9): 907-16, 2011 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-21478273

RESUMO

The ubiquitous deregulation of Myc in human cancers makes it an intriguing therapeutic target, a notion supported by recent studies in Ras-driven lung tumors showing that inhibiting endogenous Myc triggers ubiquitous tumor regression. However, neither the therapeutic mechanism nor the applicability of Myc inhibition to other tumor types driven by other oncogenic mechanisms is established. Here, we show that inhibition of endogenous Myc also triggers ubiquitous regression of tumors in a simian virus 40 (SV40)-driven pancreatic islet tumor model. Such regression is presaged by collapse of the tumor microenvironment and involution of tumor vasculature. Hence, in addition to its diverse intracellular roles, endogenous Myc serves an essential and nonredundant role in coupling diverse intracellular oncogenic pathways to the tumor microenvironment, further bolstering its credentials as a pharmacological target.


Assuntos
Proteínas Proto-Oncogênicas c-myc/metabolismo , Microambiente Tumoral/fisiologia , Adenoma de Células das Ilhotas Pancreáticas , Animais , Antineoplásicos/farmacologia , Apoptose/fisiologia , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Doxiciclina/farmacologia , Proteínas Ativadoras de GTPase/metabolismo , Regulação Neoplásica da Expressão Gênica , Camundongos , Tumores Neuroendócrinos/irrigação sanguínea , Tumores Neuroendócrinos/patologia , Tumores Neuroendócrinos/fisiopatologia , Ligação Proteica , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Repressoras/metabolismo , Vírus 40 dos Símios/fisiologia
9.
Genes Dev ; 25(16): 1746-57, 2011 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-21852537

RESUMO

MdmX, also known as Mdm4, is a critical negative regulator of p53, and its overexpression serves to block p53 tumor suppressor function in many cancers. Consequently, inhibiting MdmX has emerged as an attractive approach to restoring p53 function in those cancers that retain functional p53. However, the consequences of acute systemic MdmX inhibition in normal adult tissues remain unknown. To determine directly the effects of systemic MdmX inhibition in normal tissues and in tumors, we crossed mdmX(-/-) mice into the p53ER(TAM) knockin background. In place of wild-type p53, p53ER(TAM) knockin mice express a variant of p53, p53ER(TAM), that is completely dependent on 4-hydroxy-tamoxifen for its activity. MdmX inhibition was then modeled by restoring p53 function in these MdmX-deficient mice. We show that MdmX is continuously required to buffer p53 activity in adult normal tissues and their stem cells. Importantly, the effects of transient p53 restoration in the absence of MdmX are nonlethal and reversible, unlike transient p53 restoration in the absence of Mdm2, which is ineluctably lethal. We also show that the therapeutic impact of restoring p53 in a tumor model is enhanced in the absence of MdmX, affording a significant extension of life span over p53 restoration in the presence of MdmX. Hence, systemic inhibition of MdmX is both a feasible therapeutic strategy for restoring p53 function in tumors that retain wild-type p53 and likely to be significantly safer than inhibition of Mdm2.


Assuntos
Linfoma/genética , Proteínas Proto-Oncogênicas c-mdm2/genética , Proteínas Proto-Oncogênicas/genética , Proteína Supressora de Tumor p53/genética , Ubiquitina-Proteína Ligases/genética , Animais , Antineoplásicos Hormonais/farmacologia , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Medula Óssea/efeitos dos fármacos , Medula Óssea/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Embrião de Mamíferos/citologia , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Expressão Gênica/efeitos dos fármacos , Immunoblotting , Estimativa de Kaplan-Meier , Fígado/efeitos dos fármacos , Fígado/metabolismo , Linfoma/tratamento farmacológico , Linfoma/patologia , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Mutação , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteínas Proto-Oncogênicas c-myc/genética , Tamoxifeno/análogos & derivados , Tamoxifeno/farmacologia , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Ubiquitina-Proteína Ligases/metabolismo
10.
Am J Respir Crit Care Med ; 195(11): 1494-1508, 2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28199128

RESUMO

RATIONALE: Improving the early detection and chemoprevention of lung cancer are key to improving outcomes. The pathobiology of early squamous lung cancer is poorly understood. We have shown that amplification of sex-determining region Y-box 2 (SOX2) is an early and consistent event in the pathogenesis of this disease, but its functional oncogenic potential remains uncertain. We tested the impact of deregulated SOX2 expression in a novel organotypic system that recreates the molecular and microenvironmental context in which squamous carcinogenesis occurs. OBJECTIVES: (1) To develop an in vitro model of bronchial dysplasia that recapitulates key molecular and phenotypic characteristics of the human disease; (2) to test the hypothesis that SOX2 deregulation is a key early event in the pathogenesis of bronchial dysplasia; and (3) to use the model for studies on pathogenesis and chemoprevention. METHODS: We engineered the inducible activation of oncogenes in immortalized bronchial epithelial cells. We used three-dimensional tissue culture to build an organotypic model of bronchial dysplasia. MEASUREMENTS AND MAIN RESULTS: We recapitulated human bronchial dysplasia in vitro. SOX2 deregulation drives dysplasia, and loss of tumor promoter 53 is a cooperating genetic event that potentiates the dysplastic phenotype. Deregulated SOX2 alters critical genes implicated in hallmarks of cancer progression. Targeted inhibition of AKT prevents the initiation of the dysplastic phenotype. CONCLUSIONS: In the appropriate genetic and microenvironmental context, acute deregulation of SOX2 drives bronchial dysplasia. This confirms its oncogenic potential in human cells and affords novel insights into the impact of SOX2 deregulation. This model can be used to test therapeutic agents aimed at chemoprevention.


Assuntos
Displasia Broncopulmonar/genética , Displasia Broncopulmonar/fisiopatologia , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/fisiopatologia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/fisiopatologia , Fatores de Transcrição SOXB1/genética , Técnicas de Cultura de Células , Humanos , Modelos Biológicos
11.
Nature ; 468(7323): 567-71, 2010 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-21107427

RESUMO

Non-small cell lung carcinoma (NSCLC) is the leading cause of cancer-related death worldwide, with an overall 5-year survival rate of only 10-15%. Deregulation of the Ras pathway is a frequent hallmark of NSCLC, often through mutations that directly activate Kras. p53 is also frequently inactivated in NSCLC and, because oncogenic Ras can be a potent trigger of p53 (ref. 3), it seems likely that oncogenic Ras signalling has a major and persistent role in driving the selection against p53. Hence, pharmacological restoration of p53 is an appealing therapeutic strategy for treating this disease. Here we model the probable therapeutic impact of p53 restoration in a spontaneously evolving mouse model of NSCLC initiated by sporadic oncogenic activation of endogenous Kras. Surprisingly, p53 restoration failed to induce significant regression of established tumours, although it did result in a significant decrease in the relative proportion of high-grade tumours. This is due to selective activation of p53 only in the more aggressive tumour cells within each tumour. Such selective activation of p53 correlates with marked upregulation in Ras signal intensity and induction of the oncogenic signalling sensor p19(ARF)( )(ref. 6). Our data indicate that p53-mediated tumour suppression is triggered only when oncogenic Ras signal flux exceeds a critical threshold. Importantly, the failure of low-level oncogenic Kras to engage p53 reveals inherent limits in the capacity of p53 to restrain early tumour evolution and in the efficacy of therapeutic p53 restoration to eradicate cancers.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/fisiopatologia , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/fisiopatologia , Proteína Supressora de Tumor p53/metabolismo , Animais , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Proliferação de Células , Modelos Animais de Doenças , Neoplasias Pulmonares/metabolismo , Camundongos , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Proteína Supressora de Tumor p53/genética , Proteínas ras/metabolismo
12.
Proc Natl Acad Sci U S A ; 110(16): E1480-9, 2013 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-23542378

RESUMO

Based on clinical presentation, glioblastoma (GBM) is stratified into primary and secondary types. The protein 53 (p53) pathway is functionally incapacitated in most GBMs by distinctive type-specific mechanisms. To model human gliomagenesis, we used a GFAP-HRas(V12) mouse model crossed into the p53ER(TAM) background, such that either one or both copies of endogenous p53 is replaced by a conditional p53ER(TAM) allele. The p53ER(TAM) protein can be toggled reversibly in vivo between wild-type and inactive conformations by administration or withdrawal of 4-hydroxytamoxifen (4-OHT), respectively. Surprisingly, gliomas that develop in GFAP-HRas(V12);p53(+/KI) mice abrogate the p53 pathway by mutating p19(ARF)/MDM2 while retaining wild-type p53 allele. Consequently, such tumors are unaffected by restoration of their p53ER(TAM) allele. By contrast, gliomas arising in GFAP-HRas(V12);p53(KI/KI) mice develop in the absence of functional p53. Such tumors retain a functional p19(ARF)/MDM2-signaling pathway, and restoration of p53ER(TAM) allele triggers p53-tumor-suppressor activity. Congruently, growth inhibition upon normalization of mutant p53 by a small molecule, Prima-1, in human GBM cultures also requires p14(ARF)/MDM2 functionality. Notably, the antitumoral efficacy of p53 restoration in tumor-bearing GFAP-HRas(V12);p53(KI/KI) animals depends on the duration and frequency of p53 restoration. Thus, intermittent exposure to p53ER(TAM) activity mitigated the selective pressure to inactivate the p19(ARF)/MDM2/p53 pathway as a means of resistance, extending progression-free survival. Our results suggest that intermittent dosing regimes of drugs that restore wild-type tumor-suppressor function onto mutant, inactive p53 proteins will prove to be more efficacious than traditional chronic dosing by similarly reducing adaptive resistance.


Assuntos
Modelos Animais de Doenças , Glioblastoma/tratamento farmacológico , Glioblastoma/fisiopatologia , Transdução de Sinais/fisiologia , Tamoxifeno/análogos & derivados , Proteína Supressora de Tumor p53/metabolismo , Animais , Sequência de Bases , Linhagem Celular Tumoral , Inibidor p16 de Quinase Dependente de Ciclina/genética , Primers do DNA/genética , Imunofluorescência , Glioblastoma/metabolismo , Técnicas Histológicas , Humanos , Immunoblotting , Estimativa de Kaplan-Meier , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Mutação/genética , Proteínas Proto-Oncogênicas c-mdm2/genética , Análise de Sequência de DNA , Transdução de Sinais/genética , Tamoxifeno/farmacologia , Tamoxifeno/uso terapêutico
13.
Cancer Cell ; 12(4): 355-66, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17936560

RESUMO

It is believed that Mdm2 suppresses p53 in two ways: transcriptional inhibition by direct binding, and degradation via its E3 ligase activity. To study these functions physiologically, we generated mice bearing a single-residue substitution (C462A) abolishing the E3 function without affecting p53 binding. Unexpectedly, homozygous mutant mice died before E7.5, and deletion of p53 rescued the lethality. Furthermore, reintroducing a switchable p53 by crossing with p53ER(TAM) mice surprisingly demonstrated that the mutant Mdm2(C462A) was rapidly degraded in a manner indistinguishable from that of the wild-type Mdm2. Hence, our data indicate that (1) the Mdm2-p53 physical interaction, without Mdm2-mediated p53 ubiquitination, cannot control p53 activity sufficiently to allow early mouse embryonic development, and (2) Mdm2's E3 function is not required for Mdm2 degradation.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Mutagênese Sítio-Dirigida , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Transcrição Gênica , Proteína Supressora de Tumor p53/metabolismo , Substituição de Aminoácidos , Animais , Células Cultivadas , Dano ao DNA , Regulação para Baixo , Embrião de Mamíferos , Fibroblastos/enzimologia , Fibroblastos/metabolismo , Fibroblastos/efeitos da radiação , Raios gama , Regulação da Expressão Gênica no Desenvolvimento/efeitos da radiação , Genótipo , Idade Gestacional , Homozigoto , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação , Fenótipo , Complexo de Endopeptidases do Proteassoma/metabolismo , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-mdm2/química , Proteínas Proto-Oncogênicas c-mdm2/deficiência , Proteínas Proto-Oncogênicas c-mdm2/genética , Transcrição Gênica/efeitos da radiação , Proteína Supressora de Tumor p53/deficiência , Proteína Supressora de Tumor p53/genética
14.
Blood ; 119(2): 411-21, 2012 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-22067385

RESUMO

In response to microenvironmental signals, macrophages undergo different activation, including the "classic" proinflammatory phenotype (also called M1), the "alternative" activation induced by the IL-4/IL-13 trigger, and the related but distinct heterogeneous M2 polarization associated with the anti-inflammatory profile. The latter is induced by several stimuli, including IL-10 and TGF-ß. Macrophage-polarized activation has profound effects on immune and inflammatory responses and in tumor biology, but information on the underlying molecular pathways is scarce. In the present study, we report that alternative polarization of macrophages requires the transcription factor c-MYC. In macrophages, IL-4 and different stimuli sustaining M2-like polarization induce c-MYC expression and its translocation to the nucleus. c-MYC controls the induction of a subset (45%) of genes associated with alternative activation. ChIP assays indicate that c-MYC directly regulates some genes associated with alternative activation, including SCARB1, ALOX15, and MRC1, whereas others, including CD209, are indirectly regulated by c-MYC. c-MYC up-regulates the IL-4 signaling mediators signal transducer and activator of transcription-6 and peroxisome proliferator-activated receptorγ, is also expressed in tumor-associated macrophages, and its inhibition blocks the expression of protumoral genes including VEGF, MMP9, HIF-1α, and TGF-ß. We conclude that c-MYC is a key player in alternative macrophage activation, and is therefore a potential therapeutic target in pathologies related to these cells, including tumors.


Assuntos
Biomarcadores Tumorais/genética , Neoplasias da Mama/metabolismo , Neoplasias do Colo/metabolismo , Regulação Neoplásica da Expressão Gênica , Macrófagos/metabolismo , Neoplasias Pancreáticas/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Biomarcadores Tumorais/metabolismo , Western Blotting , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Diferenciação Celular , Células Cultivadas , Imunoprecipitação da Cromatina , Colo/metabolismo , Colo/patologia , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Feminino , Perfilação da Expressão Gênica , Humanos , Interleucina-4/farmacologia , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/citologia , Análise de Sequência com Séries de Oligonucleotídeos , PPAR gama/genética , PPAR gama/metabolismo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Proteínas Proto-Oncogênicas c-myc/genética , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT6/genética , Fator de Transcrição STAT6/metabolismo , Transdução de Sinais
15.
Nat Med ; 13(10): 1211-8, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17906636

RESUMO

An association between inflammation and cancer has long been recognized, but the cause and effect relationship linking the two remains unclear. Myc is a pleiotropic transcription factor that is overexpressed in many human cancers and instructs many extracellular aspects of the tumor tissue phenotype, including remodeling of tumor stroma and angiogenesis. Here we show in a beta-cell tumor model that activation of Myc in vivo triggers rapid recruitment of mast cells to the tumor site-a recruitment that is absolutely required for macroscopic tumor expansion. In addition, treatment of established beta-cell tumors with a mast cell inhibitor rapidly triggers hypoxia and cell death of tumor and endothelial cells. Inhibitors of mast cell function may therefore prove therapeutically useful in restraining expansion and survival of pancreatic and other cancers.


Assuntos
Transformação Celular Neoplásica/genética , Mastócitos/metabolismo , Neovascularização Patológica/etiologia , Neoplasias Pancreáticas/irrigação sanguínea , Proteínas Proto-Oncogênicas c-myc/fisiologia , Animais , Células da Medula Óssea/citologia , Células Cultivadas , Quimiocina CCL2/metabolismo , Quimiocina CCL5/metabolismo , Fêmur/citologia , Regulação Neoplásica da Expressão Gênica , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neovascularização Patológica/genética , Neovascularização Patológica/fisiopatologia , Análise de Sequência com Séries de Oligonucleotídeos , Neoplasias Pancreáticas/etiologia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/fisiopatologia , Proteínas Proto-Oncogênicas c-myc/genética
16.
Cancer Cell ; 10(5): 345-7, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17097554

RESUMO

One of the most exciting developments in recent cancer treatment has been the move away from crude cytotoxic agents toward drugs that inhibit specific targets in specific cellular pathways. One assumption of this strategy is that maintenance of human cancers is dependent upon a limited cadre of therapeutically tractable oncogenic lesions. In this issue of Cancer Cell, an intriguing paper from Sharma et al. endorses this approach by showing that evolution appears to be working for us. They show that an innate asymmetry in the dynamics of intracellular signaling biases pathway inhibition in favor of cell death. This bias may significantly potentiate targeted cancer therapies.


Assuntos
Neoplasias/genética , Neoplasias/metabolismo , Oncogenes , Transdução de Sinais/fisiologia , Apoptose , Humanos , Mutação , Neoplasias/fisiopatologia
17.
Cancer Cell ; 10(6): 501-14, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17157790

RESUMO

There is currently much interest in the idea of restoring p53 activity in tumor cells by inhibiting Hdm2/Mdm2. However, it has remained unclear whether this would also activate p53 in normal cells. Using a switchable endogenous p53 mouse model, which allows rapid and reversible toggling of p53 status between wild-type and null states, we show that p53 is spontaneously active in all tested tissues of mdm2-deficient mice, triggering fatal pathologies that include ablation of classically radiosensitive tissues. In apoptosis-resistant tissues, spontaneous unbuffered p53 activity triggers profound inhibition of cell proliferation. Such acute spontaneous p53 activity occurs in the absence of any detectable p53 posttranslational modification, DNA damage, or p19ARF signaling and triggers rapid p53 degradation.


Assuntos
Proteínas Proto-Oncogênicas c-mdm2/fisiologia , Proteína Supressora de Tumor p53/fisiologia , Fator 1 de Ribosilação do ADP/fisiologia , Animais , Dano ao DNA , Imidazóis , Camundongos , Fosforilação , Piperazinas , Proteínas Proto-Oncogênicas c-mdm2/deficiência , Tamoxifeno/análogos & derivados , Tamoxifeno/farmacologia , Transcrição Gênica
18.
Cancer Cell ; 10(2): 113-20, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16904610

RESUMO

Overexpression of Bcl-xL, loss of p19 ARF, and loss of p53 all accelerate Myc oncogenesis. All three lesions are implicated in suppressing Myc-induced apoptosis, suggesting that this is a common mechanism by which they synergize with Myc. However, using an acutely switchable model of Myc-induced tumorigenesis, we demonstrate that each lesion cooperates with Myc in vivo by a distinct mechanism. While Bcl-xL blocks Myc-induced apoptosis, inactivation of p19 ARF enhances it. However, this increase in apoptosis is matched by increased Myc-induced proliferation. p53 inactivation shares features of both lesions, partially suppressing apoptosis while augmenting proliferation. Bcl-xL and p19 ARF loss together synergize to further accelerate Myc oncogenesis. Thus, differing lesions cooperate oncogenically with Myc by discrete mechanisms that can themselves synergize with each other.


Assuntos
Apoptose , Transformação Celular Neoplásica , Genes myc/fisiologia , Proteína Supressora de Tumor p14ARF/fisiologia , Proteína bcl-X/fisiologia , Animais , Proliferação de Células , Inibidor p16 de Quinase Dependente de Ciclina , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Células Secretoras de Insulina/patologia , Camundongos , Camundongos Transgênicos , Proteína Supressora de Tumor p53/fisiologia
19.
Nature ; 455(7213): 679-83, 2008 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-18716624

RESUMO

Myc is a pleiotropic basic helix-loop-helix leucine zipper transcription factor that coordinates expression of the diverse intracellular and extracellular programs that together are necessary for growth and expansion of somatic cells. In principle, this makes inhibition of Myc an attractive pharmacological approach for treating diverse types of cancer. However, enthusiasm has been muted by lack of direct evidence that Myc inhibition would be therapeutically efficacious, concerns that it would induce serious side effects by inhibiting proliferation of normal tissues, and practical difficulties in designing Myc inhibitory drugs. We have modelled genetically both the therapeutic impact and the side effects of systemic Myc inhibition in a preclinical mouse model of Ras-induced lung adenocarcinoma by reversible, systemic expression of a dominant-interfering Myc mutant. We show that Myc inhibition triggers rapid regression of incipient and established lung tumours, defining an unexpected role for endogenous Myc function in the maintenance of Ras-dependent tumours in vivo. Systemic Myc inhibition also exerts profound effects on normal regenerating tissues. However, these effects are well tolerated over extended periods and rapidly and completely reversible. Our data demonstrate the feasibility of targeting Myc, a common downstream conduit for many oncogenic signals, as an effective, efficient and tumour-specific cancer therapy.


Assuntos
Terapia Genética , Neoplasias Pulmonares/terapia , Modelos Biológicos , Proteínas Proto-Oncogênicas c-myc/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-myc/genética , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Adenocarcinoma/terapia , Animais , Trato Gastrointestinal/citologia , Trato Gastrointestinal/metabolismo , Trato Gastrointestinal/patologia , Genes Dominantes/genética , Genes ras , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Masculino , Camundongos , Mutação/genética , Proteína Oncogênica p21(ras)/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Pele/citologia , Pele/metabolismo , Pele/patologia , Testículo/citologia , Testículo/metabolismo , Testículo/patologia , Transgenes/genética
20.
Nat Genet ; 37(7): 718-26, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15924142

RESUMO

To investigate the functions of the p53 tumor suppressor, we created a new knock-in gene replacement mouse model in which the endogenous Trp53 gene is substituted by one encoding p53ER(TAM), a p53 fusion protein whose function is completely dependent on ectopic provision of 4-hydroxytamoxifen. We show here that both tissues in vivo and cells in vitro derived from such mice can be rapidly toggled between wild-type and p53 knockout states. Using this rapid perturbation model, we define the kinetics, dependence, persistence and reversibility of p53-mediated responses to DNA damage in tissues in vivo and to activation of the Ras oncoprotein and stress in vitro. This is the first example to our knowledge of a new class of genetic model that allows the specific, rapid and reversible perturbation of the function of a single endogenous gene in vivo.


Assuntos
Neoplasias/genética , Tamoxifeno/análogos & derivados , Proteína Supressora de Tumor p53/fisiologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Células Cultivadas , Dano ao DNA/efeitos dos fármacos , Embrião de Mamíferos/citologia , Fibroblastos/metabolismo , Raios gama , Regulação Neoplásica da Expressão Gênica , Genes p53 , Genes ras/genética , Intestino Delgado/efeitos dos fármacos , Intestino Delgado/patologia , Intestino Delgado/efeitos da radiação , Camundongos , Camundongos Transgênicos , Modelos Animais , Neoplasias/metabolismo , Neoplasias/patologia , Baço/efeitos dos fármacos , Baço/patologia , Baço/efeitos da radiação , Tamoxifeno/farmacologia , Timo/efeitos dos fármacos , Timo/patologia , Timo/efeitos da radiação , Fatores de Tempo , Proteína Supressora de Tumor p53/genética , Irradiação Corporal Total
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa