Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 23(20)2022 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-36293286

RESUMEN

The tyrosine kinase receptor encoded by the MET oncogene has been extensively studied. Surprisingly, one extracellular domain, PSI, evolutionary conserved between plexins, semaphorins, and integrins, has no established function. The MET PSI sequence contains two CXXC motifs, usually found in protein disulfide isomerases (PDI). Using a scrambled oxidized RNAse enzymatic activity assay in vitro, we show, for the first time, that the MET extracellular domain displays disulfide isomerase activity, abolished by PSI domain antibodies. PSI domain deletion or mutations of CXXC sites to AXXA or SXXS result in a significant impairment of the cleavage of the MET 175 kDa precursor protein, abolishing the maturation of α and ß chains, of, respectively, 50 kDa and 145 kDa, disulfide-linked. The uncleaved precursor is stuck in the Golgi apparatus and, interestingly, is constitutively phosphorylated. However, no signal transduction is observed as measured by AKT and MAPK phosphorylation. Consequently, biological responses to the MET ligand-hepatocyte growth factor (HGF)-such as growth and epithelial to mesenchymal transition, are hampered. These data show that the MET PSI domain is functional and is required for the maturation, surface expression, and biological functions of the MET oncogenic protein.


Asunto(s)
Factor de Crecimiento de Hepatocito , Semaforinas , Factor de Crecimiento de Hepatocito/metabolismo , Proteína Disulfuro Isomerasas/genética , Ligandos , Transición Epitelial-Mesenquimal , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-met/genética , Proteínas Proto-Oncogénicas c-met/metabolismo , Semaforinas/genética , Oncogenes , Disulfuros , Integrinas/genética , Ribonucleasas/genética
2.
Lab Invest ; 100(10): 1330-1344, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32404931

RESUMEN

Glioblastomas (GBM) can be classified into three major transcriptional subgroups (proneural, mesenchymal, classical), underlying different molecular alterations, prognosis, and response to therapy. However, transcriptional analysis is not routinely feasible and assessment of a simplified method for glioblastoma subclassification is required. We propose an integrated molecular and immunohistochemical approach aimed at identifying GBM subtypes in routine paraffin-embedded material. RNA-sequencing analysis was performed on representative samples (n = 51) by means of a "glioblastoma transcriptional subtypes (GliTS) redux" custom gene signature including a restricted number (n = 90) of upregulated genes validated on the TCGA dataset. With this dataset, immunohistochemical profiles, based on expression of a restricted panel of gene classifiers, were integrated by a machine-learning approach to generate a GliTS based on protein quantification that allowed an efficient GliTS assignment when applied to an extended cohort (n = 197). GliTS redux maintained high levels of correspondence with the original GliTS classification using the TCGA dataset. The machine-learning approach designed an immunohistochemical (IHC)-based classification, whose concordance was 79.5% with the transcriptional- based classification, and reached 90% for the mesenchymal subgroup. Distribution and survival of GliTS were in line with reported data, with the mesenchymal subgroup given the worst prognosis. Notably, the algorithm allowed the identification of cases with comparable probability to be assigned to different GliTS, thus falling within overlapping regions and reflecting an extreme heterogeneous phenotype that mirrors the underlying genetic and biological tumor heterogeneity. Indeed, while mesenchymal and classical subgroups were well segregated, the proneural types frequently showed a mixed proneural/classical phenotype, predicted as proneural by the algorithm, but with comparable probability of being assigned to the classical subtype. These cases, characterized by concomitant high expression of EGFR and proneural biomarkers, showed lower survival. Collectively, these data indicate that a restricted panel of highly sensitive immunohistochemical markers can efficiently predict GliTS with high accuracy and significant association with different clinical outcomes.


Asunto(s)
Neoplasias Encefálicas/clasificación , Neoplasias Encefálicas/metabolismo , Perfilación de la Expresión Génica , Glioblastoma/clasificación , Glioblastoma/metabolismo , Anciano , Algoritmos , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Neoplasias Encefálicas/genética , Análisis por Conglomerados , Estudios de Cohortes , Femenino , Perfilación de la Expresión Génica/métodos , Perfilación de la Expresión Génica/estadística & datos numéricos , Regulación Neoplásica de la Expresión Génica , Glioblastoma/genética , Humanos , Inmunohistoquímica , Aprendizaje Automático , Masculino , Persona de Mediana Edad , RNA-Seq
3.
Int J Mol Sci ; 21(15)2020 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-32722178

RESUMEN

Previous studies showed that the hepatocyte growth factor (HGF)-Met receptor axis plays long-lasting cardioprotection against doxorubicin anti-cancer therapy. Here, we explored the mechanism(s) underlying the HGF protective effect. DNA damage was monitored by histone H2AX phosphorylation and apoptosis by proteolytic cleavage of caspase 3. In doxorubicin-treated H9c2 cardiomyoblasts, the long-lasting cardioprotection is mediated by activation of the Ras/Raf/Mek/Erk (extracellular signal-regulated kinase 1,2) signaling pathway and requires Stat3 (signal transducer and activator of transcription 3) activation. The HGF protection was abrogated by the Erk1,2 inhibitor, PD98059. This translated into reduced Y705 phosphorylation and impaired nuclear translocation of Stat3, showing crosstalk between Erk1,2 and Stat3 signaling. An array of 29 cytokines, known to activate Stat3, was interrogated to identify the molecule(s) linking the two pathways. The analysis showed a selective increase in expression of the tissue inhibitor of metalloproteinases-1 (Timp1). Consistently, inhibition in cardiomyoblasts of Timp1 translation by siRNAs blunted both Stat3 activation and the cardioprotective effect of HGF. Thus, Timp1 is responsible for the generation of a feed-forward loop of Stat3 activation and helps cardiomyocytes to survive during the genotoxic stress induced by anthracyclines.


Asunto(s)
Doxorrubicina/efectos adversos , Factor de Crecimiento de Hepatocito/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Mioblastos Cardíacos/metabolismo , Factor de Transcripción STAT3/metabolismo , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Animales , Línea Celular , Doxorrubicina/farmacología , Flavonoides/farmacología , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Mioblastos Cardíacos/patología , Ratas , Factores de Tiempo
4.
Stem Cells ; 35(11): 2218-2228, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28895245

RESUMEN

Glioblastoma (GBM) is a lethal tumor that displays remarkable genetic heterogeneity. It is also known that GBM contains a cell hierarchy driven by GBM stem-like cells (GSCs), responsible for tumor generation, therapeutic resistance, and relapse. An important and still open issue is whether phylogenetically related GSCs can be found in matched primary and recurrent GBMs, and reflect tumor genetic evolution under therapeutic pressure. To address this, we analyzed the mutational profile of GSCs isolated from either human primary GBMs (primary GSCs) or their matched tumors recurring after surgery and chemoradiotherapy (recurrent GSCs). We found that recurrent GSCs can accumulate temozolomide-related mutations over primary GSCs, following both linear and branched patterns. In the latter case, primary and recurrent GSCs share a common set of lesions, but also harbor distinctive mutations indicating that primary and recurrent GSCs derive from a putative common ancestor GSC by divergent genetic evolution. Interestingly, TP53 mutations distinctive of recurrent GSCs were detectable at low frequency in the corresponding primary tumors and likely marked pre-existent subclones that evolved under therapeutic pressure and expanded in the relapsing tumor. Consistently, recurrent GSCs displayed in vitro greater therapeutic resistance than primary GSCs. Overall, these data indicate that (a) phylogenetically related GSCs are found in matched primary and recurrent GBMs and (b) recurrent GSCs likely pre-exist in the untreated primary tumor and are both mutagenized and positively selected by chemoradiotherapy. Stem Cells 2017;35:2218-2228.


Asunto(s)
Dosificación de Gen/genética , Glioblastoma/genética , Células Madre Neoplásicas/metabolismo , Adulto , Animales , Evolución Molecular , Femenino , Glioblastoma/patología , Humanos , Masculino , Ratones , Persona de Mediana Edad , Células Madre Neoplásicas/patología
5.
Nat Rev Cancer ; 6(8): 637-45, 2006 08.
Artículo en Inglés | MEDLINE | ID: mdl-16862193

RESUMEN

Metastasis follows the inappropriate activation of a genetic programme termed invasive growth, which is a physiological process that occurs during embryonic development and post-natal organ regeneration. Burgeoning evidence indicates that invasive growth is also executed by stem and progenitor cells, and is usurped by cancer stem cells. The MET proto-oncogene, which is expressed in both stem and cancer cells, is a key regulator of invasive growth. Recent findings indicate that the MET tyrosine-kinase receptor is a sensor of adverse microenvironmental conditions (such as hypoxia) and drives cell invasion and metastasis through the transcriptional activation of a set of genes that control blood coagulation.


Asunto(s)
Invasividad Neoplásica/genética , Neoplasias/genética , Neoplasias/patología , Proteínas Proto-Oncogénicas/fisiología , Receptores de Factores de Crecimiento/fisiología , Células Madre/fisiología , Hipoxia de la Célula/fisiología , Humanos , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas c-met
6.
Proc Natl Acad Sci U S A ; 108(34): 14240-5, 2011 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-21831840

RESUMEN

Recent observations suggest that p53 mutations are responsible not only for growth of primary tumors but also for their dissemination. However, mechanisms involved in p53-mediated control of cell motility and invasion remain poorly understood. By using the primary ovarian surface epithelium cell culture, we show that conditional inactivation of p53 or expression of its mutant forms results in overexpression of MET receptor tyrosine kinase, a crucial regulator of invasive growth. At the same time, cells acquire increased MET-dependent motility and invasion. Wild-type p53 negatively regulates MET expression by two mechanisms: (i) transactivation of MET-targeting miR-34, and (ii) inhibition of SP1 binding to MET promoter. Both mechanisms are not functional in p53 absence, but mutant p53 proteins retain partial MET promoter suppression. Accordingly, MET overexpression, cell motility, and invasion are particularly high in p53-null cells. These results identify MET as a critical effector of p53 and suggest that inhibition of MET may be an effective antimetastatic approach to treat cancers with p53 mutations. These results also show that the extent of advanced cancer traits, such as invasion, may be determined by alterations in individual components of p53/MET regulatory network.


Asunto(s)
Movimiento Celular , Regulación Neoplásica de la Expresión Génica , Proteínas Proto-Oncogénicas c-met/genética , Receptores de Factores de Crecimiento/genética , Proteína p53 Supresora de Tumor/metabolismo , Animales , Línea Celular Tumoral , Epitelio/metabolismo , Epitelio/patología , Femenino , Silenciador del Gen , Humanos , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Proteínas Mutantes/metabolismo , Invasividad Neoplásica , Ovario/metabolismo , Ovario/patología , Regiones Promotoras Genéticas/genética , Unión Proteica , Proteínas Proto-Oncogénicas c-met/metabolismo , Receptores de Factores de Crecimiento/metabolismo , Factor de Transcripción Sp1/metabolismo
7.
STAR Protoc ; 4(4): 102705, 2023 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-37971942

RESUMEN

Cultures enriched in glioblastoma stem-like cells (GSCs) are prominent in vitro models to investigate molecular determinants and therapeutic targets of glioblastoma; however, conventional GSC derivation protocols fail to preserve GSC heterogeneity. Here, we present a protocol for the propagation of heterogeneous GSC cultures starting from cell resuspensions containing the entire tumor mass. We describe steps for isolation of GSCs and their maintenance and expansion in culture. We then detail procedures for preliminary analysis to be performed on freshly isolated material. For complete details on the use and execution of this protocol, please refer to De Bacco et al.1.


Asunto(s)
Glioblastoma , Humanos , Glioblastoma/patología
8.
Mol Oncol ; 17(7): 1280-1301, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-36862005

RESUMEN

In colorectal cancer, the mechanisms underlying tumor aggressiveness require further elucidation. Taking advantage of a large panel of human metastatic colorectal cancer xenografts and matched stem-like cell cultures (m-colospheres), here we show that the overexpression of microRNA 483-3p (miRNA-483-3p; also known as MIR-483-3p), encoded by a frequently amplified gene locus, confers an aggressive phenotype. In m-colospheres, endogenous or ectopic miRNA-483-3p overexpression increased proliferative response, invasiveness, stem cell frequency, and resistance to differentiation. Transcriptomic analyses and functional validation found that miRNA-483-3p directly targets NDRG1, known as a metastasis suppressor involved in EGFR family downregulation. Mechanistically, miRNA-483-3p overexpression induced the signaling pathway triggered by ERBB3, including AKT and GSK3ß, and led to the activation of transcription factors regulating epithelial-mesenchymal transition (EMT). Consistently, treatment with selective anti-ERBB3 antibodies counteracted the invasive growth of miRNA-483-3p-overexpressing m-colospheres. In human colorectal tumors, miRNA-483-3p expression inversely correlated with NDRG1 and directly correlated with EMT transcription factor expression and poor prognosis. These results unveil a previously unrecognized link between miRNA-483-3p, NDRG1, and ERBB3-AKT signaling that can directly support colorectal cancer invasion and is amenable to therapeutic targeting.


Asunto(s)
Neoplasias del Colon , Neoplasias Colorrectales , MicroARNs , Neoplasias del Recto , Humanos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Regulación hacia Abajo/genética , Línea Celular Tumoral , MicroARNs/genética , MicroARNs/metabolismo , Neoplasias Colorrectales/patología , Neoplasias del Colon/genética , Factores de Transcripción/metabolismo , Neoplasias del Recto/genética , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica , Movimiento Celular/genética , Invasividad Neoplásica/genética
9.
Virchows Arch ; 482(3): 463-475, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36346458

RESUMEN

The aim of this study is to envisage a streamlined pathological workup to rule out CUPs in patients presenting with MUOs. Sixty-four MUOs were classified using standard histopathology. Clinical data, immunocytochemical markers, and results of molecular analysis were recorded. MUOs were histologically subdivided in clear-cut carcinomas (40 adenocarcinomas, 11 squamous, and 3 neuroendocrine carcinomas) and unclear-carcinoma features (5 undifferentiated and 5 sarcomatoid tumors). Cytohistology of 7/40 adenocarcinomas suggested an early metastatic cancer per se. In 33/40 adenocarcinomas, CK7/CK20 expression pattern, gender, and metastasis sites influenced tissue-specific marker selection. In 23/40 adenocarcinomas, a "putative-immunophenotype" of tissue of origin addressed clinical-diagnostic examinations, identifying 9 early metastatic cancers. Cell lineage markers were used to confirm squamous and neuroendocrine differentiation. Pan-cytokeratins were used to confirm the epithelial nature of poorly differentiated tumors, followed by tissue and cell lineage markers, which identified one melanoma. In total, 47/64 MUOs (73.4%) were confirmed CUP. Molecular analysis, feasible in 37/47 CUPs (78.7%), had no diagnostic impact. Twenty CUP patients, mainly with squamous carcinomas and adenocarcinomas with putative-gynecologic-immunophenotypes, presented with only lymph node metastases and had longer median time to progression and overall survival (< 0.001), compared with patients with other metastatic patterns. We propose a simplified histology-driven workup which could efficiently rule out CUPs and identify early metastatic cancer.


Asunto(s)
Adenocarcinoma , Carcinoma de Células Escamosas , Neoplasias Primarias Desconocidas , Humanos , Femenino , Neoplasias Primarias Desconocidas/diagnóstico , Neoplasias Primarias Desconocidas/patología , Inmunohistoquímica , Adenocarcinoma/metabolismo , Queratinas/análisis , Carcinoma de Células Escamosas/diagnóstico , Biomarcadores de Tumor/análisis
10.
EMBO Mol Med ; 15(3): e16104, 2023 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-36722641

RESUMEN

The genetic changes sustaining the development of cancers of unknown primary (CUP) remain elusive. The whole-exome genomic profiling of 14 rigorously selected CUP samples did not reveal specific recurring mutation in known driver genes. However, by comparing the mutational landscape of CUPs with that of most other human tumor types, it emerged a consistent enrichment of changes in genes belonging to the axon guidance KEGG pathway. In particular, G842C mutation of PlexinB2 (PlxnB2) was predicted to be activating. Indeed, knocking down the mutated, but not the wild-type, PlxnB2 in CUP stem cells resulted in the impairment of self-renewal and proliferation in culture, as well as tumorigenic capacity in mice. Conversely, the genetic transfer of G842C-PlxnB2 was sufficient to promote CUP stem cell proliferation and tumorigenesis in mice. Notably, G842C-PlxnB2 expression in CUP cells was associated with basal EGFR phosphorylation, and EGFR blockade impaired the viability of CUP cells reliant on the mutated receptor. Moreover, the mutated PlxnB2 elicited CUP cell invasiveness, blocked by EGFR inhibitor treatment. In sum, we found that a novel activating mutation of the axon guidance gene PLXNB2 sustains proliferative autonomy and confers invasive properties to stem cells isolated from cancers of unknown primary, in EGFR-dependent manner.


Asunto(s)
Neoplasias Primarias Desconocidas , Células Madre Neoplásicas , Proteínas del Tejido Nervioso , Animales , Humanos , Ratones , Orientación del Axón , Receptores ErbB/genética , Mutación , Recurrencia Local de Neoplasia , Neoplasias Primarias Desconocidas/genética , Proteínas del Tejido Nervioso/genética , Células Madre Neoplásicas/patología
11.
Clin Cancer Res ; 29(7): 1252-1266, 2023 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-36648487

RESUMEN

PURPOSE: Current glioma diagnostic guidelines call for molecular profiling to stratify patients into prognostic and treatment subgroups. In case the tumor tissue is inaccessible, cerebrospinal fluid (CSF) has been proposed as a reliable tumor DNA source for liquid biopsy. We prospectively investigated the use of CSF for molecular characterization of newly diagnosed gliomas. EXPERIMENTAL DESIGN: We recruited two cohorts of newly diagnosed patients with glioma, one (n = 45) providing CSF collected in proximity of the tumor, the other (n = 39) CSF collected by lumbar puncture (LP). Both cohorts provided tumor tissues by surgery concomitant with CSF sampling. DNA samples retrieved from CSF and matched tumors were systematically characterized and compared by comprehensive (NGS, next-generation sequencing) or targeted (ddPCR, droplet digital PCR) methodologies. Conventional and molecular diagnosis outcomes were compared. RESULTS: We report that tumor DNA is abundant in CSF close to the tumor, but scanty and mostly below NGS sensitivity threshold in CSF from LP. Indeed, tumor DNA is mostly released by cells invading liquoral spaces, generating a gradient that attenuates by departing from the tumor. Nevertheless, in >60% of LP CSF samples, tumor DNA is sufficient to assess a selected panel of genetic alterations (IDH and TERT promoter mutations, EGFR amplification, CDKN2A/B deletion: ITEC protocol) and MGMT methylation that, combined with imaging, enable tissue-agnostic identification of main glioma molecular subtypes. CONCLUSIONS: This study shows potentialities and limitations of CSF liquid biopsy in achieving molecular characterization of gliomas at first clinical presentation and proposes a protocol to maximize diagnostic information retrievable from CSF DNA.


Asunto(s)
Neoplasias Encefálicas , Glioma , Humanos , Glioma/diagnóstico , Glioma/genética , Glioma/patología , Mutación , Pronóstico , Biopsia Líquida , ADN de Neoplasias , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/genética , Biomarcadores de Tumor/genética
12.
Cell Rep ; 42(8): 112816, 2023 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-37505981

RESUMEN

Glioblastoma (GBM) is known as an intractable, highly heterogeneous tumor encompassing multiple subclones, each supported by a distinct glioblastoma stem cell (GSC). The contribution of GSC genetic and transcriptional heterogeneity to tumor subclonal properties is debated. In this study, we describe the systematic derivation, propagation, and characterization of multiple distinct GSCs from single, treatment-naive GBMs (GSC families). The tumorigenic potential of each GSC better correlates with its transcriptional profile than its genetic make-up, with classical GSCs being inherently more aggressive and mesenchymal more dependent on exogenous growth factors across multiple GBMs. These GSCs can segregate and recapitulate different histopathological aspects of the same GBM, as shown in a paradigmatic tumor with two histopathologically distinct components, including a conventional GBM and a more aggressive primitive neuronal component. This study provides a resource for investigating how GSCs with distinct genetic and/or phenotypic features contribute to individual GBM heterogeneity and malignant escalation.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Humanos , Glioblastoma/patología , Neoplasias Encefálicas/metabolismo , Amplificación de Genes , Células Madre Neoplásicas/metabolismo , Carcinogénesis/patología , Línea Celular Tumoral
13.
Life Sci Alliance ; 5(10)2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35636967

RESUMEN

MET is an oncogene encoding the tyrosine kinase receptor for hepatocyte growth factor (HGF). Upon ligand binding, MET activates multiple signal transducers, including PI3K/AKT, STAT3, and MAPK. When mutated or amplified, MET becomes a "driver" for the onset and progression of cancer. The most frequent mutations in the MET gene affect the splicing sites of exon 14, leading to the deletion of the receptor's juxtamembrane domain (MET∆14). It is currently believed that, as in gene amplification, MET∆14 kinase is constitutively active. Our analysis of MET in carcinoma cell lines showed that MET∆14 strictly depends on HGF for kinase activation. Compared with wt MET, ∆14 is sensitive to lower HGF concentrations, with more sustained kinase response. Using three different models, we have demonstrated that MET∆14 activation leads to robust phosphorylation of AKT, leading to a distinctive transcriptomic signature. Functional studies revealed that ∆14 activation is predominantly responsible for enhanced protection from apoptosis and cellular migration. Thus, the unique HGF-dependent ∆14 oncogenic activity suggests consideration of HGF in the tumour microenvironment to select patients for clinical trials.


Asunto(s)
Proteínas Proto-Oncogénicas c-akt , Proteínas Proto-Oncogénicas c-met , Humanos , Ligandos , Oncogenes , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-met/genética , Proteínas Proto-Oncogénicas c-met/metabolismo
14.
J Biol Chem ; 285(34): 26335-40, 2010 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-20554517

RESUMEN

Met, the tyrosine kinase receptor for the hepatocyte growth factor is a prominent regulator of cancer cell invasiveness and has emerged as a promising therapeutic target. Binding of the anti-Met monoclonal antibody DN30 to its epitope induces the proteolytic cleavage of Met, thereby impairing the invasive growth of tumors. The molecular mechanism controlling this therapeutic shedding process has so far been unknown. Here, we report that A Disintegrin And Metalloproteinase (ADAM)-10, but not ADAM-17, is required for DN30-induced Met shedding. Knockdown of ADAM-10 in different tumor cell lines or abrogation of its proteolytic activity by natural or synthetic inhibitors abolished Met down-regulation on the cell surface as well as reduction of Met activation. Moreover, hepatocyte growth factor-induced tumor cell migration and invasion were impaired upon ADAM-10 knockdown. Thus, the therapeutic effect of DN30 involves ADAM-10-dependent Met shedding, linking for the first time a specific metalloprotease to target therapy against a receptor tyrosine kinase.


Asunto(s)
Proteínas ADAM/fisiología , Secretasas de la Proteína Precursora del Amiloide/fisiología , Anticuerpos/uso terapéutico , Proteínas de la Membrana/fisiología , Proteínas Proto-Oncogénicas c-met/metabolismo , Receptores de Factores de Crecimiento/metabolismo , Proteína ADAM10 , Anticuerpos/farmacología , Línea Celular Tumoral , Factor de Crecimiento de Hepatocito/farmacología , Humanos , Invasividad Neoplásica/prevención & control , Metástasis de la Neoplasia/tratamiento farmacológico , Metástasis de la Neoplasia/prevención & control , Proteínas Proto-Oncogénicas c-met/efectos de los fármacos , Receptores de Factores de Crecimiento/efectos de los fármacos
15.
Curr Opin Cell Biol ; 15(5): 565-71, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-14519391

RESUMEN

Adhesion molecules, although catalytically inactive, are able to translate environmental cues into complex intracellular signals. They can do this by associating with tyrosine kinase receptors for growth factors, which can prime, integrate or feedback adhesion-based signals. Recent results show that reciprocal crosstalk between the two systems is only one facet of such a collaboration, and that unconventional and alternative hierarchies can be established in which, on the one hand, cell adhesion can trigger ligand-independent activation of growth factor receptors, and, on the other, growth factors can induce adhesion molecules to propagate adhesion-independent signals.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Sustancias de Crecimiento/metabolismo , Factor de Crecimiento de Hepatocito , Proteínas Tirosina Quinasas/metabolismo , Receptores de Factores de Crecimiento/metabolismo , Transducción de Señal/fisiología , Cadherinas/metabolismo , Adhesión Celular/fisiología , Matriz Extracelular/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal , Integrinas/metabolismo , Mitógenos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Mapeo de Interacción de Proteínas
16.
Nature ; 434(7031): 396-400, 2005 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-15772665

RESUMEN

The close relationship between activation of blood coagulation and cancer is an old enigma. In 1865, migrans trombophlebitis ('a condition of the blood that predisposes it to spontaneous coagulation') was described as a forewarning of occult malignancy (Trousseau's sign). This pioneering observation emphasized the existence of haemostasis disorders associated with cancer onset; this phenomenon has since been extensively reported in clinical and epidemiological studies, but has so far resisted a mechanistic explanation. Here we report a mouse model of sporadic tumorigenesis based on genetic manipulation of somatic cells. Targeting the activated, human MET oncogene to adult liver caused slowly progressing hepatocarcinogenesis. This was preceded and accompanied by a syndrome manifesting first with blood hypercoagulation (venous thromboses), and then evolving towards fatal internal haemorrhages. The pathogenesis of this syndrome is driven by the transcriptional response to the oncogene, including prominent upregulation of plasminogen activator inhibitor type 1 (PAI-1) and cyclooxygenase-2 (COX-2) genes. In vivo analysis showed that both proteins support the thrombohaemorrhagic phenotype, thus providing direct genetic evidence for the long-sought-after link between oncogene activation and haemostasis.


Asunto(s)
Coagulación Sanguínea , Transformación Celular Neoplásica/genética , Hemostasis/fisiología , Neoplasias Hepáticas/genética , Oncogenes/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Receptores de Factores de Crecimiento/genética , Receptores de Factores de Crecimiento/metabolismo , Animales , Ciclooxigenasa 2 , Dimerización , Enfermedades Hematológicas/genética , Enfermedades Hematológicas/patología , Hemostasis/genética , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , Neoplasias Hepáticas/patología , Proteínas de la Membrana , Ratones , Ratones SCID , Inhibidor 1 de Activador Plasminogénico/metabolismo , Prostaglandina-Endoperóxido Sintasas/metabolismo , Proteínas Proto-Oncogénicas c-met , Transcripción Genética/genética , Regulación hacia Arriba/genética
17.
Mol Cell Oncol ; 8(5): 1990677, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34859149

RESUMEN

By exploiting an integrated experimental platform based on patient-derived cancer stem cells, we identified a glioblastoma subset characterized by inheritable Erb-B2 Receptor Tyrosine Kinase 3 (ERBB3) overexpression, metabolic dependency on ERBB3 signaling, and liability to ERBB3 targeting. We provide insights on why some glioblastomas may rely on ERBB3 and how to recognize them.

18.
Cell Rep ; 36(4): 109455, 2021 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-34320350

RESUMEN

In glioblastoma (GBM), the most frequent and lethal brain tumor, therapies suppressing recurrently altered signaling pathways failed to extend survival. However, in patient subsets, specific genetic lesions can confer sensitivity to targeted agents. By exploiting an integrated model based on patient-derived stem-like cells, faithfully recapitulating the original GBMs in vitro and in vivo, here, we identify a human GBM subset (∼9% of all GBMs) characterized by ERBB3 overexpression and nuclear accumulation. ERBB3 overexpression is driven by inheritable promoter methylation or post-transcriptional silencing of the oncosuppressor miR-205 and sustains the malignant phenotype. Overexpressed ERBB3 behaves as a specific signaling platform for fibroblast growth factor receptor (FGFR), driving PI3K/AKT/mTOR pathway hyperactivation, and overall metabolic upregulation. As a result, ERBB3 inhibition by specific antibodies is lethal for GBM stem-like cells and xenotransplants. These findings highlight a subset of patients eligible for ERBB3-targeted therapy.


Asunto(s)
Glioblastoma/genética , MicroARNs/metabolismo , Receptor ErbB-3/metabolismo , Anticuerpos/metabolismo , Apoptosis , Línea Celular Tumoral , Factor 2 de Crecimiento de Fibroblastos , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor , Humanos , MicroARNs/genética , Oligodendroglía/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Pronóstico , Proteínas Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor ErbB-3/antagonistas & inhibidores , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal , Esferoides Celulares/patología , Serina-Treonina Quinasas TOR/metabolismo
19.
Nat Commun ; 12(1): 2498, 2021 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-33941777

RESUMEN

Cancers of unknown primary (CUPs), featuring metastatic dissemination in the absence of a primary tumor, are a biological enigma and a fatal disease. We propose that CUPs are a distinct, yet unrecognized, pathological entity originating from stem-like cells endowed with peculiar and shared properties. These cells can be isolated in vitro (agnospheres) and propagated in vivo by serial transplantation, displaying high tumorigenicity. After subcutaneous engraftment, agnospheres recapitulate the CUP phenotype, by spontaneously and quickly disseminating, and forming widespread established metastases. Regardless of different genetic backgrounds, agnospheres invariably display cell-autonomous proliferation and self-renewal, mostly relying on unrestrained activation of the MAP kinase/MYC axis, which confers sensitivity to MEK inhibitors in vitro and in vivo. Such sensitivity is associated with a transcriptomic signature predicting that more than 70% of CUP patients could be eligible to MEK inhibition. These data shed light on CUP biology and unveil an opportunity for therapeutic intervention.


Asunto(s)
Carcinogénesis/patología , Metástasis de la Neoplasia/patología , Neoplasias Primarias Desconocidas/patología , Células Madre Neoplásicas/patología , Esferoides Celulares/patología , Animales , Carcinogénesis/genética , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica/genética , Xenoinjertos , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Metástasis de la Neoplasia/genética , Trasplante de Neoplasias , Neoplasias Primarias Desconocidas/genética , Células Tumorales Cultivadas
20.
Br J Pharmacol ; 177(13): 3107-3122, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32133617

RESUMEN

BACKGROUND AND PURPOSE: Doxorubicin anti-cancer therapy is associated with cardiotoxicity, resulting from DNA damage response (DDR). Hepatocyte growth factor (HGF) protects cardiomyocytes from injury, but its effective use is compromised by low biodistribution. In this study, we have investigated whether the activation of the HGF receptor-encoded by the Met gene-by an agonist monoclonal antibody (mAb) could protect against doxorubicin-induced cardiotoxicity. EXPERIMENTAL APPROACH: The mAb (5 mg·kg-1 ) was injected in vivo into C57BL/6J mice, before doxorubicin (three doses of 7 mg·kg-1 ). Cardiac functions were evaluated through MRI after treatment termination. Heart histological staining and mRNA levels of genes associated with heart failure (Acta1 and Nppa), inflammation (IL-6), and fibrosis (Ctgf, Col1a2, Timp1, and Mmp9) were assessed. MAb (100 nM) was administered in vitro to H9c2 cardiomyoblasts before addition of doxorubicin (25 µM). DDR and apoptosis markers were evaluated by quantitative western blotting, flow cytometry, and immunofluorescence. Stattic was used for pharmacological inactivation of STAT3. KEY RESULTS: In vivo, administration of the mAb alleviated doxorubicin-induced cardiac dysfunction and fibrosis. In vitro, mAb mimicked the response to HGF by (a) inhibiting histone H2AX phosphorylation at S139, (b) quenching the expression of the DNA repair enzyme PARP1, and (c) reducing the proteolytic activation of caspase 3. The MET-driven cardioprotection involved, at least in vitro, the phosphorylation of STAT3. CONCLUSION AND IMPLICATIONS: The MET agonist mAb provides a new tool for cardioprotection against anthracycline cardiotoxicity.


Asunto(s)
Cardiotoxicidad , Doxorrubicina , Animales , Antibióticos Antineoplásicos/toxicidad , Apoptosis , Cardiotoxicidad/metabolismo , Doxorrubicina/toxicidad , Ratones , Ratones Endogámicos C57BL , Miocitos Cardíacos/metabolismo , Distribución Tisular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA