Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Am Assoc Lab Anim Sci ; 61(5): 412-418, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-35944976

RESUMEN

Naked mole rats (Heterocephalus glaber) are a unique rodent species originating in Africa and are increasingly being used in research. Their needs and characteristics differ from those of other rodents used in research. Unique housing systems are necessary to address the special macro- and microenvironmental requirements of NMRs. Naked mole rats are one of the 2 known eusocial mammalian species, are extremely long-living, are active burrowers, and are accustomed to a subterranean environment. Unlike typical rats and mice, naked mole rats need specific, unique housing systems that mimic their natural subterranean environment to support health and longevity. Here we provide an overview of naked mole rats and a housing method that can be used in research settings.


Asunto(s)
Vivienda , Ratas Topo , Animales , Longevidad , Ratones
2.
Neurooncol Adv ; 4(1): vdac072, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35855490

RESUMEN

Background: Neurofibromatosis type 2 (NF2) is an autosomal dominant genetic disease characterized by development of schwannomas on the VIIIth (vestibular) cranial nerves. Bromodomain and extra-terminal domain (BET) proteins regulate gene transcription and their activity is required in a variety of cancers including malignant peripheral nerve sheath tumors. The use of BET inhibitors as a therapeutic option to treat NF2 schwannomas has not been explored and is the focus of this study. Methods: A panel of normal and NF2-null Schwann and schwannoma cell lines were used to characterize the impact of the BET inhibitor JQ1 in vitro and in vivo. The mechanism of action was explored by chromatin immunoprecipitation of the BET BRD4, phospho-kinase arrays and immunohistochemistry (IHC) of BRD4 in vestibular schwannomas. Results: JQ1 inhibited proliferation of NF2-null schwannoma and Schwann cell lines in vitro and in vivo. Further, loss of NF2 by CRISPR deletion or siRNA knockdown increased sensitivity of cells to JQ1. Loss of function experiments identified BRD4, and to a lesser extent BRD2, as BET family members mediating the majority of JQ1 effects. IHC demonstrated elevated levels of BRD4 protein in human vestibular schwannomas. Analysis of signaling pathways effected by JQ1 treatment suggests that the effects of JQ1 treatment are mediated, at least in part, via inhibition of PI3K/Akt signaling. Conclusions: NF2-deficient Schwann and schwannoma cells are sensitive to BET inhibition, primarily mediated by BRD4, which is overexpressed in human vestibular schwannomas. Our results suggest BRD4 regulates PI3K signaling and likely impedes NF2 schwannoma growth via this inhibition. These findings implicate BET inhibition as a therapeutic option for NF2-deficient schwannomas.

3.
J Biol Chem ; 297(2): 100962, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34265306

RESUMEN

The Hippo pathway is a key regulatory pathway that is tightly regulated by mechanical cues such as tension, pressure, and contact with the extracellular matrix and other cells. At the distal end of the pathway is the yes-associated protein (YAP), a well-characterized transcriptional regulator. Through binding to transcription factors such as the TEA Domain TFs (TEADs) YAP regulates expression of several genes involved in cell fate, proliferation and death decisions. While the function of YAP as direct transcriptional regulator has been extensively characterized, only a small number of studies examined YAP function as a regulator of gene expression via microRNAs. We utilized bioinformatic approaches, including chromatin immunoprecipitation sequencing and RNA-Seq, to identify potential new targets of YAP regulation and identified miR-30a as a YAP target gene in Schwann cells. We find that YAP binds to the promoter and regulates the expression of miR-30a. Moreover, we identify several YAP-regulated genes that are putative miR-30a targets and focus on two of these, protein tyrosine pohosphatase non-receptor type 13 (PTPN13) and Kruppel like factor 9. We find that YAP regulation of Schwann cell proliferation and death is mediated, to a significant extent, through miR-30a regulation of PTPN13 in Schwann cells. These findings identify a new regulatory function by YAP, mediated by miR-30a, to downregulate expression of PTPN13 and Kruppel like factor 9. These studies expand our understanding of YAP function as a regulator of miRNAs and illustrate the complexity of YAP transcriptional functions.


Asunto(s)
Factores de Transcripción de Tipo Kruppel/metabolismo , MicroARNs , Proteína Tirosina Fosfatasa no Receptora Tipo 13/metabolismo , Células de Schwann/patología , Proteínas Adaptadoras Transductoras de Señales , Diferenciación Celular , Línea Celular Tumoral , Proliferación Celular , Regulación hacia Abajo , Humanos , Transducción de Señal , Transcripción Genética , Proteínas Señalizadoras YAP
4.
Cancer Res ; 80(12): 2512-2522, 2020 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-32409309

RESUMEN

The Hippo pathway regulates cell proliferation and organ size through control of the transcriptional regulators YAP (yes-associated protein) and TAZ. Upon extracellular stimuli such as cell-cell contact, the pathway negatively regulates YAP through cytoplasmic sequestration. Under conditions of low cell density, YAP is nuclear and associates with enhancer regions and gene promoters. YAP is mainly described as a transcriptional activator of genes involved in cell proliferation and survival. Using a genome-wide approach, we show here that, in addition to its known function as a transcriptional activator, YAP functions as a transcriptional repressor by interacting with the multifunctional transcription factor Yin Yang 1 (YY1) and Polycomb repressive complex member enhancer of zeste homologue 2 (EZH2). YAP colocalized with YY1 and EZH2 on the genome to transcriptionally repress a broad network of genes mediating a host of cellular functions, including repression of the cell-cycle kinase inhibitor p27, whose role is to functionally promote contact inhibition. This work unveils a broad and underappreciated aspect of YAP nuclear function as a transcriptional repressor and highlights how loss of contact inhibition in cancer is mediated in part through YAP repressive function. SIGNIFICANCE: This study provides new insights into YAP as a broad transcriptional repressor of key regulators of the cell cycle, in turn influencing contact inhibition and tumorigenesis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Ciclo Celular/genética , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Neoplasias/genética , Factores de Transcripción/metabolismo , Transcripción Genética , Factor de Transcripción YY1/metabolismo , Animales , Carcinogénesis/genética , Fraccionamiento Celular , Línea Celular Tumoral , Núcleo Celular/metabolismo , Proliferación Celular/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes/genética , Humanos , Ratones , Neoplasias/patología , Regiones Promotoras Genéticas/genética , Transducción de Señal/genética , Ensayos Antitumor por Modelo de Xenoinjerto , Proteínas Señalizadoras YAP
5.
Cell Rep ; 29(6): 1660-1674.e7, 2019 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-31693903

RESUMEN

The incidence of human papilloma virus (HPV)-associated head and neck squamous cell carcinoma (HNSCC) is increasing and implicated in more than 60% of all oropharyngeal carcinomas (OPSCCs). Although whole-genome, transcriptome, and proteome analyses have identified altered signaling pathways in HPV-induced HNSCCs, additional tools are needed to investigate the unique pathobiology of OPSCC. Herein, bioinformatics analyses of human HPV(+) HNSCCs revealed that all tumors express full-length E6 and identified molecular subtypes based on relative E6 and E7 expression levels. To recapitulate the levels, stoichiometric ratios, and anatomic location of E6/E7 expression, we generated a genetically engineered mouse model whereby balanced expression of E6/E7 is directed to the oropharyngeal epithelium. The addition of a mutant PIK3CAE545K allele leads to the rapid development of pre-malignant lesions marked by immune cell accumulation, and a subset of these lesions progress to OPSCC. This mouse provides a faithful immunocompetent model for testing treatments and investigating mechanisms of immunosuppression.


Asunto(s)
Modelos Animales de Enfermedad , Neoplasias de Cabeza y Cuello/virología , Proteínas Oncogénicas Virales/metabolismo , Neoplasias Orofaríngeas/virología , Proteínas E7 de Papillomavirus/metabolismo , Proteínas Represoras/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/virología , Animales , Carcinogénesis/genética , Carcinogénesis/metabolismo , Línea Celular Tumoral , Fosfatidilinositol 3-Quinasa Clase I/genética , Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Femenino , Expresión Génica , Neoplasias de Cabeza y Cuello/genética , Neoplasias de Cabeza y Cuello/inmunología , Neoplasias de Cabeza y Cuello/metabolismo , Humanos , Inmunocompetencia , Sitios Internos de Entrada al Ribosoma/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células T Asesinas Naturales/inmunología , Células T Asesinas Naturales/metabolismo , Proteínas Oncogénicas Virales/genética , Neoplasias Orofaríngeas/genética , Neoplasias Orofaríngeas/inmunología , Neoplasias Orofaríngeas/metabolismo , Proteínas E7 de Papillomavirus/genética , Empalme del ARN/genética , RNA-Seq , Proteínas Represoras/genética , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Carcinoma de Células Escamosas de Cabeza y Cuello/inmunología , Carcinoma de Células Escamosas de Cabeza y Cuello/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo
6.
Oncogene ; 38(36): 6370-6381, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31312020

RESUMEN

Inactivation of the tumor suppressor NF2/merlin underlies neurofibromatosis type 2 (NF2) and some sporadic tumors. Previous studies have established that merlin mediates contact inhibition of proliferation; however, the exact mechanisms remain obscure and multiple pathways have been implicated. We have previously reported that merlin inhibits Ras and Rac activity during contact inhibition, but how merlin regulates Ras activity has remained elusive. Here we demonstrate that merlin can directly interact with both Ras and p120RasGAP (also named RasGAP). While merlin does not increase the catalytic activity of RasGAP, the interactions with Ras and RasGAP may fine-tune Ras signaling. In vivo, loss of RasGAP in Schwann cells, unlike the loss of merlin, failed to promote tumorigenic growth in an orthotopic model. Therefore, modulation of Ras signaling through RasGAP likely contributes to, but is not sufficient to account for, merlin's tumor suppressor activity. Our study provides new insight into the mechanisms of merlin-dependent Ras regulation and may have additional implications for merlin-dependent regulation of other small GTPases.


Asunto(s)
Neurofibromina 2/fisiología , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Proteínas Activadoras de ras GTPasa/metabolismo , Animales , Células Cultivadas , Proteínas Activadoras de GTPasa/metabolismo , Genes Supresores de Tumor , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Neurofibromatosis 2/genética , Neurofibromatosis 2/metabolismo , Neurofibromina 2/metabolismo , Unión Proteica , Transducción de Señal/genética
7.
Oncogene ; 37(32): 4372-4384, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29743592

RESUMEN

The RAS proteins are the most frequently mutated oncogenes in cancer, with highest frequency found in pancreatic, lung, and colon tumors. Moreover, the activity of RAS is required for the proliferation and/or survival of these tumor cells and thus represents a high-value target for therapeutic development. Direct targeting of RAS has proven challenging for multiple reasons stemming from the biology of the protein, the complexity of downstream effector pathways and upstream regulatory networks. Thus, significant efforts have been directed at identifying downstream targets on which RAS is dependent. These efforts have proven challenging, in part due to confounding factors such as reliance on two-dimensional adherent monolayer cell cultures that inadequately recapitulate the physiologic context to which cells are exposed in vivo. To overcome these issues, we implemented a high-throughput screening (HTS) approach using a spheroid-based 3-dimensional culture format, thought to more closely reflect conditions experienced by cells in vivo. Using isogenic cell pairs, differing in the status of KRAS, we identified Proscillaridin A as a selective inhibitor of cells harboring the oncogenic KRasG12V allele. Significantly, the identification of Proscillaridin A was facilitated by the 3D screening platform and would not have been discovered employing standard 2D culturing methods.


Asunto(s)
Mutación/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Antineoplásicos/farmacología , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales/métodos , Humanos , Fenotipo , Proscilaridina/farmacología , Transducción de Señal/genética
8.
Elife ; 62017 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-28464980

RESUMEN

The Hippo-YAP pathway is a central regulator of cell contact inhibition, proliferation and death. There are conflicting reports regarding the role of Angiomotin (Amot) in regulating this pathway. While some studies suggest a YAP-inhibitory function other studies indicate Amot is required for YAP activity. Here, we describe an Amot-dependent complex comprised of Amot, YAP and Merlin. The phosphorylation of Amot at Serine 176 shifts localization of this complex to the plasma membrane, where it associates with the tight-junction proteins Pals1/PATJ and E-cadherin. Conversely, hypophosphorylated Amot shifts localization of the complex to the nucleus, where it facilitates the association of YAP and TEAD, induces transcriptional activation of YAP target genes and promotes YAP-dependent cell proliferation. We propose that phosphorylation of AmotS176 is a critical post-translational modification that suppresses YAP's ability to promote cell proliferation and tumorigenesis by altering the subcellular localization of an essential YAP co-factor.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas de la Membrana/metabolismo , Neurofibromina 2/metabolismo , Fosfoproteínas/metabolismo , Multimerización de Proteína , Procesamiento Proteico-Postraduccional , Angiomotinas , Membrana Celular/química , Núcleo Celular/química , Células HEK293 , Células Hep G2 , Humanos , Proteínas de Microfilamentos , Fosforilación , Unión Proteica , Factores de Transcripción , Proteínas Señalizadoras YAP
9.
Oncotarget ; 7(34): 54515-54525, 2016 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-27363027

RESUMEN

Neurofibromatosis type 2 (NF2) is a dominantly inherited autosomal disease characterized by schwannomas of the 8th cranial nerve. The NF2 tumor suppressor gene encodes for Merlin, a protein implicated as a suppressor of multiple cellular signaling pathways. To identify potential drug targets in NF2-associated malignancies we assessed the consequences of inhibiting the tyrosine kinase receptor MET. We identified crizotinib, a MET and ALK inhibitor, as a potent inhibitor of NF2-null Schwann cell proliferation in vitro and tumor growth in vivo. To identify the target/s of crizotnib we employed activity-based protein profiling (ABPP), leading to identification of FAK1 (PTK2) as the relevant target of crizotinib inhibition in NF2-null schwannoma cells. Subsequent studies confirm that inhibition of FAK1 is sufficient to suppress tumorigenesis in animal models of NF2 and that crizotinib-resistant forms of FAK1 can rescue the effects of treatment. These studies identify a FDA approved drug as a potential treatment for NF2 and delineate the mechanism of action in NF2-null Schwann cells.


Asunto(s)
Quinasa 1 de Adhesión Focal/antagonistas & inhibidores , Neurilemoma/tratamiento farmacológico , Neurofibromina 2/fisiología , Inhibidores de Proteínas Quinasas/farmacología , Pirazoles/farmacología , Piridinas/farmacología , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Crizotinib , Humanos , Ratones , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Cancer Res ; 76(12): 3507-19, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27216189

RESUMEN

The Hippo-YAP pathway has emerged as a major driver of tumorigenesis in many human cancers. YAP is a transcriptional coactivator and while details of YAP regulation are quickly emerging, it remains unknown what downstream targets are critical for the oncogenic functions of YAP. To determine the mechanisms involved and to identify disease-relevant targets, we examined the role of YAP in neurofibromatosis type 2 (NF2) using cell and animal models. We found that YAP function is required for NF2-null Schwann cell survival, proliferation, and tumor growth in vivo Moreover, YAP promotes transcription of several targets including PTGS2, which codes for COX-2, a key enzyme in prostaglandin biosynthesis, and AREG, which codes for the EGFR ligand, amphiregulin. Both AREG and prostaglandin E2 converge to activate signaling through EGFR. Importantly, treatment with the COX-2 inhibitor celecoxib significantly inhibited the growth of NF2-null Schwann cells and tumor growth in a mouse model of NF2. Cancer Res; 76(12); 3507-19. ©2016 AACR.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Ciclooxigenasa 2/fisiología , Receptores ErbB/fisiología , Neurofibromatosis 2/etiología , Fosfoproteínas/fisiología , Transducción de Señal/fisiología , Anfirregulina/fisiología , Carcinogénesis , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Humanos , Células de Schwann/fisiología , Factores de Transcripción , Proteínas Señalizadoras YAP
11.
Sci Signal ; 6(291): ra77, 2013 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-24003254

RESUMEN

The Hippo-Yap signaling pathway regulates a number of developmental and adult cellular processes, including cell fate determination, tissue growth, and tumorigenesis. Members of the scaffold protein angiomotin (Amot) family interact with several Hippo pathway components, including Yap (Yes-associated protein), and either stimulate or inhibit Yap activity. We used a combination of genetic, biochemical, and transcriptional approaches to assess the functional consequences of the Amot-Yap interaction in mice and in human cells. Mice with a liver-specific Amot knockout exhibited reduced hepatic "oval cell" proliferation and tumorigenesis in response to toxin-induced injury or when crossed with mice lacking the tumor suppressor Nf2. Biochemical examination of the Amot-Yap interaction revealed that the p130 splicing isoform of Amot (Amot-p130) and Yap interacted in both the cytoplasm and nucleus, which involved binding of PPxY and LPxY motifs in Amot-p130 to WW domains of Yap. In the cytoplasm, Amot-p130 prevented the phosphorylation of Yap by blocking access of the WW domains to the kinase Lats1. Within the nucleus, Amot-p130 was associated with the transcriptional complex containing Yap and Teads (TEA domain family members) and contributed to the regulation of a subset of Yap target genes, many of which are associated with tumorigenesis. These findings indicated that Amot acts as a Yap cofactor, preventing Yap phosphorylation and augmenting its activity toward a specific set of genes that facilitate tumorigenesis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Transformación Celular Neoplásica/metabolismo , Células Epiteliales/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Neoplasias Hepáticas/metabolismo , Hígado/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Microfilamentos/metabolismo , Proteínas de Neoplasias/metabolismo , Fosfoproteínas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Secuencias de Aminoácidos , Angiomotinas , Animales , Proteínas de Ciclo Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Células Epiteliales/patología , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Hígado/patología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Proteínas de Microfilamentos/genética , Proteínas de Neoplasias/genética , Fosfoproteínas/genética , Fosforilación/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estructura Terciaria de Proteína , Factores de Transcripción , Proteínas Señalizadoras YAP
12.
J Biol Chem ; 288(40): 29105-14, 2013 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-23960073

RESUMEN

The p21-activated kinases (PAKs) are immediate downstream effectors of the Rac/Cdc42 small G-proteins and implicated in promoting tumorigenesis in various types of cancer including breast and lung carcinomas. Recent studies have established a requirement for the PAKs in the pathogenesis of Neurofibromatosis type 2 (NF2), a dominantly inherited cancer disorder caused by mutations at the NF2 gene locus. Merlin, the protein product of the NF2 gene, has been shown to negatively regulate signaling through the PAKs and the tumor suppressive functions of Merlin are mediated, at least in part, through inhibition of the PAKs. Knockdown of PAK1 and PAK2 expression, through RNAi-based approaches, impairs the proliferation of NF2-null schwannoma cells in culture and inhibits their ability to form tumors in vivo. These data implicate the PAKs as potential therapeutic targets. High-throughput screening of a library of small molecules combined with a structure-activity relationship approach resulted in the identification of FRAX597, a small-molecule pyridopyrimidinone, as a potent inhibitor of the group I PAKs. Crystallographic characterization of the FRAX597/PAK1 complex identifies a phenyl ring that traverses the gatekeeper residue and positions the thiazole in the back cavity of the ATP binding site, a site rarely targeted by kinase inhibitors. FRAX597 inhibits the proliferation of NF2-deficient schwannoma cells in culture and displayed potent anti-tumor activity in vivo, impairing schwannoma development in an orthotopic model of NF2. These studies identify a novel class of orally available ATP-competitive Group I PAK inhibitors with significant potential for the treatment of NF2 and other cancers.


Asunto(s)
Carcinogénesis/patología , Neurilemoma/tratamiento farmacológico , Neurilemoma/enzimología , Neurofibromatosis 2/tratamiento farmacológico , Piridonas/uso terapéutico , Pirimidinas/uso terapéutico , Pirimidinonas/uso terapéutico , Bibliotecas de Moléculas Pequeñas/uso terapéutico , Quinasas p21 Activadas/antagonistas & inhibidores , Animales , Carcinogénesis/efectos de los fármacos , Dominio Catalítico , Proliferación Celular/efectos de los fármacos , Descubrimiento de Drogas , Humanos , Ratones , Modelos Moleculares , Neurilemoma/patología , Neurofibromatosis 2/enzimología , Neurofibromatosis 2/patología , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Piridonas/química , Piridonas/farmacología , Pirimidinas/química , Pirimidinas/farmacología , Pirimidinonas/química , Pirimidinonas/farmacología , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Quinasas p21 Activadas/metabolismo
13.
Cancer Res ; 73(19): 5974-84, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-23943799

RESUMEN

The Notch pathway has been implicated in a number of malignancies with different roles that are cell- and tissue-type dependent. Notch1 is a putative oncogene in non-small cell lung cancer (NSCLC) and activation of the pathway represents a negative prognostic factor. To establish the role of Notch1 in lung adenocarcinoma, we directly assessed its requirement in Kras-induced tumorigenesis in vivo using an autochthonous model of lung adenocarcinoma with concomitant expression of oncogenic Kras and deletion of Notch1. We found that Notch1 function is required for tumor initiation via suppression of p53-mediated apoptosis through the regulation of p53 stability. These findings implicate Notch1 as a critical effector in Kras-driven lung adenocarcinoma and as a regulator of p53 at a posttranslational level. Moreover, our study provides new insights to explain, at a molecular level, the correlation between Notch1 activity and poor prognosis in patients with NSCLC carrying wild-type p53. This information is critical for design and implementation of new therapeutic strategies in this cohort of patients representing 50% of NSCLC cases.


Asunto(s)
Adenocarcinoma/patología , Apoptosis , Transformación Celular Neoplásica , Neoplasias Pulmonares/patología , Proteínas Proto-Oncogénicas p21(ras)/fisiología , Receptor Notch1/fisiología , Proteína p53 Supresora de Tumor/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Animales , Western Blotting , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Ciclo Celular , Proliferación Celular , Técnica del Anticuerpo Fluorescente , Humanos , Técnicas para Inmunoenzimas , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Ratones , Ratones Noqueados , Mutación/genética , Transducción de Señal , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/química
14.
Chem Biol ; 19(4): 518-28, 2012 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-22520758

RESUMEN

The retinoblastoma protein pRb is essential for regulating many cellular activities through its binding and inhibition of E2F transcription activators, and pRb inactivation leads to many cancers. pRb activity can be perturbed by viral oncoproteins including human papillomavirus (HPV) that share an LxCxE motif. Because there are no treatments for existing HPV infection leading to nearly all cervical cancers and other cancers to a lesser extent, we screened for compounds that inhibit the ability of HPV-E7 to disrupt pRb/E2F complexes. This lead to the identification of thiadiazolidinedione compounds that bind to pRb with mid-high nanomolar dissociation constants, are competitive with the binding of viral oncoproteins containing an LxCxE motif, and are selectively cytotoxic in HPV-positive cells alone and in mice. These inhibitors provide a promising scaffold for the development of therapies to treat HPV-mediated pathologies.


Asunto(s)
Proteínas E7 de Papillomavirus/metabolismo , Proteína de Retinoblastoma/antagonistas & inhibidores , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Factores de Transcripción E2F/metabolismo , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Células HeLa , Humanos , Ratones , Datos de Secuencia Molecular , Papillomaviridae/efectos de los fármacos , Papillomaviridae/metabolismo , Proteínas E7 de Papillomavirus/antagonistas & inhibidores , Proteína de Retinoblastoma/metabolismo , Alineación de Secuencia , Tiadiazoles/química , Tiadiazoles/farmacología
15.
PLoS One ; 7(12): e52133, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23284900

RESUMEN

Pancreatic ductal adenocarcinoma is believed to arise from precursor lesions termed pancreatic intraepithelial neoplasia (PanIN). Mouse models have demonstrated that targeted expression of activated K-ras to mature acinar cells in the pancreas induces the spontaneous development of PanIN lesions; implying acinar-to-ductal metaplasia (ADM) is a key event in this process. Recent studies suggest Notch signaling is a key regulator of ADM. To assess if Notch1 is required for K-ras driven ADM we employed both an in vivo mouse model and in vitro explant culture system, in which an oncogenic allele of K-ras is activated and Notch1 is deleted simultaneously in acinar cells. Our results demonstrate that oncogenic K-ras is sufficient to drive ADM both in vitro and in vivo but that loss of Notch1 has a minimal effect on this process. Interestingly, while loss of Notch1 in vivo does not affect the severity of PanIN lesions observed, the overall numbers of lesions were greater in mice with deleted Notch1. This suggests Notch1 deletion renders acinar cells more susceptible to formation of K-ras-induced PanINs.


Asunto(s)
Células Acinares/patología , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Genes ras , Receptor Notch1/genética , Animales , Carcinoma in Situ/genética , Carcinoma in Situ/metabolismo , Carcinoma in Situ/patología , Carcinoma Ductal Pancreático/metabolismo , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Modelos Animales de Enfermedad , Eliminación de Gen , Metaplasia , Ratones , Pancreatitis/genética , Pancreatitis/metabolismo , Pancreatitis/patología , Lesiones Precancerosas/genética , Lesiones Precancerosas/metabolismo , Lesiones Precancerosas/patología , Receptor Notch1/metabolismo , Transducción de Señal
16.
Cancer Cell ; 19(4): 527-40, 2011 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-21481793

RESUMEN

The Merlin/NF2 tumor suppressor restrains cell growth and tumorigenesis by controlling contact-dependent inhibition of proliferation. We have identified a tight-junction-associated protein complex comprising Merlin, Angiomotin, Patj, and Pals1. We demonstrate that Angiomotin functions downstream of Merlin and upstream of Rich1, a small GTPase Activating Protein, as a positive regulator of Rac1. Merlin, through competitive binding to Angiomotin, releases Rich1 from the Angiomotin-inhibitory complex, allowing Rich1 to inactivate Rac1, ultimately leading to attenuation of Rac1 and Ras-MAPK pathways. Patient-derived Merlin mutants show diminished binding capacities to Angiomotin and are unable to dissociate Rich1 from Angiomotin or inhibit MAPK signaling. Depletion of Angiomotin in Nf2(-/-) Schwann cells attenuates the Ras-MAPK signaling pathway, impedes cellular proliferation in vitro and tumorigenesis in vivo.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/fisiología , Proteínas de la Membrana/fisiología , Proteínas de Microfilamentos/fisiología , Neurofibromina 2/fisiología , Transducción de Señal/fisiología , Uniones Estrechas/fisiología , Proteínas Supresoras de Tumor/fisiología , Secuencia de Aminoácidos , Angiomotinas , Animales , Proliferación Celular , Proteínas Activadoras de GTPasa/fisiología , Humanos , Sistema de Señalización de MAP Quinasas , Ratones , Ratones SCID , Datos de Secuencia Molecular , Nucleósido-Fosfato Quinasa/fisiología , Nervios Periféricos/química , Células de Schwann/química , Proteínas de Uniones Estrechas , Proteína de Unión al GTP rac1/fisiología
17.
Cancer Res ; 70(11): 4280-6, 2010 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-20484026

RESUMEN

K-ras is the most commonly mutated oncogene in pancreatic cancer and its activation in murine models is sufficient to recapitulate the spectrum of lesions seen in human pancreatic ductal adenocarcinoma (PDAC). Recent studies suggest that Notch receptor signaling becomes reactivated in a subset of PDACs, leading to the hypothesis that Notch1 functions as an oncogene in this setting. To determine whether Notch1 is required for K-ras-induced tumorigenesis, we used a mouse model in which an oncogenic allele of K-ras is activated and Notch1 is deleted simultaneously in the pancreas. Unexpectedly, the loss of Notch1 in this model resulted in increased tumor incidence and progression, implying that Notch1 can function as a tumor suppressor gene in PDAC.


Asunto(s)
Carcinoma Ductal Pancreático/genética , Genes Supresores de Tumor , Genes ras , Neoplasias Pancreáticas/genética , Receptor Notch1/genética , Animales , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patología , Progresión de la Enfermedad , Eliminación de Gen , Regulación Neoplásica de la Expresión Génica , Ratones , Ratones Transgénicos , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Receptor Notch1/deficiencia , Receptor Notch1/metabolismo , Transducción de Señal , beta Catenina/metabolismo
18.
Mol Cell Biol ; 25(22): 10052-9, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16260618

RESUMEN

The band 4.1 proteins are cytoskeletal proteins, harboring a conserved FERM domain highly homologous to the N-terminal FERM domain of ezrin, radixin, moesin, and merlin. Recently, a truncated form of the 4.1B protein, termed Dal-1, was identified in a screen as down regulated in adenocarcinoma of the lung and was mapped to chromosome 18p11.3, which is lost in 38% of primary non-small cell lung carcinoma tumors. Analysis of several meningiomas has shown that Dal-1 expression was lost in 76% of the tumors. To further elucidate the function of the 4.1B/Dal-1 gene in development and tumorigenesis we generated mice deficient for this allele. The 4.1B/Dal-1 null mice develop normally and are fertile. Rates of cellular proliferation and apoptosis in brain, mammary, and lung tissues from the 4.1B/Dal-1 null mice were indistinguishable from those seen with wild-type mice. Aging studies indicate that these mice do not have a propensity to develop tumors. Analysis of fibroblasts from these mice demonstrated that the growth characteristics and kinetics of these cells were not different from those of cells from the wild-type mice. These findings indicate that the 4.1B gene is not required for normal development and that 4.1B/Dal-1 does not function as a tumor suppressor gene.


Asunto(s)
Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/fisiología , Predisposición Genética a la Enfermedad , Proteínas de la Membrana/genética , Proteínas de la Membrana/fisiología , Neoplasias/genética , Neoplasias/patología , Alelos , Animales , Apoptosis , Southern Blotting , Western Blotting , Encéfalo/metabolismo , Muerte Celular , Proliferación Celular , Células Cultivadas , Clonación Molecular , Cartilla de ADN/química , Femenino , Genotipo , Inmunohistoquímica , Cinética , Pulmón/metabolismo , Glándulas Mamarias Animales/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Modelos Genéticos , Mutación , Neoplasias/metabolismo , Reacción en Cadena de la Polimerasa
19.
Wound Repair Regen ; 12(3): 384-92, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15225218

RESUMEN

The MRL mouse has been shown to display an epimorphic regenerative response after ear hole punching leading to complete closure within 30 days and cartilage regrowth. The regenerative capacity of the MRL has also been seen after a severe cryoinjury to the heart leads to complete healing without scarring and functional myocardium. The wound healing ear hole closure response that occurs in MRL mice has been shown to be genetically controlled. We have previously identified 11 quantitative trait loci (QTL) that govern healing in an intercross of (MRL x C57BL/6 J) mice. However, it is desirable to use another poorly healing mouse strain to elucidate the full range of genetic factors that affect this important process. In the current study, we have used an inbred subspecies of the mouse, M. castaneus, and have confirmed a number of loci identified previously. In addition, we report three new healing QTL. Furthermore, in this strain combination, we note a strong sexual dimorphism also observed in the MRL x C57BL/6 cross, both in the healing trait and in the QTL that control it.


Asunto(s)
Cartílago Auricular/fisiología , Regeneración/genética , Animales , Cartílago Auricular/lesiones , Ligamiento Genético , Genotipo , Ratones , Ratones Endogámicos , Modelos Animales , Factores Sexuales , Cicatrización de Heridas/genética
20.
Mamm Genome ; 15(1): 53-61, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14727142

RESUMEN

Iddm4 is one of several susceptibility genes that have been identified in the BB rat model of type 1 diabetes. The BB rat allele of this gene confers dominant predisposition to diabetes induction by immune perturbation in both the diabetes-prone and the diabetes-resistant substrains, whereas the Wistar Furth (WF) allele confers resistance. We have positioned the gene in a 2.8-cM region on rat Chromosome (Chr) 4, proximal to Lyp/Ian4l1. We have produced a radiation hybrid map of the Iddm4-region that includes a number of rat genes with their mouse and human orthologs. We present a comparative map of the rat Iddm4 region in rat, human, and mouse, assigning the gene to a 6.3-Mb segment between PTN and ZYX at 7q32 in the human genome, and to a 5.7-Mb segment between Ptn and Zyx in the mouse genome.


Asunto(s)
Proteínas Portadoras/genética , Mapeo Cromosómico , Diabetes Mellitus Tipo 1/genética , Animales , Cromosomas/genética , Diabetes Mellitus Tipo 1/patología , Marcadores Genéticos , Predisposición Genética a la Enfermedad , Genoma , Humanos , Escala de Lod , Ratones , Repeticiones de Microsatélite , Mapeo de Híbrido por Radiación , Ratas , Ratas Endogámicas BB , Ratas Endogámicas WF
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...