Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Cancer Sci ; 108(11): 2273-2280, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28869796

RESUMEN

We previously reported that eribulin mesylate (eribulin), a tubulin-binding drug (TBD), could remodel tumor vasculature (i.e. increase tumor vessels and perfusion) in human breast cancer xenograft models. However, the role of this vascular remodeling in antitumor effects is not fully understood. Here, we investigated the effects of eribulin-induced vascular remodeling on antitumor activities in multiple human cancer xenograft models. Microvessel densities (MVD) were evaluated by immunohistochemistry (CD31 staining), and antitumor effects were examined in 10 human cancer xenograft models. Eribulin significantly increased MVD compared to the controls in six out of 10 models with a correlation between enhanced MVD levels and antitumor effects (R2  = 0.54). Because of increased MVD, we next used radiolabeled liposomes to examine whether eribulin treatment would result in increased tumoral accumulation levels of these macromolecules and, indeed, we found that eribulin, unlike vinorelbine (another TBD) enhanced them. As eribulin increased accumulation of radiolabeled liposomes, we postulated that this treatment might enhance the antitumor effect of Doxil (a liposomal anticancer agent) and facilitate recruitment of immune cells into the tumor. As expected, eribulin enhanced antitumor activity of Doxil in a post-erlotinib treatment H1650 (PE-H1650) xenograft model. Furthermore, infiltrating CD11b-positive immune cells were significantly increased in multiple eribulin-treated xenografted tumors, and natural killer (NK) cell depletion reduced the antitumor effects of eribulin. These findings suggest a contribution of the immune cells for antitumor activities of eribulin. Taken together, our results suggest that vascular remodeling induced by eribulin acts as a microenvironment modulator and, consequently, this alteration enhanced the antitumor effects of eribulin.


Asunto(s)
Furanos/administración & dosificación , Cetonas/administración & dosificación , Neoplasias/tratamiento farmacológico , Microambiente Tumoral/efectos de los fármacos , Remodelación Vascular/efectos de los fármacos , Animales , Doxorrubicina/administración & dosificación , Doxorrubicina/análogos & derivados , Femenino , Células HCT116 , Humanos , Ratones , Neoplasias/patología , Polietilenglicoles/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Nat Commun ; 15(1): 2192, 2024 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-38467634

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) has the worst prognosis of all cancers. To improve PDAC therapy, we establish screening systems based on organoid and co-culture technologies and find a payload of antibody-drug conjugate (ADC), a bromodomain and extra-terminal (BET) protein degrader named EBET. We select CEACAM6/CD66c as an ADC target and developed an antibody, #84.7, with minimal reactivity to CEACAM6-expressing normal cells. EBET-conjugated #84.7 (84-EBET) has lethal effects on various PDAC organoids and bystander efficacy on CEACAM6-negative PDAC cells and cancer-associated fibroblasts. In mouse studies, a single injection of 84-EBET induces marked tumor regression in various PDAC-patient-derived xenografts, with a decrease in the inflammatory phenotype of stromal cells and without significant body weight loss. Combination with standard chemotherapy or PD-1 antibody induces more profound and sustained regression without toxicity enhancement. Our preclinical evidence demonstrates potential efficacy by delivering BET protein degrader to PDAC and its microenvironment via CEACAM6-targeted ADC.


Asunto(s)
Carcinoma Ductal Pancreático , Inmunoconjugados , Neoplasias Pancreáticas , Humanos , Ratones , Animales , Inmunoconjugados/farmacología , Inmunoconjugados/uso terapéutico , Neoplasias Pancreáticas/genética , Carcinoma Ductal Pancreático/genética , Línea Celular Tumoral , Microambiente Tumoral , Antígenos CD , Moléculas de Adhesión Celular , Proteínas Ligadas a GPI
3.
Mol Cancer Ther ; 22(1): 12-24, 2023 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-36279567

RESUMEN

Innate and adaptive resistance to cancer therapies, such as chemotherapies, molecularly targeted therapies, and immune-modulating therapies, is a major issue in clinical practice. Subpopulations of tumor cells expressing the receptor tyrosine kinase AXL become enriched after treatment with antimitotic drugs, causing tumor relapse. Elevated AXL expression is closely associated with drug resistance in clinical samples, suggesting that AXL plays a pivotal role in drug resistance. Although several molecules with AXL inhibitory activity have been developed, none have sufficient activity and selectivity to be clinically effective when administered in combination with a cancer therapy. Here, we report a novel small molecule, ER-851, which is a potent and highly selective AXL inhibitor. To investigate resistance mechanisms and identify driving molecules, we conducted a comprehensive gene expression analysis of chemoresistant tumor cells in mouse xenograft models of genetically engineered human lung cancer and human triple-negative breast cancer. Consistent with the effect of AXL knockdown, cotreatment of ER-851 and antimitotic drugs produced an antitumor effect and prolonged relapse-free survival in the mouse xenograft model of human triple-negative breast cancer. Importantly, when orally administered to BALB/c mice, this compound did not induce retinal toxicity, a known side effect of chronic MER inhibition. Together, these data strongly suggest that AXL is a therapeutic target for overcoming drug resistance and that ER-851 is a promising candidate therapeutic agent for use against AXL-expressing antimitotic-resistant tumors.


Asunto(s)
Antimitóticos , Neoplasias de la Mama Triple Negativas , Humanos , Animales , Ratones , Tirosina Quinasa del Receptor Axl , Antimitóticos/farmacología , Proteínas Proto-Oncogénicas/metabolismo , Resistencia a Antineoplásicos , Línea Celular Tumoral , Inhibidores de Proteínas Quinasas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
4.
J Gen Virol ; 93(Pt 3): 588-593, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22071515

RESUMEN

Extremely low infectivity has hampered direct (cell-free) infection studies of human T-cell leukemia virus type I (HTLV-I). In order to break through this barrier, we examined the susceptibility of many kinds of cells to HTLV-I and found a feline kidney cell line, 8C, that is highly susceptible to HTLV-I and produced remarkable amounts of infectious progeny viruses. Tax1 protein encoded by HTLV-I is known as a transcription activator for viral and cellular genes. We found that the 8C cells expressing the Tax1 protein (8C/TaxWT cells) can produce more progeny viruses than 8C cells when the cells were exposed to cell-free HTLV-I. A large number of syncytia were also induced in these cells. Here, we propose 8C/TaxWT cells as a useful tool to study the cell-free HTLV-I infection.


Asunto(s)
Expresión Génica , Productos del Gen tax/metabolismo , Virus Linfotrópico T Tipo 1 Humano/fisiología , Replicación Viral , Animales , Gatos , Línea Celular , Productos del Gen tax/genética , Virus Linfotrópico T Tipo 1 Humano/crecimiento & desarrollo , Virología/métodos , Cultivo de Virus/métodos
5.
Biochem Biophys Res Commun ; 410(4): 878-84, 2011 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-21703241

RESUMEN

Oncogenic stimuli such as H-Ras induce oncogene-induced senescence (OIS) in fibroblasts to protect against transformation. Here we found that a population of the human diploid fibroblasts can escape from OIS induced by H-RasV12. We designated these OIS-escaped cells as OISEC (OIS-escaped cells). OISEC lost the expression of p16 which plays an important role for cell cycle arrest for induction of senescence, but OISEC preserved the p16 expression machinery and exhibited senescence by the treatment with hydrogen peroxide (H(2)O(2)) as stress-induced premature senescence (SIPS). OISEC did not possess anchorage-independent growth potential, and functional disruption of p53 and Rb by SV40 early region encoding large T and small t antigens, induced the aneuploidy phenotype and colony-forming potential of OISEC together with the exhibition of in vivo tumor formation. Finally, we also found that the distinctive feature of OISEC is expression of transcription factors, Oct3/4, SOX2, and Nanog which is closely related to stem-like cell features. This study highlights the presence of a cell population which escaped from OIS, and this OISEC may transform into malignant cancer cells by the additional hits of several genes in vivo.


Asunto(s)
Transformación Celular Neoplásica/patología , Senescencia Celular , Fibroblastos/patología , Genes ras , Animales , Transformación Celular Neoplásica/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Diploidia , Humanos , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Estrés Fisiológico/genética
6.
Biochem Biophys Res Commun ; 393(2): 201-6, 2010 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-20122898

RESUMEN

MAPK phosphatase-7 (MKP-7) was identified as a JNK-specific phosphatase. However, despite its high specificity for JNK, MKP-7 interacts also with ERK. We previously showed that as a physiological consequence of their interaction, activated ERK phosphorylates MKP-7 at Ser-446, and stabilizing MKP-7. In the present study, we analyzed MKP-7 function in activation of ERK. A time-course experiment showed that both MKP-7 and its phosphatase-dead mutant prolonged mitogen-induced ERK phosphorylation, suggesting that MKP-7 functions as a scaffold for ERK. An important immunohistological finding was that nuclear translocation of phospho-ERK following PMA stimulation was blocked by co-expressed MKP-7 and, moreover, that phospho-ERK co-localized with MKP-7 in the cytoplasm. Reporter gene analysis indicated that MKP-7 blocks ERK-mediated transcription. Overall, our data indicate that MKP-7 down-regulates ERK-dependent gene expression by blocking nuclear accumulation of phospho-ERK.


Asunto(s)
Citoplasma/enzimología , Fosfatasas de Especificidad Dual/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/metabolismo , Activación Transcripcional , Transporte Activo de Núcleo Celular , Animales , Células COS , Núcleo Celular/enzimología , Chlorocebus aethiops , Regulación hacia Abajo , Factor de Crecimiento Epidérmico/farmacología , Humanos , Fosforilación , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo
7.
Cancer Sci ; 100(9): 1675-9, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19555392

RESUMEN

MgcRacGAP plays critical roles in cell division through regulating Rho family small GTPases. As we previously reported, phosphorylation of MgcRacGAP on serine 387 (S387) is induced by Aurora B kinase at the midbody during cytokinesis, which is a critical step of cytokinesis. Phosphorylation of S387-MgcRacGAP converts it from RacGAP to RhoGAP, leading to completion of cytokinesis. Here we show that MgcRacGAP is prominently phosphorylated on S387 even in the interphase of v-Src-transformed NIH3T3 cells in the cytoplasm, but not in the interphase of parental NIH3T3 or H-RasV12-transformed NIH3T3 cells. Interestingly, levels of phosphorylation on S387 (pS387) correlated with soft agar colony-forming abilities of v-Src-transformed NIH3T3 cells. Expression of a phosphorylation-mimic mutant MgcRacGAP-S387D enhanced colony formation of v-Src-transformed NIH3T3 cells. Surprisingly, a Rac1 inhibitor but not kinase inhibitors including Aurora B kinase inhibitor specifically inhibited phosphorylation of S387-MgcRacGAP in v-Src-transformed NIH3T3 cells, suggesting the v-Src-induced pathological positive feedback mechanisms towards Rac1 activation using pS387-MgcRacGAP. These results indicated the difference in the mechanisms between v-Src- and H-RasV12-induced transformation, and should shed some light on pathological roles of disordered phosphorylation of MgcRacGAP at S387 in v-Src-induced cell transformation.


Asunto(s)
Transformación Celular Neoplásica , Proteínas Activadoras de GTPasa/metabolismo , Proteína Oncogénica pp60(v-src)/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Aminoquinolinas/farmacología , Animales , Aurora Quinasa B , Aurora Quinasas , Western Blotting , Ensayo de Unidades Formadoras de Colonias , Proteínas Activadoras de GTPasa/antagonistas & inhibidores , Ratones , Células 3T3 NIH , Proteína Oncogénica pp60(v-src)/genética , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Pirimidinas/farmacología , Proteína de Unión al GTP rac1/genética , Proteína de Unión al GTP rac1/metabolismo
8.
Sci Rep ; 9(1): 8656, 2019 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-31209263

RESUMEN

Despite their outstanding antitumour activity in mice, the limited supply from the natural sources has prevented drug discovery/development based on intact halichondrins. We achieved a total synthesis of C52-halichondrin-B amine (E7130) on a >10 g scale with >99.8% purity under GMP conditions. Interestingly, E7130 not only is a novel microtubule dynamics inhibitor but can also increase intratumoural CD31-positive endothelial cells and reduce α-SMA-positive cancer-associated fibroblasts at pharmacologically relevant compound concentrations. According to these unique effects, E7130 significantly augment the effect of antitumour treatments in mouse models and is currently in a clinical trial. Overall, our work demonstrates that a total synthesis can address the issue of limited material supply in drug discovery/development even for the cases of complex natural products.


Asunto(s)
Antineoplásicos Fitogénicos/síntesis química , Neoplasias de la Mama/tratamiento farmacológico , Carcinoma de Células Escamosas/tratamiento farmacológico , Éteres Cíclicos/síntesis química , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Macrólidos/síntesis química , Moduladores de Tubulina/síntesis química , Actinas/genética , Actinas/metabolismo , Animales , Antineoplásicos Fitogénicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica , Productos Biológicos/síntesis química , Productos Biológicos/farmacología , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Fibroblastos Asociados al Cáncer/efectos de los fármacos , Fibroblastos Asociados al Cáncer/metabolismo , Fibroblastos Asociados al Cáncer/patología , Carcinoma de Células Escamosas/mortalidad , Carcinoma de Células Escamosas/patología , Cetuximab/farmacología , Descubrimiento de Drogas , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales/patología , Éteres Cíclicos/farmacología , Femenino , Expresión Génica/efectos de los fármacos , Neoplasias de Cabeza y Cuello/mortalidad , Neoplasias de Cabeza y Cuello/patología , Humanos , Macrólidos/farmacología , Ratones , Ratones Endogámicos BALB C , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/genética , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Análisis de Supervivencia , Moduladores de Tubulina/farmacología , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Biochem Biophys Res Commun ; 373(3): 392-6, 2008 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-18570889

RESUMEN

The AKT pathway is frequently activated in glioblastoma, and as such, inhibitors of this pathway could prove very useful as anti-glioblastoma therapies. Here we established immortalized astrocytes expressing Renilla luciferase as well as those expressing both an active form of AKT and firefly luciferase. Since both luciferase activities represent the numbers of corresponding cell lines, novel inhibitors of the AKT pathway can be identified by treating co-cultures containing the two types of luciferase-expressing cells with individual compounds. Indeed, such a screening system succeeded in identifying fumitremorgin C as an efficient inhibitor of the AKT pathway, which was further confirmed by the ability of fumitremorgin C to selectively inhibit the growth of immortalized astrocytes expressing an active form of AKT. The present study proposes a broadly applicable approach for identifying therapeutic agents that target the pathways and/or molecules responsible for cancer development.


Asunto(s)
Antineoplásicos/aislamiento & purificación , Astrocitos/enzimología , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales/métodos , Inhibidores de Proteínas Quinasas/aislamiento & purificación , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Humanos , Indoles/farmacología , Luciferasas de Luciérnaga/genética , Luciferasas de Renilla/genética , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/biosíntesis , Proteínas Proto-Oncogénicas c-akt/genética
10.
Methods Mol Biol ; 1763: 153-163, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29476497

RESUMEN

Most of preclinical cancer studies use xenograft models established from human cell lines or patient-derived cancer cells subcutaneously implanted into the flank of immunocompromised mice. These models are often assumed to represent the original diseases and are valuable tools, at least to some extent, for understanding both the basic biology of cancer and for proof-of-concept studies of molecularly targeted therapies. However, analyzing the cellular behavior of individual components within xenografts, including tumor cells, stromal cells, immune cells, and blood vessels, is challenging. In particular, it has been difficult and urgently required to trace the whole process of heterogeneous tumor microenvironment formation mediated by various components described above. Here we demonstrate a method for monitoring this process using a window device system that we have recently developed and a subcutaneous xenograft model that accurately recapitulates the histology of human lung adenocarcinoma. Use of our imaging window device and a multiphoton laser scanning microscope provides a powerful tool for investigating tumor heterogeneity and responses to drug treatments in an in vivo live imaging system.


Asunto(s)
Adenocarcinoma/patología , Neoplasias Pulmonares/patología , Imagen Molecular/instrumentación , Animales , Femenino , Humanos , Inyecciones Subcutáneas , Ratones , Ratones Desnudos , Ratones SCID , Células Tumorales Cultivadas , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Mol Cancer ; 6: 36, 2007 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-17547775

RESUMEN

BACKGROUND: A novel alkylating agent, temozolomide, has proven efficacious in the treatment of malignant gliomas. However, expression of O6-methylguanine-DNA methyltransferase (MGMT) renders glioma cells resistant to the treatment, indicating that identification of mechanisms underlying the gene regulation of MGMT is highly required. Although glioma-derived cell lines have been widely employed to understand such mechanisms, those models harbor numerous unidentified genetic lesions specific for individual cell lines, which complicates the study of specific molecules and pathways. RESULTS: We established glioma models by transforming normal human astrocyte cells via retroviral-mediated gene transfer of defined genetic elements and found that MGMT was downregulated in the transformed cells. Interestingly, inhibitors of DNA methylation and histone deacetylation failed to increase MGMT protein levels in the transformed astrocyte cells as well as cultured glioblastoma cell lines, whereas the treatment partially restored mRNA levels. These observations suggest that downregulation of MGMT may depend largely on cellular factors other than promoter-hypermethylation of MGMT genes, which is being used in the clinic to nominate patients for temozolomide treatment. Furthermore, we discovered that Valproic acid, one of histone deacetylase inhibitors, suppressed growth of the transformed astrocyte cells without increasing MGMT protein, suggesting that such epigenetic compounds may be used to some types of gliomas in combination with alkylating agents. CONCLUSION: Normal human astrocyte cells allow us to generate experimental models of human gliomas by direct manipulation with defined genetic elements, in contrast to tumor-derived cell lines which harbor numerous unknown genetic abnormalities. Thus, we propose that the study using the transformed astrocyte cells would be useful for identifying the mechanisms underlying MGMT regulation in tumor and for the development of rational drug combination in glioma therapies.


Asunto(s)
Astrocitos/enzimología , Metilasas de Modificación del ADN/genética , Enzimas Reparadoras del ADN/genética , Glioma/tratamiento farmacológico , Glioma/enzimología , Proteínas Supresoras de Tumor/genética , Células 3T3 , Animales , Antineoplásicos/uso terapéutico , Astrocitos/citología , Astrocitos/efectos de los fármacos , División Celular , Metilasas de Modificación del ADN/metabolismo , Enzimas Reparadoras del ADN/metabolismo , Dacarbazina/análogos & derivados , Dacarbazina/uso terapéutico , Humanos , Ratones , Proteínas Recombinantes/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Temozolomida , Transfección , Trasplante Heterólogo , Proteínas Supresoras de Tumor/metabolismo , Ácido Valproico/farmacología
12.
Mol Cell Biol ; 22(20): 7015-23, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12242282

RESUMEN

v-Crk, an oncogene product of avian sarcoma virus CT10, efficiently transforms chicken embryo fibroblasts (CEF). We have recently reported that constitutive activation of the phosphoinositide 3-kinase (PI3K)/AKT pathway plays a critical role in the v-Crk-induced transformation of CEF. In the present study we investigated the molecular mechanism by which v-Crk activates the PI3K/AKT pathway. First, we found that v-Crk promotes the association of the p85 regulatory subunit of PI3K with focal adhesion kinase (FAK) by inducing the phosphorylation of the Y397 residue in FAK. This FAK phosphorylation needs activation of the Src family tyrosine kinase(s) for which the v-Crk SH2 domain is responsible. v-Crk was unable to activate the PI3K/AKT pathway in FAK-null cells, indicating the functional importance of FAK. In addition, we found that H-Ras is also required for the activation of the PI3K/AKT pathway. The v-Crk-induced activation of AKT was greatly enhanced by the overexpression of H-Ras or its guanine nucleotide exchange factor mSOS, which binds to the v-Crk SH3 domain, whereas a dominant-negative mutant of H-Ras almost completely suppressed this activation. Furthermore, we showed that v-Crk stimulates the interaction of H-Ras with the Ras binding domain in the PI3K p110 catalytic subunit. Our data indicated that the v-Crk-induced activation of PI3K/AKT pathway was cooperatively achieved by two distinct interactions. One is the interaction of p85 with tyrosine-phosphorylated FAK promoted by the v-Crk SH2 domain, and another is the interaction of p110 with H-Ras dictated by the v-Crk SH3 domain.


Asunto(s)
Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Proteínas Proto-Oncogénicas , Proteínas Oncogénicas de Retroviridae/metabolismo , Transducción de Señal , Animales , Virus del Sarcoma Aviar/metabolismo , Embrión de Pollo , Activación Enzimática , Quinasa 1 de Adhesión Focal , Proteína-Tirosina Quinasas de Adhesión Focal , Humanos , Proteína Oncogénica v-crk , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Proteínas Tirosina Quinasas/genética , Proteínas Proto-Oncogénicas c-akt , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Oncogénicas de Retroviridae/genética , Transformación Genética , Tirosina/metabolismo , Familia-src Quinasas/genética , Familia-src Quinasas/metabolismo
13.
Mol Cell Biol ; 23(17): 6139-49, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12917336

RESUMEN

PTEN is a tumor suppressor frequently inactivated in brain, prostate, and uterine cancers that acts as a phosphatase on phosphatidylinositol-3,4,5-trisphosphate, antagonizing the activity of the phosphatidylinositol 3'-OH kinase. PTEN manifests its tumor suppressor function in most tumor cells by inducing G(1)-phase cell cycle arrest. To study the mechanism of cell cycle arrest, we established a tetracycline-inducible expression system for PTEN in cell lines lacking this gene. Expression of wild-type PTEN but not of mutant forms unable to dephosphorylate phosphoinositides reduced the expression of cyclin D1. Cyclin D1 reduction was accompanied by a marked decrease in endogenous retinoblastoma (Rb) protein phosphorylation on cyclin D/CDK4-specific sites, showing an early negative effect of PTEN on Rb inactivation. PTEN expression also prevented cyclin D1 from localizing to the nucleus during the G(1)- to S-phase cell cycle transition. The PTEN-induced localization defect and the cell growth arrest could be rescued by the expression of a nucleus-persistent mutant form of cyclin D1, indicating that an important effect of PTEN is at the level of nuclear availability of cyclin D1. Constitutively active Akt/PKB kinase counteracted the effect of PTEN on cyclin D1 translocation. The data are consistent with an oncogenesis model in which a lack of PTEN fuels the cell cycle by increasing the nuclear availability of cyclin D1 through the Akt/PKB pathway.


Asunto(s)
Ciclo Celular/fisiología , Núcleo Celular/metabolismo , Ciclina D1/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Proteínas Serina-Treonina Quinasas , Proteínas Supresoras de Tumor/metabolismo , Transporte Activo de Núcleo Celular/fisiología , Ciclo Celular/efectos de los fármacos , División Celular/genética , Células Cultivadas , Cromonas/farmacología , Ciclina D1/efectos de los fármacos , Quinasa 4 Dependiente de la Ciclina , Quinasas Ciclina-Dependientes/metabolismo , Inhibidores Enzimáticos/farmacología , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/metabolismo , Humanos , Cloruro de Litio/farmacología , Morfolinas/farmacología , Mutación , Fosfohidrolasa PTEN , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosfoproteínas Fosfatasas/metabolismo , Monoéster Fosfórico Hidrolasas/genética , Fosforilación , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Proteína de Retinoblastoma/metabolismo , Proteínas Supresoras de Tumor/genética
14.
Cancer Res ; 77(21): 6001-6010, 2017 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-28882998

RESUMEN

Immune surveillance is a critical component of the antitumor response in vivo, yet the specific components of the immune system involved in this regulatory response remain unclear. In this study, we demonstrate that autoantibodies can mitigate tumor growth in vitro and in vivo We generated two cancer cell lines, embryonal carcinoma and glioblastoma cell lines, from monkey-induced pluripotent stem cells (iPSC) carrying a homozygous haplotype of major histocompatibility complex (MHC, Mafa in Macaca fascicularis). To establish a monkey cancer model, we transplanted these cells into monkeys carrying the matched Mafa haplotype in one of the chromosomes. Neither Mafa-homozygous cancer cell line grew in monkeys carrying the matched Mafa haplotype heterozygously. We detected in the plasma of these monkeys an IgG autoantibody against GRP94, a heat shock protein. Injection of the plasma prevented growth of the tumor cells in immunodeficient mice, whereas plasma IgG depleted of GRP94 IgG exhibited reduced killing activity against cancer cells in vitro These results indicate that humoral immunity, including autoantibodies against GRP94, plays a role in cancer immune surveillance. Cancer Res; 77(21); 6001-10. ©2017 AACR.


Asunto(s)
Autoanticuerpos/inmunología , Proteínas HSP70 de Choque Térmico/inmunología , Células Madre Pluripotentes Inducidas/inmunología , Complejo Mayor de Histocompatibilidad/inmunología , Proteínas de la Membrana/inmunología , Neoplasias/inmunología , Animales , Autoanticuerpos/sangre , Autoanticuerpos/metabolismo , Carcinoma Embrionario/genética , Carcinoma Embrionario/inmunología , Carcinoma Embrionario/patología , Línea Celular Tumoral , Células Cultivadas , Femenino , Glioblastoma/genética , Glioblastoma/inmunología , Glioblastoma/patología , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Haplotipos , Homocigoto , Células Madre Pluripotentes Inducidas/metabolismo , Macaca fascicularis , Complejo Mayor de Histocompatibilidad/genética , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Trasplante de Neoplasias/métodos , Neoplasias/genética , Neoplasias/patología
15.
PLoS One ; 12(4): e0175147, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28380052

RESUMEN

It has been proposed that a subpopulation of tumour cells with stem cell-like characteristics, known as cancer stem cells (CSCs), drives tumour initiation and generates tumour heterogeneity, thus leading to cancer metastasis, recurrence, and drug resistance. Although there has been substantial progress in CSC research into many solid tumour types, an understanding of the biology of CSCs in lung cancer remains elusive, mainly because of their heterogeneous origins and high plasticity. Here, we demonstrate that engineered lung cancer cells derived from normal human airway basal epithelial cells possessed CSC-like characteristics in terms of multilineage differentiation potential and strong tumour-initiating ability. Moreover, we established an in vitro 3D culture system that allowed the in vivo differentiation process of the CSC-like cells to be recapitulated. This engineered CSC model provides valuable opportunities for studying the biology of CSCs and for exploring and evaluating novel therapeutic approaches and targets in lung CSCs.


Asunto(s)
Ingeniería Celular/métodos , Pulmón/citología , Células Madre Neoplásicas/fisiología , Mucosa Respiratoria/citología , Animales , Transformación Celular Neoplásica/metabolismo , Células Cultivadas , Femenino , Humanos , Pulmón/patología , Neoplasias Pulmonares/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Trasplante de Neoplasias
16.
Oncogene ; 24(36): 5648-55, 2005 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-16007212

RESUMEN

Ras-induced transformation is characterized not only by uncontrolled proliferation but also by drastic morphological changes accompanied by the disruption of the actin cytoskeleton. Previously, we reported that human fibroblasts are more resistant than rodent fibroblasts to Ras-induced transformation. To explore the molecular basis for the difference in susceptibility to Ras-induced transformation, we investigated the effect of activated H-Ras on the actin cytoskeleton in human diploid fibroblasts and in rat embryo fibroblasts, both of which are immortalized by SV40 early region. We demonstrate here that Ras-induced morphological changes, decreased expression of tropomyosin isoforms, and suppression of the ROCK/LIMK/Cofilin pathway observed in the rat fibroblasts were not detected in the human fibroblasts even with high expression levels of Ras. We also show that activation of the MEK/ERK pathway sufficed to induce all of these alterations in the rat fibroblasts, whereas the human fibroblasts were refractory to these MEK/ERK-mediated changes. In addition to morphological changes, we demonstrated that the expression of activated Ras induced an invasive phenotype in the rat, but not in the human fibroblasts. These studies provide evidence for the existence of human-specific mechanisms that resist Ras/MEK/ERK-mediated transformation.


Asunto(s)
Actinas/metabolismo , Citoesqueleto/metabolismo , Diploidia , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Invasividad Neoplásica/patología , Proteínas ras/metabolismo , Factores Despolimerizantes de la Actina , Animales , Células Cultivadas , Regulación hacia Abajo , Matriz Extracelular/metabolismo , Fibroblastos , Humanos , Sistema de Señalización de MAP Quinasas , Proteínas de Microfilamentos/metabolismo , Peso Molecular , Fosforilación , Isoformas de Proteínas/metabolismo , Ratas , Tropomiosina/metabolismo , Proteínas ras/genética
17.
Oncogene ; 24(54): 7984-90, 2005 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-16103879

RESUMEN

Oncogenic protein provokes cell cycle arrest termed premature senescence. In this process Ras has been known to induce cyclin-dependent kinase inhibitor (CKI) p16(INK4A) in primary fibroblasts. Here, we present a novel finding that human chimeric oncoprotein SYT-SSX1 induces CKI p21(WAF1/CIP1) (p21) for suppression of cell growth. In human synovial sarcoma cell lines, the expression levels of p21 were high and the transcriptional activity of the p21 gene promoter was significantly elevated. The transient expression of SYT-SSX1-induced activation of the p21 gene promoter in human diploid fibroblasts. The N-terminus deletion form of SYT-SSX1, which failed to bind to hBRM one of the chromatin remodeling factors, preserved the p21 induction ability. This effect of SYT-SSX1 was similar in extent in both wild-type and p53-deficient HCT116 cell lines. Furthermore, the introduction of mutation in Sp1/Sp3 binding sites of the p21 gene promoter abolished the SYT-SSX1-induced transcriptional activity of its promoter. In SW13 cells, the stable expression of SYT-SSX1 suppressed cell growth in culture. These results suggest that SYT-SSX1 is able to induce p21 in a manner independent on hBRM and p53 but dependent on Sp1/Sp3.


Asunto(s)
Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Regulación Neoplásica de la Expresión Génica , Sarcoma Sinovial/genética , Animales , Células COS , Línea Celular , Línea Celular Tumoral , Chlorocebus aethiops , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Genes Reporteros , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Humanos , Immunoblotting , Luciferasas/metabolismo , Ratones , Microscopía Confocal , Células 3T3 NIH , Proteínas de Fusión Oncogénica/metabolismo , Regiones Promotoras Genéticas , Proteínas Recombinantes de Fusión/metabolismo , Sarcoma Sinovial/patología
18.
Oncogene ; 22(38): 5946-57, 2003 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-12955073

RESUMEN

CrkII belongs to the adaptor protein family that plays a crucial role in signal transduction. In order to better understand the biological functions of CrkII, we focused on the regulation of gene expression by CrkII. Various transcriptional control elements were examined for their activation by CrkII-expression, and we found that CrkII selectively activates the serum response element (SRE), a transcriptional control element of immediate-early genes. This SRE activation induced by CrkII-overexpression was mediated by the serum response factor (SRF) via Rho. Indeed, we confirmed that the amount of activated Rho was increased in the CrkII-expressing cells. Moreover, we showed that when overexpressed, CrkII induces the cellular transformation of NIH 3T3 cells and that a dominant negative mutant of Rho suppresses this transformation, strongly suggesting that activation of Rho is essential for the transforming activity by CrkII. Furthermore, we also found that CrkII and Galpha12, a member of the heterotrimeric G proteins, synergistically activates Rho as well as the SRF, and that an SH3 mutant of CrkII can inhibit the Galpha12-induced activation of SRF. These results strongly suggest that CrkII is involved in the activation of Rho and SRF by Galpha12. Our study provides strong evidence that Rho activation plays a crucial role in CrkII-mediated signals to induce gene expression and cellular transformation.


Asunto(s)
Transformación Celular Neoplásica , Proteínas Proto-Oncogénicas/metabolismo , Elemento de Respuesta al Suero/fisiología , Factor de Respuesta Sérica/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Células 3T3/metabolismo , Células 3T3/patología , Animales , Subunidades alfa de la Proteína de Unión al GTP G12-G13 , Regulación de la Expresión Génica , Genes Reporteros , Proteínas de Unión al GTP Heterotriméricas/genética , Proteínas de Unión al GTP Heterotriméricas/metabolismo , Ratones , Mutación , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-crk , Factor de Respuesta Sérica/genética , Transducción de Señal , Activación Transcripcional , Proteínas de Unión al GTP rho/genética
19.
Oncogene ; 23(52): 8527-34, 2004 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-15378021

RESUMEN

The c-abl tyrosine kinase is the proto-oncogene of the v-abl oncogene of the Abelson murine leukemia virus. Although mutational variants of c-Abl can exhibit gain of function and can produce a transformed phenotype, the function of c-Abl in transformation remained unclear. Here, we report that the loss of c-abl facilitates transformation. c-abl-knockout mouse embryonic fibroblasts (MEFs) immortalized by SV40 T antigen acquired anchorage-independent growth, and by constructing mutational variants of T antigen we showed that binding of large T antigen to p53 and RB was necessary to induce anchorage-independent growth. Although c-abl/p53 double-knockout MEFs did not undergo anchorage-independent growth, those expressing human papilloma virus 16 E7, which mainly inactivates RB, did. Our results show that the loss of c-abl facilitates anchorage-independent growth in the context of p53 and RB deficiency, and suggest that loss of function of c-abl facilitates some types of transformation.


Asunto(s)
Fibroblastos/metabolismo , Proteínas Proto-Oncogénicas c-abl/deficiencia , Proteína de Retinoblastoma/deficiencia , Proteína p53 Supresora de Tumor/deficiencia , Animales , Antígenos Transformadores de Poliomavirus/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Ratones , Proteínas Oncogénicas Virales/metabolismo , Proteínas E7 de Papillomavirus , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas c-abl/genética , Proteínas Proto-Oncogénicas c-abl/metabolismo
20.
Trends Mol Med ; 10(11): 542-8, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15519280

RESUMEN

Long-standing difficulties in the in vitro transformation of human cells have been overcome. Using telomerase, several successful oncogene-mediated transformations of human cells have been reported and the following cellular requirements for human cell transformation have been proposed: the maintenance of telomere sequences, the inactivation of Rb and p53 pathways, the perturbation of protein phosphatase 2A (PP2A) and the expression of activated Ras. Even when all of these requirements are fulfilled, however, the transformed phenotypes of human cells seem to be much less malignant than those of rodent cells meeting the same requirements. This suggests the existence of undefined cell-autonomous mechanisms that render human cells resistant to malignant transformation.


Asunto(s)
Transformación Celular Neoplásica/genética , Células Cultivadas , Modelos Animales , Proteínas Oncogénicas/fisiología , Roedores , Animales , Línea Celular , Senescencia Celular/genética , Silenciador del Gen , Genes Supresores de Tumor , Humanos , Mitógenos/farmacología , Roedores/genética , Transducción de Señal/genética , Telómero/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA