Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.354
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 296: 100285, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33450231

RESUMEN

DNA methylation regulates gene transcription and is involved in various physiological processes in mammals, including development and hematopoiesis. It is catalyzed by DNA methyltransferases including Dnmt1, Dnmt3a, and Dnmt3b. For Dnmt3b, its effects on transcription can result from its own DNA methylase activity, the recruitment of other Dnmts to mediate methylation, or transcription repression in a methylation-independent manner. Low-frequency mutations in human DNMT3B are found in hematologic malignancies including cutaneous T-cell lymphomas, hairy cell leukemia, and diffuse large B-cell lymphomas. Moreover, Dnmt3b is a tumor suppressor in oncogene-driven lymphoid and myeloid malignancies in mice. However, it is poorly understood how the different Dnmt3b activities contribute to these outcomes. We modulated Dnmt3b activity in vivo by generating Dnmt3b+/- mice expressing one wild-type allele as well as Dnmt3b+/CI and Dnmt3bCI/CI mice where one or both alleles express catalytically inactive Dnmt3bCI. We show that 43% of Dnmt3b+/- mice developed T-cell lymphomas, chronic lymphocytic leukemia, and myeloproliferation over 18 months, thus resembling phenotypes previously observed in Dnmt3a+/- mice, possibly through regulation of shared target genes. Interestingly, Dnmt3b+/CI and Dnmt3bCI/CI mice survived postnatal development and were affected by B-cell rather than T-cell malignancies with decreased penetrance. Genome-wide hypomethylation, increased expression of oncogenes such as Jdp2, STAT1, and Trip13, and p53 downregulation were major events contributing to Dnmt3b+/- lymphoma development. We conclude that Dnmt3b catalytic activity is critical to prevent B-cell transformation in vivo, whereas accessory and methylation-independent repressive functions are important to prevent T-cell transformation.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/genética , Leucemia Linfocítica Crónica de Células B/genética , Linfoma de Células B/genética , Linfoma de Células T/genética , Trastornos Mieloproliferativos/genética , Neoplasias Experimentales/genética , ATPasas Asociadas con Actividades Celulares Diversas/genética , ATPasas Asociadas con Actividades Celulares Diversas/metabolismo , Animales , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , ADN (Citosina-5-)-Metiltransferasas/deficiencia , Metilación de ADN , ADN de Neoplasias/genética , ADN de Neoplasias/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Heterocigoto , Homocigoto , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Leucemia Linfocítica Crónica de Células B/enzimología , Leucemia Linfocítica Crónica de Células B/patología , Linfoma de Células B/enzimología , Linfoma de Células B/patología , Linfoma de Células T/enzimología , Linfoma de Células T/patología , Masculino , Ratones , Ratones Noqueados , Trastornos Mieloproliferativos/enzimología , Trastornos Mieloproliferativos/patología , Neoplasias Experimentales/enzimología , Neoplasias Experimentales/patología , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , ADN Metiltransferasa 3B
2.
J Biol Chem ; 295(28): 9663-9675, 2020 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-32467231

RESUMEN

Acute myeloid leukemia (AML) with mixed lineage leukemia 1 (MLL1) gene rearrangement is characterized by increased expression of a set of homeodomain transcription factors, including homeobox A9 (HOXA9) and HOXA10. The target genes for these regulators include fibroblast growth factor 2 (FGF2) and Ariadne RBR E3 ubiquitin ligase 2 (ARIH2). FGF2 induces leukemia stem cell expansion in MLL1-rearranged AML. ARIH2 encodes TRIAD1, an E3 ubiquitin ligase required for termination of emergency granulopoiesis and leukemia suppressor function in MLL1-rearranged AML. Receptor tyrosine kinases (RTKs), including the FGF receptor, are TRIAD1 substrates that are possibly relevant to these activities. Using transcriptome analysis, we found increased activity of innate immune response pathways and RTK signaling in bone marrow progenitors from mice with MLL1-rearranged AML. We hypothesized that sustained RTK signaling, because of decreased TRIAD1 activity, impairs termination of emergency granulopoiesis during the innate immune response and contributes to leukemogenesis in this AML subtype. Consistent with this, we found aberrantly sustained emergency granulopoiesis in a murine model of MLL1-rearranged AML, associated with accelerated leukemogenesis. Treating these mice with an inhibitor of TRIAD1-substrate RTKs terminated emergency granulopoiesis, delayed leukemogenesis during emergency granulopoiesis, and normalized innate immune responses when combined with chemotherapy. Emergency granulopoiesis also hastened postchemotherapy relapse in mice with MLL1-rearranged AML, but remission was sustained by ongoing RTK inhibition. Our findings suggest that the physiological stress of infectious challenges may drive AML progression in molecularly defined subsets and identify RTK inhibition as a potential therapeutic approach to counteract this process.


Asunto(s)
Reordenamiento Génico , N-Metiltransferasa de Histona-Lisina/metabolismo , Leucemia Mieloide Aguda/enzimología , Leucopoyesis , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Neoplasias Experimentales/enzimología , Animales , Receptores ErbB/genética , Receptores ErbB/metabolismo , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Ratones , Proteína de la Leucemia Mieloide-Linfoide/genética , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Células Madre Neoplásicas/patología , Recurrencia , Transducción de Señal/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
3.
Small ; 17(7): e2007177, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33502119

RESUMEN

Probing the dynamics and quantifying the activities of intracellular protein kinases that coordinate cell growth and division and constitute biomarkers and pharmacological targets in hyperproliferative and pathological disorders remain a challenging task. Here engineering and characterization of a nanobiosensor of the mitotic kinase CDK1, through multifunctionalization of carbon nanotubes with a CDK1-specific fluorescent peptide reporter, are described. This original reporter of CDK1 activity combines the sensitivity of a fluorescent biosensor with the unique physico-chemical and biological properties of nanotubes for multifunctionalization and efficient intracellular penetration. The functional versatility of this nanobiosensor enables implementation to quantify CDK1 activity in a sensitive and dose-dependent fashion in complex biological environments in vitro, to monitor endogenous kinase in living cells and directly within tumor xenografts in mice by fluorescence imaging, thanks to a ratiometric quantification strategy accounting for response relative to concentration in space and in time.


Asunto(s)
Proteína Quinasa CDC2 , Nanotubos de Carbono , Neoplasias Experimentales/enzimología , Animales , Proteína Quinasa CDC2/metabolismo , Línea Celular Tumoral , Humanos , Ratones , Fosforilación
4.
J Biochem Mol Toxicol ; 35(9): e22838, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34273909

RESUMEN

Colorectal cancer (CRC) is the third most common type of cancer. Here, we studied the inhibitory effect of IRAK1 and IRAK4 as a preventive strategy using a colitis-induced tumorigenesis mouse model. CRC clinical data were obtained from the Gene Expression Omnibus (GEO). An experimental inflammation-dependent CRC model was induced by treatment with azoxymethane (AOM) and then dextran sodium sulfate (DSS) in C57BL/6 mice. Mice were administered an IRAK1/4 inhibitor by intraperitoneal injection at 3 mg/kg twice each week for 9 weeks. The IRAK1/4 inhibitor attenuated histological changes and prevented tumor growth. Tumor-associated proteins, including p65 and Ki-67, were downregulated by the IRAK1/4 inhibitor in AOM/DSS-treated mice. Additionally, IRAK1/4 inhibitor administration effectively decreased the expression of inflammatory cytokines. Furthermore, we observed that IRAK1/4 inhibitor treatment attenuated colitis-induced tumorigenesis by inhibiting epithelial-mesenchymal transition. These observations indicate that inhibition of IRAK1 and IRAK4 may suppress experimental colitis-induced tumorigenesis by inhibiting inflammatory responses and epithelial-mesenchymal transition.


Asunto(s)
Carcinogénesis/efectos de los fármacos , Neoplasias Asociadas a Colitis/tratamiento farmacológico , Colitis/tratamiento farmacológico , Transición Epitelial-Mesenquimal/efectos de los fármacos , Quinasas Asociadas a Receptores de Interleucina-1/antagonistas & inhibidores , Proteínas de Neoplasias/antagonistas & inhibidores , Neoplasias Experimentales/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Animales , Carcinogénesis/inducido químicamente , Carcinogénesis/metabolismo , Colitis/inducido químicamente , Colitis/enzimología , Neoplasias Asociadas a Colitis/inducido químicamente , Neoplasias Asociadas a Colitis/enzimología , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Inflamación/enzimología , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Masculino , Ratones , Proteínas de Neoplasias/metabolismo , Neoplasias Experimentales/inducido químicamente , Neoplasias Experimentales/enzimología
5.
Int J Mol Sci ; 22(9)2021 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-33946854

RESUMEN

Hexokinases are a family of ubiquitous exose-phosphorylating enzymes that prime glucose for intracellular utilization. Hexokinase 2 (HK2) is the most active isozyme of the family, mainly expressed in insulin-sensitive tissues. HK2 induction in most neoplastic cells contributes to their metabolic rewiring towards aerobic glycolysis, and its genetic ablation inhibits malignant growth in mouse models. HK2 can dock to mitochondria, where it performs additional functions in autophagy regulation and cell death inhibition that are independent of its enzymatic activity. The recent definition of HK2 localization to contact points between mitochondria and endoplasmic reticulum called Mitochondria Associated Membranes (MAMs) has unveiled a novel HK2 role in regulating intracellular Ca2+ fluxes. Here, we propose that HK2 localization in MAMs of tumor cells is key in sustaining neoplastic progression, as it acts as an intersection node between metabolic and survival pathways. Disrupting these functions by targeting HK2 subcellular localization can constitute a promising anti-tumor strategy.


Asunto(s)
Hexoquinasa/fisiología , Proteínas de Neoplasias/fisiología , Neoplasias/enzimología , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Apoptosis/fisiología , Autofagia/fisiología , Señalización del Calcio/fisiología , Hipoxia de la Célula , Péptidos de Penetración Celular/uso terapéutico , Inducción Enzimática , Regulación Neoplásica de la Expresión Génica , Glucólisis/fisiología , Hexoquinasa/antagonistas & inhibidores , Humanos , Membranas Intracelulares/enzimología , Ratones , MicroARNs/genética , Mitocondrias/metabolismo , Terapia Molecular Dirigida , Proteínas de Neoplasias/antagonistas & inhibidores , Neoplasias/terapia , Neoplasias Experimentales/enzimología , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Procesamiento Proteico-Postraduccional , Ratas , Ubiquitinación
6.
Molecules ; 26(6)2021 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-33801057

RESUMEN

A cannabinoid anticancer para-quinone, HU-331, which was synthesized by our group five decades ago, was shown to have very high efficacy against human cancer cell lines in-vitro and against in-vivo grafts of human tumors in nude mice. The main mechanism was topoisomerase IIα catalytic inhibition. Later, several groups synthesized related compounds. In the present presentation, we review the publications on compounds synthesized on the basis of HU-331, summarize their published activities and mechanisms of action and report the synthesis and action of novel quinones, thus expanding the structure-activity relationship in these series.


Asunto(s)
Cannabidiol/análogos & derivados , Proteínas de Neoplasias/antagonistas & inhibidores , Neoplasias Experimentales , Proteínas de Unión a Poli-ADP-Ribosa/antagonistas & inhibidores , Quinonas , Inhibidores de Topoisomerasa II , Animales , Cannabidiol/química , Cannabidiol/uso terapéutico , ADN-Topoisomerasas de Tipo II/metabolismo , Humanos , Ratones , Ratones Desnudos , Proteínas de Neoplasias/metabolismo , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/enzimología , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Quinonas/química , Quinonas/uso terapéutico , Inhibidores de Topoisomerasa II/química , Inhibidores de Topoisomerasa II/uso terapéutico
7.
Br J Haematol ; 190(6): 877-890, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32232850

RESUMEN

Future progress in the treatment of multiple myeloma (MM) requires both the characterisation of key drivers of the disease and novel, innovative approaches to tackle these vulnerabilities. The present study focussed on the pre-clinical evaluation of a novel drug class, BMI-1 modulators, in MM. We demonstrate potent activity of PTC-028 and PTC596 in a comprehensive set of in vitro and in vivo models, including models of drug resistance and stromal support. Treatment of MM cells with PTC-028 and PTC596 downregulated BMI-1 protein levels, which was found to correlate with drug activity. Surprisingly, BMI-1 was dispensable for the activity of BMI-1 modulators and MM cell growth. Our data rather point to mitotic arrest accompanied by myeloid cell leukaemia-1 (MCL-1) loss as key anti-MM mechanisms and reveal impaired MYC and AKT signalling activity due to BMI-1 modulator treatment. Moreover, we observed a complete eradication of MM after PTC596 treatment in the 5TGM.1 in vivo model and define epigenetic compounds and B cell leukaemia/lymphoma 2 homology domain 3 (BH3) mimetics as promising combination partners. These results bring into question the postulated role of BMI-1 as an essential MM gene and confirm BMI-1 modulators as potent anti-mitotic agents with encouraging pre-clinical activity that supports their rapid translation into clinical trials.


Asunto(s)
Antineoplásicos/farmacología , Bencimidazoles/farmacología , Mitosis/efectos de los fármacos , Mieloma Múltiple , Proteínas de Neoplasias/antagonistas & inhibidores , Neoplasias Experimentales , Complejo Represivo Polycomb 1/antagonistas & inhibidores , Pirazinas/farmacología , Animales , Femenino , Humanos , Masculino , Ratones , Mieloma Múltiple/dietoterapia , Mieloma Múltiple/enzimología , Mieloma Múltiple/patología , Proteínas de Neoplasias/metabolismo , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/enzimología , Neoplasias Experimentales/patología , Complejo Represivo Polycomb 1/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
8.
PLoS Comput Biol ; 15(4): e1006878, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-31026276

RESUMEN

Drosophila provides an inexpensive and quantitative platform for measuring whole animal drug response. A complementary approach is virtual screening, where chemical libraries can be efficiently screened against protein target(s). Here, we present a unique discovery platform integrating structure-based modeling with Drosophila biology and organic synthesis. We demonstrate this platform by developing chemicals targeting a Drosophila model of Medullary Thyroid Cancer (MTC) characterized by a transformation network activated by oncogenic dRetM955T. Structural models for kinases relevant to MTC were generated for virtual screening to identify unique preliminary hits that suppressed dRetM955T-induced transformation. We then combined features from our hits with those of known inhibitors to create a 'hybrid' molecule with improved suppression of dRetM955T transformation. Our platform provides a framework to efficiently explore novel kinase inhibitors outside of explored inhibitor chemical space that are effective in inhibiting cancer networks while minimizing whole body toxicity.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma Neuroendocrino , Evaluación Preclínica de Medicamentos/métodos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas , Neoplasias de la Tiroides , Animales , Carcinoma Neuroendocrino/enzimología , Carcinoma Neuroendocrino/metabolismo , Biología Computacional/métodos , Drosophila , Modelos Biológicos , Neoplasias Experimentales/enzimología , Neoplasias Experimentales/metabolismo , Proteínas Quinasas/efectos de los fármacos , Proteínas Quinasas/metabolismo , Neoplasias de la Tiroides/enzimología , Neoplasias de la Tiroides/metabolismo
9.
Molecules ; 25(12)2020 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-32560527

RESUMEN

Natural compounds of various origins are intensively investigated for their antitumor activity. Potential benefits of antitumor therapy can be achieved when cytotoxic agents kill cancer cells and these dying cancer cells drive adoptive immunity to the tumor. This strategy was successfully demonstrated for chemotherapeutic drugs that induce immunogenic type of cell death (ICD) with release of DAMPs (danger associated molecular patterns) and exposure of "eat me" signals. In this study, we demonstrated that recombinant human milk peptide lactaptin (RL2) induces death of cancer cells with ICD hallmarks in vitro with the release of ATP and high-mobility group box 1 protein (HMGB1) and exposure of calreticulin and HSP70 on the external cell membrane. RL2-treated cancer cells were efficiently engulfed by phagocytic cells. Using the syngeneic mouse model, we demonstrated that RL2-treated MX-7 rhabdomyosarcoma cells confer long-term immune-mediated protection against challenge with live MX-7 cells. We also analyzed the combinatorial antitumor effect of vaccination with RL2-treated cells and the inhibition of indoleamine 2,3-dioxygenase (IDO) with ethyl pyruvate. Compared to solo anti-tumor immunization with RL2-treated cells, additional chemical inhibition of IDO demonstrated better long-term antitumor responses than vaccination alone.


Asunto(s)
Antineoplásicos , Caseínas , Inhibidores Enzimáticos , Indolamina-Pirrol 2,3,-Dioxigenasa/antagonistas & inhibidores , Proteínas de Neoplasias/antagonistas & inhibidores , Neoplasias Experimentales/tratamiento farmacológico , Vacunación , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Caseínas/química , Caseínas/farmacología , Muerte Celular , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Células MCF-7 , Ratones , Proteínas de Neoplasias/metabolismo , Neoplasias Experimentales/enzimología , Neoplasias Experimentales/patología , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacología
10.
Genesis ; 57(10): e23323, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31218818

RESUMEN

Neuroblastoma, an embryonal tumor arising from the sympathetic ganglia and adrenal medulla, is among the most intractable pediatric cancers. Although a variety of genetic changes have been identified in neuroblastoma, how they contribute to its pathogenesis remains largely unclear. Recent studies have identified alterations of the anaplastic lymphoma kinase (ALK) gene in neuroblastoma; ALK F1174L (a phenylalanine-to-leucine substitution at codon 1174) represents one of the most frequent of these somatic mutations, and is associated with amplification of the MYCN gene, the most reliable marker for the poor survival. We engineered the mouse Alk locus so that ALK F1174L is expressed by its endogenous promoter and can be induced in a spatiotemporally controlled fashion using Cre-loxP system. Although expression of ALK F1174L resulted in enhanced proliferation of sympathetic ganglion progenitors and increased the size of the sympathetic ganglia, it was insufficient to cause neuroblastoma. However, lethal neuroblastoma frequently developed in mice co-expressing ALK F1174L and MYCN, even in a genetic background where MYCN alone does not cause overt tumors. These data reveal that physiological expression of ALK F1174L significantly potentiates the oncogenic ability of MYCN in vivo. Our conditional mutant mice provide a valuable platform for investigating the pathogenesis of neuroblastoma.


Asunto(s)
Quinasa de Linfoma Anaplásico/genética , Neoplasias Experimentales/genética , Neuroblastoma/etiología , Animales , Carcinogénesis/genética , Femenino , Ganglios Simpáticos/crecimiento & desarrollo , Ingeniería Genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes/genética , Mutagénesis Insercional , Proteína Proto-Oncogénica N-Myc/biosíntesis , Neoplasias Experimentales/enzimología , Neoplasias Experimentales/etiología , Neuroblastoma/enzimología , Neuroblastoma/genética
11.
Biochem Biophys Res Commun ; 496(3): 806-813, 2018 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-29395081

RESUMEN

Methionine sulfoxide reductase B1 (MsrB1), a member of the selenoprotein family and contributes significantly to the reduction of methionine sulfoxides produced from reactive oxygen species (ROS). However, few studies have examined the role of MsrB1 in tumors. Here We tested the proliferation and invasion in MsrB1 knockdown u2os cells under H2O2/thioredoxin. As shown in our result, knockdown of MsrB1 inhibited the proliferation of u2os cells and regulates mitogen-activated protein kinase (MAPK) pathway by down-regulation of Erk, MeK phosphorylation and p53 expression in u2os cells. In a xenograft tumorigenicity mice, MsrB1 knockdown effectively inhibited tumor growth. Furthermore, MsrB1 knockdown resulted in migration and invasion reducement of u2os cells. MsrB1 regulates epithelial-mesenchymal transition (EMT) via affecting cytoskeleton by increasing E-cadherin expression and decreasing N-cadherin, TGF-ß1, slug, fibronectin, vimentin, c-myc, snail and ß-catenin expressions. In vivo, MsrB1 shRNAi can inhibit lung metastasis in metastasis model. In conclusion, MsrB1 regulates proliferation and invasion of u2os cells by affecting MAPK pathway and EMT, and MsrB1 gene may be a novel therapeutic target against tumors.


Asunto(s)
Transición Epitelial-Mesenquimal , Quinasas MAP Reguladas por Señal Extracelular , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Metionina Sulfóxido Reductasas/metabolismo , Neoplasias Experimentales/enzimología , Neoplasias Experimentales/patología , Línea Celular Tumoral , Proliferación Celular , Humanos , Invasividad Neoplásica
12.
Biochem Biophys Res Commun ; 503(3): 1378-1384, 2018 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-30029880

RESUMEN

BACKGROUND: PTK6 is involved in cell proliferation, invasion and migration. Patients with lower PTK6 expression predicts poor prognosis of LSCC. However, the mechanism of PTK6 in LSCC progression remains unclear. We investigated the role of PTK6 in the pathogenesis of LSCC. METHODS: Human LSCC tissues and paired adjacent non-tumor tissues were obtained to evaluate PTK6 expression. The biological function of PTK6 in LSCC was determined by overexpression of PTK6 in Hep-2 cells in vitro and in nude mice. The potential PTK6 target factors and signaling pathways were identified by Western blotting assay and immunohistochemical staining. RESULTS: PTK6 was downregulated in tissues of human LSCC. Biological function investigation of PTK6 demonstrated that overexpression of PTK6 significantly decreased cell growth, clonogenicity, invasion and migration capacity in vitro and suppressed xenograft tumor growth as well as lung metastasis in vivo. PTK6 suppresses LSCC proliferation mainly by inhibiting c-myc and cyclinD1 expression. In addition, PTK6 promotes cell apoptosis in LSCC. Moreover, PTK6 mitigated LSCC invasion and migration through regulating EMT and MMP-9. CONCLUSION: PTK6 plays a tumor suppressor role in LSCC by regulating c-myc and cyclinD1 expression, cell apoptosis, EMT and MMP-9.


Asunto(s)
Carcinoma de Células Escamosas/enzimología , Carcinoma de Células Escamosas/patología , Progresión de la Enfermedad , Neoplasias Laríngeas/enzimología , Neoplasias Laríngeas/patología , Metástasis de la Neoplasia , Proteínas de Neoplasias/genética , Proteínas Tirosina Quinasas/genética , Animales , Apoptosis , Carcinoma de Células Escamosas/metabolismo , Proliferación Celular , Supervivencia Celular , Humanos , Neoplasias Laríngeas/metabolismo , Ratones , Ratones Desnudos , Proteínas de Neoplasias/metabolismo , Neoplasias Experimentales/enzimología , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Proteínas Tirosina Quinasas/metabolismo , Células Tumorales Cultivadas
13.
Toxicol Appl Pharmacol ; 352: 87-96, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29802912

RESUMEN

Previously, we proved that caffeic acid (CA), a major dietary phenolic acid, prevents skin carcinogenesis by modulating inflammatory signaling in mouse skin. However, the actual mechanisms of CA against UVB (280-320 nm) induced photocarcinogenesis remains unclear. The present results confirms that CA significantly inhibits single UVB-induced CPDs formation, oxidative DNA damage, ROS generation and frequency of apoptotic cell death in human dermal fibroblasts (HDFa). Furthermore, CA prevents UVB-induced expression of PI3K and AKT kinases through activation of PTEN which subsequently promotes XPC dependant NER proteins such as XPC, XPE, TFIIH (p44) and ERCC1 in HDFa cells and mouse skin tissue. Further, CA directly activates PTEN through hydrogen bond and hydrophobic interactions. Taken together, these findings suggest that CA prevents UVB-induced photodamage through the activation of PTEN expression in human dermal fibroblasts and mouse skin.


Asunto(s)
Anticarcinógenos/farmacología , Ácidos Cafeicos/farmacología , Transformación Celular Neoplásica/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Neoplasias Experimentales/prevención & control , Neoplasias Inducidas por Radiación/prevención & control , Fosfohidrolasa PTEN/metabolismo , Transducción de Señal/efectos de los fármacos , Neoplasias Cutáneas/prevención & control , Piel/efectos de los fármacos , Rayos Ultravioleta/efectos adversos , Animales , Apoptosis/efectos de los fármacos , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Transformación Celular Neoplásica/efectos de la radiación , Daño del ADN/efectos de los fármacos , Reparación del ADN/efectos de los fármacos , Enzimas Reparadoras del ADN/metabolismo , Fibroblastos/enzimología , Fibroblastos/patología , Fibroblastos/efectos de la radiación , Humanos , Ratones , Neoplasias Experimentales/enzimología , Neoplasias Experimentales/patología , Neoplasias Inducidas por Radiación/enzimología , Neoplasias Inducidas por Radiación/patología , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/efectos de la radiación , Transducción de Señal/efectos de la radiación , Piel/enzimología , Piel/patología , Piel/efectos de la radiación , Neoplasias Cutáneas/enzimología , Neoplasias Cutáneas/patología
14.
Bioorg Med Chem Lett ; 28(5): 915-921, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29422388

RESUMEN

Carbonic anhydrase IX is overexpressed in many solid tumors including hypoxic tumors and is a potential target for cancer therapy and diagnosis. Reported imaging agents targeting CA-IX are successful mostly in clear cell renal carcinoma as SKRC-52 and no candidate was approved yet in clinical trials for imaging of CA-IX. To validate CA-IX as a valid target for imaging of hypoxic tumor, we designed and synthesized novel [18F]-PET tracer (1) based on acetazolamide which is one of the well-known CA-IX inhibitors and performed imaging study in CA-IX expressing hypoxic tumor model as 4T1 and HT-29 in vivo models other than SKRC-52. [18F]-acetazolamide (1) was found to be insufficient for the specific accumulation in CA-IX expressing tumor. This study might be useful to understand in vivo behavior of acetazolamide PET tracer and can contribute to the development of successful PET imaging agents targeting CA-IX in future. Additional study is needed to understand the mechanism of poor targeting of CA-IX, as if CA-IX is not reliable as a sole target for imaging of CA-IX expressing hypoxic solid tumors.


Asunto(s)
Acetazolamida/química , Anhidrasa Carbónica IX/análisis , Inhibidores de Anhidrasa Carbónica/química , Carcinoma de Células Renales/enzimología , Neoplasias Renales/enzimología , Tomografía de Emisión de Positrones , Acetazolamida/síntesis química , Acetazolamida/farmacocinética , Animales , Anhidrasa Carbónica IX/biosíntesis , Anhidrasa Carbónica IX/metabolismo , Inhibidores de Anhidrasa Carbónica/síntesis química , Inhibidores de Anhidrasa Carbónica/farmacocinética , Carcinoma de Células Renales/diagnóstico , Radioisótopos de Flúor , Humanos , Neoplasias Renales/diagnóstico , Ratones , Ratones Endogámicos BALB C , Neoplasias Experimentales/diagnóstico , Neoplasias Experimentales/enzimología , Distribución Tisular
15.
Anal Bioanal Chem ; 410(26): 6771-6777, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29909457

RESUMEN

γ-Glutamyl transpeptidase (GGT), overexpressed in various cancer cells, has been perceived as a latent tumor biomarker. Thus, developing near-infrared (NIR) fluorescent GGT probes is highly desired for in vivo tumor imaging and studies. To our knowledge, however, such a GGT probe is still rare. Herein, we construct a new GGT-activatable NIR fluorescent probe HCAGlu by incorporating γ-glutamyl group as a recognition unit directly into a NIR hemicyanine fluorophore. HCAGlu exhibits a highly sensitive and selective NIR fluorescence off-on response to GGT. The probe has been applied to cell and histological section imaging, which demonstrates the ability of HCAGlu in distinguishing different GGT-expression levels in situ in biological samples. Notably, in vivo fluorescence imaging in tumor-bearing mice has been performed, verifying that probe HCAGlu can rapidly produce a distinct fluorescence signal in the tumor site via both intravenous and intratumoral injections. The simplicity and excellent performance of HCAGlu make it of high potential in studying the physiological function of GGT in vivo. Graphical abstract A near-infrared fluorescent probe for imaging γ-glutamyl transpeptidase in biological samples.


Asunto(s)
Colorantes Fluorescentes/química , Rayos Infrarrojos , Neoplasias Experimentales/diagnóstico por imagen , gamma-Glutamiltransferasa/metabolismo , Animales , Fluorescencia , Colorantes Fluorescentes/análisis , Células HeLa , Células Hep G2 , Humanos , Límite de Detección , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Células 3T3 NIH , Neoplasias Experimentales/enzimología , Análisis Espectral/métodos
16.
J Cell Biochem ; 118(11): 3635-3642, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-27171483

RESUMEN

We have established an orthotopic nude-mouse model of gastric cancer carcinomatosis peritonitis, a recalcitrant disease in human patients. Human MKN45 poorly-differentiated human gastric cancer cells developed carcinomatosis peritonitis upon orthotopic transplantation in nude mice. The MKN45 cells expressed the fluorescent ubiquitination-based cell cycle indicator (FUCCI) that color codes the phases of the cell cycle. The intra-peritoneal tumors and ascites contained mostly quiescent G1 /Go cancer cells visualized as red by FUCCI imaging. Cisplatinum (CDDP) treatment did not reduce bloody ascites, and larger tumors formed in the peritoneal cavity after CDDP treatment in an early-stage carcinomatosis peritonitis orthotopic mouse model. Paclitaxel-treated mice had reduced ascites, but also had large tumor masses in the peritonium after treatment with cancer cells mostly in G0 /G1 , visualized by FUCCI red. In contrast, OBP-301 telomerase-dependent adenovirus-treated mice had no ascites and only small tumor nodules consisting of cancer cells mostly in S/G2 phases in the early-stage carcinomatosis peritonitis model, visualized by FUCCI green. Furthermore, OBP-301 significantly reduced the size of tumors (P < 0.01) and ascites even in a late-stage carcinomatosis peritonitis model. These results suggest that quiescent peritoneally-disseminated gastric cancer cells are resistant to conventional chemotherapy, but OBP-301 significantly reduced the weight of the tumors and increased survival, suggesting clinical potential. J. Cell. Biochem. 118: 3635-3642, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Adenocarcinoma/enzimología , Adenoviridae , Proteínas de Neoplasias/metabolismo , Neoplasias Experimentales/enzimología , Neoplasias Peritoneales/enzimología , Neoplasias Gástricas/enzimología , Telomerasa/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/patología , Animales , Fase G2/genética , Humanos , Ratones Desnudos , Proteínas de Neoplasias/genética , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Neoplasias Peritoneales/genética , Neoplasias Peritoneales/patología , Fase S/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Telomerasa/genética
17.
Biochem Biophys Res Commun ; 487(3): 613-618, 2017 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-28433633

RESUMEN

EGFR-mutant lung adenocarcinomas contain a subpopulation of cells that have undergone epithelial-to-mesenchymal transition and can grow independently of EGFR. To kill these cancer cells, we need a novel therapeutic approach other than EGFR inhibitors. If a molecule is specifically expressed on the cell surface of such EGFR-independent EGFR-mutant cancer cells, it can be a therapeutic target. We found that a mesenchymal EGFR-independent subline derived from HCC827 cells, an EGFR-mutant lung adenocarcinoma cell line, expressed angiotensin-converting enzyme 2 (ACE2) to a greater extent than its parental cells. ACE2 was also expressed at least partially in most of the primary EGFR-mutant lung adenocarcinomas examined, and the ACE2 expression level in the cancer cells was much higher than that in normal lung epithelial cells. In addition, we developed an anti-ACE2 mouse monoclonal antibody (mAb), termed H8R64, that was internalized by ACE2-expressing cells. If an antibody-drug conjugate consisting of a humanized mAb based on H8R64 and a potent anticancer drug were produced, it could be effective for the treatment of EGFR-mutant lung adenocarcinomas.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Anticuerpos Monoclonales Humanizados/farmacología , Antineoplásicos/farmacología , Receptores ErbB/genética , Neoplasias Pulmonares/tratamiento farmacológico , Terapia Molecular Dirigida , Peptidil-Dipeptidasa A/metabolismo , Adenocarcinoma/enzimología , Adenocarcinoma/genética , Adenocarcinoma/patología , Adenocarcinoma del Pulmón , Enzima Convertidora de Angiotensina 2 , Inhibidores de la Enzima Convertidora de Angiotensina/química , Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Animales , Anticuerpos Monoclonales Humanizados/química , Anticuerpos Monoclonales Humanizados/uso terapéutico , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Receptores ErbB/metabolismo , Humanos , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Ratones , Ratones Endogámicos BALB C , Mutación , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/enzimología , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Peptidil-Dipeptidasa A/genética , Relación Estructura-Actividad
18.
Analyst ; 142(14): 2624-2630, 2017 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-28608874

RESUMEN

In recent years, many activatable fluorescent probes have been developed for hNQO1 detection. However, most of the reported fluorescent probes are susceptible to the interferences of endogenous fluorescence and have the drawback of inadequate penetration depth. Very recently, researchers have reported a two-photon excitation (TPE) fluorescent probe for hNQO1 detection. Nevertheless, this probe only exhibits a compromised signal-to-background ratio, and has not been applied to image hNQO1 in living tissues. Herein, a novel TPE fluorescent probe, trimethyl locked quinone caged Acedan (Q3CA-P), has been developed for hNQO1 detection and imaging in living cells and tissues. Q3CA-P displays over 25-fold enhancement in fluorescence intensity toward hNQO1 with a Stokes shift over 100 nm in one-photon excitation and exhibits a very low detection limit of 5.6 ng mL-1. The imaging experiments performed in tumour cells and tissue slices using Q3CA-P demonstrate that Q3CA-P could image the endogenous hNQO1 with high selectivity and sensitivity with a TPE probing depth of 120 µm. Thus, our probe may have great potential for use in cancer diagnosis and image-guided surgery.


Asunto(s)
Colorantes Fluorescentes , NAD(P)H Deshidrogenasa (Quinona)/análisis , Neoplasias Experimentales/diagnóstico por imagen , Fotones , Animales , Fluorescencia , Células HT29 , Células HeLa , Humanos , Neoplasias Experimentales/enzimología , Ratas Desnudas
19.
Analyst ; 142(10): 1813-1820, 2017 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-28466919

RESUMEN

Oxidative stress, a disturbance in the balance between oxidant/antioxidant ratios, is associated with cancer, aging, inflammation, neurodegenerative diseases and other conditions. γ-Glutamyltranspeptidase (GGT) is a redox-related enzyme that plays a key role in mitigating the effects of oxidative stress by maintaining cellular glutathione (GSH) metabolism and homeostasis. Therefore, oxidative stress will upregulate the intracellular GGT level. To better understand the major pathophysiological resist mechanism to oxidative injury in mediating many disease states, we designed and synthesized a novel two-photon (TP) fluorescent turn-on probe, Np-Glu, for GGT detection and bioimaging. Under the optimized conditions, Np-Glu exhibited remarkable fluorescence enhancement (150-fold), good selectivity and high sensitivity (LOD is 0.033 U L-1), with a wide linear concentration range (0-50 U L-1). More importantly, the probe Np-Glu was successfully applied in one-photon and TP fluorescence imaging of GGT activity in an oxidative stress model in living cells and tissues, suggesting Np-Glu as an ideal indicator for clinical and biological samples.


Asunto(s)
Colorantes Fluorescentes , Neoplasias Hepáticas/enzimología , Neoplasias Experimentales/enzimología , Estrés Oxidativo , gamma-Glutamiltransferasa/metabolismo , Animales , Glutatión/metabolismo , Células Hep G2 , Humanos , Ratones Desnudos , Fotones
20.
J Immunol ; 194(10): 5014-21, 2015 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-25870245

RESUMEN

The efficiency of immune-mediated clearance of cancer cells is hampered by immunosuppressive mediators in the malignant microenvironment, including NADPH oxidase-derived reactive oxygen species. We aimed at defining the effects of histamine, an inhibitor of the myeloid NADPH oxidase/NOX2, on the development of Ag-presenting dendritic cells (DCs) from myeloid precursors and the impact of these mechanisms for tumor growth. Histamine was found to promote the maturation of human DCs from monocytes by increasing the expression of HLA-DR and costimulatory molecules, which resulted in improved induction of Th cells with Th0 polarity. Experiments using wild-type and NOX2-deficient myelomonoblastic cells showed that histamine facilitated myeloid cell maturation only in cells capable of generating reactive oxygen species. Treatment of mice with histamine reduced the growth of murine EL-4 lymphomas in parallel with an increment of tumor-infiltrating DCs in NOX2-sufficient mice but not in NOX2-deficient (gp91(phox) (-/-)) mice. We propose that strategies to target the myeloid NADPH oxidase may facilitate the development of endogenous DCs in cancer.


Asunto(s)
Células Dendríticas/inmunología , Histamina/inmunología , NADPH Oxidasas/inmunología , Neoplasias Experimentales/inmunología , Animales , Diferenciación Celular/inmunología , Técnicas de Cocultivo , Células Dendríticas/citología , Citometría de Flujo , Humanos , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Monocitos/citología , Monocitos/inmunología , Neoplasias Experimentales/enzimología , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA