Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Plant Cell Physiol ; 65(4): 671-679, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38226464

RESUMEN

Nutritropism is a positive tropism toward nutrients in plant roots. An NH4+ gradient is a nutritropic stimulus in rice (Oryza sativa L.). When rice roots are exposed to an NH4+ gradient generated around nutrient sources, root tips bend toward and coil around the sources. The molecular mechanisms are largely unknown. Here, we analyzed the transcriptomes of the inside and outside of bending root tips exhibiting nutritropism to reveal nutritropic signal transduction. Tissues facing the nutrient sources (inside) and away (outside) were separately collected by laser microdissection. Principal component analysis revealed distinct transcriptome patterns between the two tissues. Annotations of 153 differentially expressed genes implied that auxin, gibberellin and ethylene signaling were activated differentially between the sides of the root tips under nutritropism. Exogenous application of transport and/or biosynthesis inhibitors of these phytohormones largely inhibited the nutritropism. Thus, signaling and de novo biosynthesis of the three phytohormones are necessary for nutritropism. Expression patterns of IAA genes implied that auxins accumulated more in the inside tissues, meaning that ammonium stimulus is transduced to auxin signaling in nutritropism similar to gravity stimulus in gravitropism. SAUR and expansin genes, which are known to control cell wall modification and to promote cell elongation in shoot gravitropism, were highly expressed in the inside tissues rather than the outside tissues, and our transcriptome data are unexplainable for differential elongation in root nutritropism.


Asunto(s)
Etilenos , Perfilación de la Expresión Génica , Regulación de la Expresión Génica de las Plantas , Giberelinas , Ácidos Indolacéticos , Oryza , Transducción de Señal , Oryza/genética , Oryza/metabolismo , Oryza/fisiología , Ácidos Indolacéticos/metabolismo , Giberelinas/metabolismo , Etilenos/metabolismo , Reguladores del Crecimiento de las Plantas/metabolismo , Meristema/genética , Meristema/metabolismo , Transcriptoma , Raíces de Plantas/metabolismo , Raíces de Plantas/genética , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo
2.
Med Sci (Basel) ; 11(2)2023 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-37218983

RESUMEN

Chemokines are a group of cytokines involved in the mobilization of leukocytes, which play a role in host defense and a variety of pathological conditions, including cancer. Interferon (IFN)-inducible chemokines C-X-C motif ligand 9 (CXCL), CXCL10, and CXCL11 are anti-tumor chemokines; however, the differential anti-tumor effects of IFN-inducible chemokines are not completely understood. In this study, we investigated the anti-tumor effects of IFN-inducible chemokines by transferring chemokine expression vectors into a mouse squamous cell carcinoma cell line, SCCVII, to generate a cell line stably expressing chemokines and transplanted it into nude mice. The results showed that CXCL9- and CXCL11-expressing cells markedly inhibited tumor growth, whereas CXCL10-expressing cells did not inhibit growth. The NH2-terminal amino acid sequence of mouse CXCL10 contains a cleavage sequence by dipeptidyl peptidase 4 (DPP4), an enzyme that cleaves the peptide chain of chemokines. IHC staining indicated DPP4 expression in the stromal tissue, suggesting CXCL10 inactivation. These results suggest that the anti-tumor effects of IFN-inducible chemokines are affected by the expression of chemokine-cleaving enzymes in tumor tissues.


Asunto(s)
Carcinoma de Células Escamosas , Quimiocina CXCL10 , Quimiocina CXCL11 , Quimiocina CXCL9 , Animales , Ratones , Línea Celular , Quimiocina CXCL10/metabolismo , Dipeptidil Peptidasa 4 , Interferón gamma/farmacología , Ratones Desnudos , Quimiocina CXCL9/metabolismo , Quimiocina CXCL11/metabolismo
3.
Biomedicines ; 10(4)2022 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-35453513

RESUMEN

Combined treatment of human oral squamous cell carcinoma (OSCCs) with DNA methyltransferase inhibitors (DNMTis), histone methyltransferase inhibitors (HMTis), and histone deacetylase inhibitors (HDACis), and the molecular mechanisms underlying their anticancer effects, have not been fully elucidated. Herein, we investigated the cytotoxic effects of combined DNMTis (5-Aza-deoxycytidine: 5-Aza-dC, RG108), HMTis (3-deazaneplanocin A: DZNep), and HDACis (trichostatin A: TSA) treatment on human OSCC cells and explored their molecular mechanisms. Combined 5-Aza-dC, or RG108, and TSA treatment significantly decreased HSC-2 and Ca9-22 cell viability. Combinatorial DZNep and TSA treatment also decreased Ca9-22 cell viability. Although caspase 3/7 activation was not observed in HSC-2 cells following combined treatment, caspase activity was significantly increased in Ca9-22 cells treated with DZNep and TSA. Moreover, combined treatment with 5-Aza-dC, RG108, and TSA increased the proportion of HSC-2 and Ca9-22 cells in the S and G2/M phases. Meanwhile, increased phosphorylation of the histone variant H2A.X, a marker of double-stranded DNA breaks, was observed in both cells after combination treatment. Hence, the decreased viability induced by combined treatment with epigenomic inhibitors results from apoptosis and cell cycle arrest in S and G2/M phases. Thus, epigenomic therapy comprising combined low concentrations of DNMTi, HMTi, and HDACi is effective against OSCC.

4.
Cancers (Basel) ; 13(8)2021 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-33921389

RESUMEN

Chemokines and cytokines in the tumor microenvironment influence immune cell infiltration and activation. To elucidate their role in immune cell recruitment during oral cancer development, we generated a mouse tongue cancer model using the carcinogen 4-nitroquinoline 1-oxide (4NQO) and investigated the carcinogenetic process and chemokine/cytokine gene expression kinetics in the mouse tongue. C57/BL6 mice were administered 4NQO in drinking water, after which tongues were dissected at 16 and 28 weeks and subjected to analysis using the RT2 Profiler PCR Array, qRT-PCR, and pathologic and immunohistochemical analyses. We found that Th1-associated chemokine/cytokine (Cxcl9, Cxcl10, Ccl5, and Ifng) and Treg-associated chemokine/cytokine (Ccl17, Ccl22, and Il10) mRNA levels were simultaneously increased in premalignant lesions of 4NQO-treated mice at 16 weeks. Additionally, although levels of Gata3, a Th2 marker, were not upregulated, those of Cxcr3, Ccr4, and Foxp3 were upregulated in the tongue tissue. Furthermore, immunohistochemical analysis confirmed the infiltration of CD4+, CD8+, and Foxp3+ cells in the tongue tissue of 4NQO-treated mice, as well as significant correlations between Th1- or Treg-associated chemokine/cytokine mRNA expression and T cell infiltration. These results indicate that CD4+, CD8+, and Foxp3+ cells were simultaneously recruited through the expression of Th1- and Treg-associated chemokines in premalignant lesions of 4NQO-induced mouse tongue tissue.

5.
Cytokine ; 118: 80-92, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30017387

RESUMEN

Many tumor cells escape from cancer immunosurveillance and resist treatment with interferons (IFNs). Although the mechanism underlying IFN resistance is mostly attributed to a deficiency of components of the IFN-signaling pathway, some types of tumor cells resist IFN-mediated cell growth arrest despite the presence of an intact JAK/STAT signaling pathway. However, the molecular mechanisms underlying the unresponsiveness to IFNs independent of the defective JAK/STAT pathway remain to be clarified. To elucidate the mechanisms underlying IFNγ resistance, we examined the anti-proliferative effect of IFNγ on mouse tumor cell lines. Mouse squamous cell carcinoma (SCCVII) cells were resistant to IFNγ-mediated cell growth arrest despite the presence of the IFNγ-induced STAT1-dependent signaling pathway, whereas IFNγ inhibited cell growth of B16/F1 cells, a well-known IFNγ-sensitive mouse melanoma cell line, at the G1 phase of the cell cycle. Treatment of SCCVII cells with IFNγ neither downregulated the expression of cyclin D1, cyclin A2, and cyclin E1 nor induced a hypo-phosphorylated, active form of retinoblastoma protein (pRb). Interestingly, the hyper-phosphorylated, inactive form of pRb was exclusively localized in the cytoplasm in SCCVII cells. The IFN-inducible 204 gene (Ifi204), whose gene product, p204, binds to pRb and exerts an anti-proliferative effect, was repressed in SCCVII cells. p204 overexpression in SCCVII significantly inhibited cell growth, and mutation of a pRb-binding LXCXE motif decreased the anti-proliferative effect. These results suggest that silencing of Ifi204/p204 induces resistance to IFNγ-mediated cell growth arrest in SCCVII cells.


Asunto(s)
Proliferación Celular/genética , Inhibidores de Crecimiento/genética , Interferón gamma/genética , Proteínas Nucleares/genética , Fosfoproteínas/genética , Animales , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Regulación hacia Abajo/genética , Fase G1/genética , Melanoma/genética , Melanoma/patología , Ratones , Fosforilación/genética , Unión Proteica/genética , Proteína de Retinoblastoma/genética , Transducción de Señal/genética
6.
Mediators Inflamm ; 2018: 3979606, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30158833

RESUMEN

During chronic inflammation from diseases, such as periodontal disease, the proinflammatory cytokines interferon-gamma (IFNγ) and tumor necrosis factor-α (TNFα) alter bone remodeling. To elucidate the underlying molecular mechanisms, we investigated the effect of IFNγ and TNFα on the proliferation and survival of clonal MC3T3-E1 mouse osteoblasts. We found that although IFNγ or TNFα alone affected cell growth and survival only marginally, costimulation with both synergistically inhibited cell growth and reduced cell viability. The diminished cell viability was due to apoptosis, as indicated by increased TUNEL staining and elevated caspase 3, 8, and 9 activities. Western blot also showed that costimulation with IFNγ and TNFα elicited cytochrome c release and downregulated B cell lymphoma 2 (Bcl-2) expression without affecting Bcl-2-associated X (Bax) protein expression. Furthermore, stable Bcl-2 overexpression significantly alleviated cell death following costimulation. Collectively, these results suggested that IFNγ and TNFα elicited osteoblast apoptosis via cytochrome c release from damaged mitochondria, caspase activation, and Bcl-2 downregulation.


Asunto(s)
Citocromos c/metabolismo , Interferón gamma/farmacología , Mitocondrias/metabolismo , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Etiquetado Corte-Fin in Situ , Ratones , Mitocondrias/efectos de los fármacos , Proteína X Asociada a bcl-2/metabolismo
7.
BMC Cancer ; 15: 573, 2015 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-26242181

RESUMEN

BACKGROUND: Tumor-associated macrophages (TAMs) are implicated in the growth, invasion and metastasis of various solid tumors. However, the phenotype of TAMs in premalignant lesions of solid tumors has not been clarified. In the present study, we identify the phenotype of TAMs in leukoplakia, an oral premalignant lesion, by immunohistochemical analysis and investigate the involvement of infiltrated T cells that participate in the polarization of TAMs. METHODS: The subjects included 30 patients with oral leukoplakia and 10 individuals with normal mucosa. Hematoxylin and eosin slides were examined for the histological grades, and immunohistochemical analysis was carried out using antibodies against CD68 (pan-MΦ), CD80 (M1 MΦ), CD163 (M2 MΦ), CD4 (helper T cells: Th), CD8 (cytotoxic T cells), CXCR3, CCR5 (Th1), CCR4 (Th2), signal transducer and activator of transcription (STAT1), phosphorylated STAT1 (pSTAT1) and chemokine CXCL9. The differences in the numbers of positively stained cells among the different histological grades were tested for statistical significance using the Kruskal-Wallis test. Correlations between different types of immune cells were determined using Spearman's rank analysis. RESULTS: An increase in the rate of CD163(+) TAM infiltration was observed in mild and moderate epithelial dysplasia, which positively correlated with the rate of intraepithelial CD4(+) Th cell infiltration. Although CCR4(+) cells rarely infiltrated, CXCR3(+) and CCR5(+) cells were observed in these lesions. Cells positive for STAT1 and chemokine CXCL9, interferon- (IFN)-induced gene products, and pSTAT1 were also observed in the same lesions. Double immunofluorescence staining demonstrated that the cells that were positive for CD163 were also positive for STAT1. CONCLUSIONS: CD163(+) TAMs in oral premalignant lesions coexpress CD163 and STAT1, suggesting that the TAMs in oral premalignant lesions possess an M1 phenotype in a Th1-dominated micromilieu.


Asunto(s)
Antígenos CD/genética , Antígenos de Diferenciación Mielomonocítica/genética , Macrófagos/metabolismo , Macrófagos/patología , Neoplasias de la Boca/genética , Neoplasias de la Boca/patología , Lesiones Precancerosas , Receptores de Superficie Celular/genética , Factor de Transcripción STAT1/genética , Células TH1/metabolismo , Adulto , Anciano , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Biomarcadores , Biopsia , Estudios de Casos y Controles , Femenino , Humanos , Leucopenia/genética , Leucopenia/inmunología , Leucopenia/patología , Macrófagos/inmunología , Masculino , Persona de Mediana Edad , Neoplasias de la Boca/inmunología , Clasificación del Tumor , Unión Proteica , Transporte de Proteínas , Receptores CXCR3/metabolismo , Receptores de Superficie Celular/metabolismo , Factor de Transcripción STAT1/metabolismo , Células TH1/inmunología , Microambiente Tumoral
8.
Mediators Inflamm ; 2013: 369693, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24459328

RESUMEN

Inducible nitric oxide synthase (iNOS) is a signature molecule involved in the classical activation of M1 macrophages and is induced by the Nos2 gene upon stimulation with Th1-cell derived interferon-gamma (IFNγ) and bacterial lipopolysaccharide (LPS). Although the anti-inflammatory cytokine IL-4 is known to inhibit Nos2 gene expression, the molecular mechanism involved in the negative regulation of Nos2 by IL-4 remains to be fully elucidated. In the present study, we investigated the mechanism of IL-4-mediated Nos2 transcriptional repression in the mouse macrophage-like cell line RAW264.7. Signal transducer and activator of transcription 6 (Stat6) knockdown by siRNA abolished the IL-4-mediated inhibition of Nos2 induced by IFNγ/LPS. Transient transfection of a luciferase reporter gene containing the 5'-flanking region of the Nos2 gene demonstrated that an octamer transcription factor (OCT) binding site in the promoter region is required for both positive regulation by IFNγ/LPS and negative regulation by IL-4. Although IL-4 had no inhibitory effect on the DNA-binding activity of constitutively expressed Oct-1, IL-4-induced Nos2-reporter transcriptional repression was partially attenuated by overexpression of the coactivator CREB-binding protein (CBP). These results suggest that a coactivator/cofactor that functionally interacts with Oct-1 is a molecular target for the IL-4-mediated inhibition of Nos2 and that IL-4-activated Stat6 represses Oct-1-dependent transcription by competing with this coactivator/cofactor.


Asunto(s)
Interleucina-4/farmacología , Óxido Nítrico Sintasa de Tipo II/genética , Factor 1 de Transcripción de Unión a Octámeros/fisiología , Animales , Células Cultivadas , Interferón gamma/farmacología , Lipopolisacáridos/farmacología , Macrófagos/enzimología , Proteínas de la Membrana/fisiología , Ratones , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Proteínas de Transporte de Catión Orgánico , Transportador 2 de Cátion Orgánico , Fosfoproteínas/fisiología , Regiones Promotoras Genéticas , Factor de Transcripción STAT6/fisiología
9.
J Oral Sci ; 54(3): 241-50, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23047035

RESUMEN

Human podoplanin is a type-1 transmembrane sialomucin-like glycoprotein that is involved in cell migration, tumor cell invasion and metastasis. Our recent study of oral squamous cell carcinoma (OSCC) demonstrated that the degree of immunohistochemical expression of podoplanin was correlated with the severity of epithelial dysplasia and significantly associated with a poor pathologic grade of differentiation. Furthermore, it has been reported that Src directly associates with the epidermal growth factor receptor (EGFR) in OSCC cells upon stimulation with EGF and phosphorylates Crk-associated substrate (Cas), podoplanin acting downstream of Src and Cas to promote cell migration. However, the molecular function of podoplanin remains unclear. In this study we performed real-time RT-PCR, Western blotting and scratch assay using OSCC cell lines in order to clarify the molecular biological function of podoplanin expression associated with various growth factors including EGF and with the Src-Cas signaling pathway. Podoplanin was found to have a marked influence on cancer cell migration and the expression of matrix metalloprotease-9 (MMP-9) in the oral cavity upon stimulation with EGF. Podoplanin promotes oral cancer cell migration, and the EGF-Src-Cas pathway is one of the possible mechanisms responsible for progression of cancer in the oral cavity.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Movimiento Celular , Sistema de Señalización de MAP Quinasas , Glicoproteínas de Membrana/fisiología , Neoplasias de la Boca/metabolismo , Proteínas de Neoplasias/fisiología , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proteína Sustrato Asociada a CrK , Factor de Crecimiento Epidérmico/fisiología , Humanos , Metaloproteinasa 9 de la Matriz/metabolismo , Glicoproteínas de Membrana/farmacología , Neoplasias de la Boca/patología , Invasividad Neoplásica , Proteínas de Neoplasias/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Microambiente Tumoral
10.
Cell Immunol ; 276(1-2): 101-9, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22608126

RESUMEN

T helper (Th)2 polarized immune responses are characteristically dominant in helminth infections. The gene expression of interferon (IFN)-γ-inducible protein 10 (IP-10/CXCL10), which promotes Th1 responses, in mouse macrophages stimulated with lipopolysaccharide (LPS) and/or IFN-γ was suppressed by excretory/secretory (ES) products of Spirometra erinaceieuropaei plerocercoids. ES products suppressed LPS- and/or IFN-γ-induced transcriptional activities of a luciferase reporter gene under the control of a 243-bp fragment of the IP-10 gene promoter/enhancer, which contains an IFN-stimulated response element (ISRE) and two κB elements. Consistent with this result, ES products inhibited ISRE-dependent heterologous promoter activities and LPS- or IFN-γ-induced ISRE-binding activity. ES products also suppressed LPS-induced IFN-ß gene expression. Furthermore, ES products suppressed nuclear factor (NF)-κB RelA (p65)-dependent transcriptional activity, whereas ES products had no effect on the κB-binding activity. These results suggest that ES products suppress the IP-10 gene expression by inhibiting the ISRE- and RelA-dependent transcriptional activities in mouse macrophages.


Asunto(s)
Quimiocina CXCL10/inmunología , Interferón gamma/inmunología , Lipopolisacáridos/inmunología , Macrófagos/inmunología , Spirometra/inmunología , Animales , Línea Celular , Quimiocina CXCL10/genética , Regulación hacia Abajo , Ratones , Fosforilación , Regiones Promotoras Genéticas , ARN Mensajero/genética , Elementos de Respuesta , Factor de Transcripción STAT1/inmunología
11.
Tumour Biol ; 33(1): 183-94, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22081310

RESUMEN

Human podoplanin, a type-1 transmembrane sialomucin-like glycoprotein, is involved in cell migration, tumor cell invasion, and metastasis. However, the role of the protein in squamous cell carcinoma (SCC) has been unclear and immunohistochemical reactivity for podoplanin differs from organ-to-organ. In the present study, immunohistochemical and molecular biological analyses were performed to examine the importance of podoplanin expression in oral precancerous and cancerous lesions and metastases. We immunohistochemically investigated the expression of podoplanin in 103 precancerous lesions, 69 primary oral squamous cell carcinomas (OSCCs), and 32 metastases, and that of E-cadherin and vimentin in primary OSCCs with metastasis. Furthermore, human OSCC-derived cell lines preincubated with fibrous growth factor-basic, epidermal growth factor (EGF), and tumor growth factor-ß1 were subjected to real-time reverse transcription polymerase chain reaction. Immunoreactivity for podoplanin was detected in 89 (86.4%) of the precancerous lesions and the intensity was correlated with the degree of epithelial dysplasia (P = 0.016). Enhanced podoplanin expression was observed in 66 (95.7%) of the OSCCs and was significantly associated with a poor pathologic grade of differentiation (P = 0.020). Epithelial-mesenchymal transition was observed in 18 (58.1%) of the primary OSCCs with metastasis to regional lymph nodes. Messenger RNA for podoplanin was markedly increased after treatment with EGF in three OSCC cell lines. The present findings suggest that podoplanin is associated with tumor development via the oral dysplasia-carcinoma sequence and could be involved in a signaling pathway governing tumor growth and invasion in OSCC.


Asunto(s)
Biomarcadores de Tumor/biosíntesis , Carcinoma de Células Escamosas/patología , Glicoproteínas de Membrana/biosíntesis , Neoplasias de la Boca/patología , Boca/patología , Lesiones Precancerosas/patología , Carcinoma de Células Escamosas/metabolismo , Femenino , Humanos , Masculino , Boca/metabolismo , Neoplasias de la Boca/metabolismo , Clasificación del Tumor , Invasividad Neoplásica , Lesiones Precancerosas/metabolismo
12.
Cancers (Basel) ; 3(4): 3726-39, 2011 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-24213108

RESUMEN

Tumor-associated macrophages (TAMs) are a major cellular component in the tumor microenvironment of many solid tumors. The functional competence of TAMs varies depending on the type of tumors and their respective microenvironments. The classically activated M1 macrophages exhibit antitumor functions, whereas the alternatively activated M2 macrophages exhibit protumor functions that contribute to tumor development and progression. Although TAMs have been detected in oral squamous cell carcinoma (OSCC), little is known about their phenotype. In the present study, we performed an immunohistochemical analysis to identify TAMs in surgically resected specimens from 50 patients with OSCC and evaluated the relationship between infiltrated TAMs and the pathological grade of OSCC. Positive staining for CD163, which has been used as a marker for M2 macrophages, was observed in OSCC specimens, and the percentages of CD163+ cells were significantly increased based on the pathological grade. CD163+ cells were detected in the tumor stroma in grade I tumors, whereas an increase in the CD163+ cells in the tumor nest was observed in higher grades of tumors. Although infiltrated CD4+ and CD8+ T cells were detected in all pathological grades of OSCC, no correlation between the infiltrated T cells and the CD163+ TAMs was observed. These results indicate that the infiltrated TAMs in OSCC have an M2 phenotype and that the M2 macrophages may participate in the development of OSCC.

13.
Am J Pathol ; 177(2): 896-907, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20595633

RESUMEN

We have previously isolated GRIM-19, a novel growth suppressor, using a genetic method. GRIM-19 ablates cell growth by inhibiting the transcription factor signal transducer and activator of transcription 3 (STAT3). Up-regulation of STAT3 and growth promotion were observed in a number of human tumors. Although the tumor-suppressive actions of GRIM-19 are known, the structural elements required for its antitumor actions are not understood. Mutational and protein sequence analyses identified a motif in the N terminus of GRIM-19 that exhibited similarity to certain RNA viral proteins. We show that disruption of specific amino acids within this motif cripples the antitumor actions of GRIM-19. These mutants fail to interact with STAT3 efficiently and consequently do not inhibit growth-promoting gene expression. More importantly, we show that a clinically observed mutation in the N terminus of GRIM-19 also weakened its interaction with STAT3 and antitumor action. Together, these studies identify a major role for the N terminus of GRIM-19 in mediating its tumor-suppressive actions.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/química , Proteínas Reguladoras de la Apoptosis/metabolismo , NADH NADPH Oxidorreductasas/química , NADH NADPH Oxidorreductasas/metabolismo , Proteínas Supresoras de Tumor/química , Proteínas Supresoras de Tumor/metabolismo , Secuencias de Aminoácidos/genética , Animales , Apoptosis , Proteínas Reguladoras de la Apoptosis/genética , Análisis Mutacional de ADN , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Ratones Desnudos , NADH NADPH Oxidorreductasas/genética , Trasplante de Neoplasias , Neoplasias/metabolismo , Neoplasias/patología , Factor de Transcripción STAT3/antagonistas & inhibidores , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Proteínas Supresoras de Tumor/genética
14.
Int J Oncol ; 36(6): 1355-65, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20428758

RESUMEN

Interleukin (IL)-23 is a heterodimeric cytokine, comprising IL-12p40 and the cloned IL-23-specific p19 subunit, was identified as a cancer-associated cytokine in a recent study. Like IL-12, IL-23 is expressed predominantly by activated dendritic cells and phagocytic cells. These cytokines antagonistically regulate local inflammatory responses in the tumor microenvironment and infiltration by intraepithelial lymphocytes. We have previously demonstrated the expression of IL-23 and its receptors in human oral squamous cell carcinoma (HOSCC) cell lines and tissue. Hence, this study investigated whether IL-23 has a role in the growth and proliferation of oral cancer cells by examining the expression kinetics of IL-23 and NF-kappaB activity, in vitro and in vivo. IL-23, which constitutively expressed in oral cancer, was enhanced by TNF-alpha and IL-23. IL-23 promotes cell proliferation in oral cancer and enhances the transport of nuclear factor-kappaB (NF-kappaB p65, RelA) to the nucleus in HSC-3 cells. Furthermore, luciferase reporter assay showed that IL-23 strongly induces RelA activity, and confirmed this finding by knockdown of IL-23 using RNA interference. Although RelA activity was down-regulated by anti-human IL-23p19 polyclonal antibody, used to neutralize the activity of IL-23, apoptosis was not induced. Immunohistochemistry revealed a weak IL-23 immunoreactivity in the cytoplasm of inflammatory infiltrating cells and in the cancer cells derived from 14 of 40 cases (35%) of oral SCC. In contrast, strong RelA immunoreactivity was observed in 30 of 40 cases of SCC (75%), especially consistent with IL-23 positive cells in SCC tissues. These data suggest that IL-23 up-regulates the growth and cell proliferation of oral cancer by promoting the nuclear transactivation of RelA.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , Interleucina-23/metabolismo , Neoplasias de la Boca/metabolismo , Factor de Transcripción ReIA/metabolismo , Anciano , Anciano de 80 o más Años , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Proliferación Celular , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Immunoblotting , Masculino , Persona de Mediana Edad , Neoplasias de la Boca/genética , Neoplasias de la Boca/patología , FN-kappa B/genética , FN-kappa B/metabolismo , ARN Interferente Pequeño , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción ReIA/genética , Activación Transcripcional , Transfección , Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Arriba
15.
Biochem Biophys Res Commun ; 387(4): 806-10, 2009 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-19646959

RESUMEN

Tumor hypoxia is associated with tumor promotion, malignant progression, and resistance to cancer therapy. The hypoxia-induced phenotypic changes in tumors result, at least partially, from the induction of hypoxia-responsive genes, such as chemokine receptor CXCR4. Hypoxia-inducible factor 1 (HIF-1) is a critical transcription factor involved in the transcriptional regulation of these genes. Although interferon-gamma (IFNgamma) exerts anti-tumor responses against various tumors, the effect of IFNgamma on HIF-1-dependent transcription remains to be determined. In this study, we examined the inhibitory effect of IFNgamma on hypoxia-induced CXCR4 gene expression in human glioblastoma cell lines and explored the mechanism of inhibition. Hypoxia (1% O(2)) and the iron chelator deferoxamine (DFX), a hypoxia mimetic, increased the levels of CXCR4 mRNA in A172 and T98G cells, and treatment with IFNgamma inhibited the expression of CXCR4 mRNA. Although hypoxia and DFX induced the expression of HIF-1alpha protein and its hypoxia response element (HRE) DNA-binding activity, IFNgamma failed to inhibit its expression or DNA-binding activity. The results of a luciferase reporter assay using a heterologous promoter construct containing the HRE sequence revealed that IFNgamma suppressed the hypoxia- and DFX-induced reporter activities. Lentivirus-mediated RNAi of signal transducer and activator of transcription 1 (STAT1) expression abolished the inhibitory effect of IFNgamma on hypoxia-induced reporter gene activity. Furthermore, this activity was not detected in a stable cell line expressing dominant-negative STAT1. These results indicate that IFNgamma-activated STAT1 functions as a negative regulator of HIF-1-dependent transcription in tumor cells.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Interferón gamma/farmacología , Neoplasias/metabolismo , Receptores CXCR4/genética , Factor de Transcripción STAT1/metabolismo , Hipoxia de la Célula/efectos de los fármacos , Hipoxia de la Célula/genética , Línea Celular Tumoral , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias/genética , Factor de Transcripción STAT1/agonistas , Transcripción Genética/efectos de los fármacos
16.
J Biol Chem ; 284(37): 24869-80, 2009 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-19596857

RESUMEN

Interferon-gamma (IFNgamma) has an antiproliferative effect on a variety of tumor cells. However, many tumor cells resist treatment with IFNs. Here, we show that IFNgamma fails to inhibit the growth of some types of oral squamous cell carcinoma (OSCC) cells that possess a fully functional IFNgamma/STAT1 (signal transducer and activator of transcription-1) signaling pathway. IFNgamma inhibited the growth of the HSC-2, HSC-3, and HSC-4 OSCC cell lines. However, Ca9-22 cells were resistant to IFNgamma despite having intact STAT1-dependent signaling, such as normal tyrosine phosphorylation, DNA binding activity, and transcriptional activity of STAT1. The growth inhibition of HSC-2 cells resulted from S-phase arrest of the cell cycle. IFNgamma inhibited cyclin A2 (CcnA2)-associated kinase activity, which correlated with the IFNgamma-mediated down-regulation of CcnA2 and Cdk2 expression at both the transcriptional and post-transcriptional level in HSC-2 cells but not in Ca9-22 cells. RNAi-mediated knockdown of CcnA2 and Cdk2 resulted in growth inhibition in both cell lines. These results indicate that the resistance of OSCC to IFNgamma is not due simply to the deficiency in STAT1-dependent signaling but results from a defect in the signaling component that mediates this IFNgamma-induced down-regulation of CcnA2 and Cdk2 expression at the transcriptional and post-transcriptional levels.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Resistencia a Antineoplásicos , Regulación Neoplásica de la Expresión Génica , Interferón gamma/metabolismo , Neoplasias de la Boca/metabolismo , Línea Celular Tumoral , Núcleo Celular/metabolismo , Proliferación Celular , Ciclina A/metabolismo , Ciclina A2 , Quinasa 2 Dependiente de la Ciclina/metabolismo , Humanos , Fosforilación , Proteínas Recombinantes/química , Factor de Transcripción STAT1/metabolismo , Transducción de Señal
17.
J Cell Physiol ; 221(1): 40-53, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19492422

RESUMEN

Osteoclasts are bone-resorbing multinucleated cells differentiated from monocyte/macrophage lineage precursors. A novel osteoclast precursor cell line, 4B12 was established from Mac-1(+)c-Fms(+)RANK(+) cells from calvaria of 14-day-old mouse embryos using immunofluorescence and cell-sorting methods. Like M-CSF-dependent bone marrow macrophages (M-BMMs), M-CSF is required for 4B12 cells to differentiate into TRAP-positive multinucleated cells [TRAP(+) MNCs] in the presence of RANKL. Bone-resorbing osteoclasts differentiated from 4B12 cells on dentine slices possess both a clear zone and ruffled borders and express osteoclast-specific genes. Bone-resorbing activity, but not TRAP, was enhanced in the presence of IL-1alpha. The number of TRAP(+) MNCs and the number of pits formed from 4B12 cells on dentine slices was fourfold higher than that from M-BMMs. 4B12 cells were identified as macrophages with Mac-1 and F4/80, yet lost these markers upon differentiation into osteoclasts as determined by confocal laser scanning microscopy. The 4B12 cells do not have the potential to differentiate into dendritic cells indicating commitment to the osteoclast lineage. 4B12 cells are readily transfectable with siRNA transfection before and after differentiation. These data show that 4B12 cells faithfully replicate the properties of primary cells and are a useful and powerful model for analyzing the molecular and cellular regulatory mechanisms of osteoclastogenesis and osteoclast function.


Asunto(s)
Diferenciación Celular , Osteoclastos/citología , Células Madre/citología , Transfección , Fosfatasa Ácida/metabolismo , Animales , Biomarcadores/metabolismo , Resorción Ósea/metabolismo , Encéfalo/citología , Encéfalo/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular , Linaje de la Célula/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Dentina/metabolismo , Embrión de Mamíferos/citología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Isoenzimas/metabolismo , Factor Estimulante de Colonias de Macrófagos/farmacología , Antígeno de Macrófago-1/metabolismo , Macrófagos/citología , Macrófagos/efectos de los fármacos , Ratones , Osteoclastos/efectos de los fármacos , Osteoclastos/patología , ARN Interferente Pequeño/metabolismo , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo , Células Madre/efectos de los fármacos , Fosfatasa Ácida Tartratorresistente
18.
Eur J Immunol ; 38(6): 1559-73, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18493979

RESUMEN

The innate immunity to viral infections induces a potent antiviral response mediated by interferons (IFN). Although IFN-gamma is detected during the acute stages of illness in the upper respiratory tract secretions and in the serum of influenza A virus-infected individuals, control of influenza A virus is not dependent upon IFN-gamma as evidenced by studies using anti-IFN-gamma Ab and IFN-gamma(-/-) mice. Thus, we hypothesized that IFN-gamma is not critical in host survival because influenza A virus has mechanisms to evade the antiviral activity of IFN-gamma. To test this, A549 cells, an epithelial cell line derived from lung adenocarcinoma, were infected with influenza virus strain A/Aichi/2/68 (H3N2) (Aichi) and/or stimulated with IFN-gamma to detect IFN-gamma-stimulated MHC class II expression. Influenza A virus infection inhibited IFN-gamma-induced up-regulation of HLA-DRalpha mRNA and the IFN-gamma induction of class II transactivator (CIITA), an obligate mediator of MHC class II expression. Nuclear translocation of Stat1alpha upon IFN-gamma stimulation was significantly inhibited in influenza A virus-infected cells and this was associated with a decrease in Tyr701 and Ser727 phosphorylation of Stat1alpha. Thus, influenza A virus subverts antiviral host defense mediated by IFN-gamma through effects on the intracellular signaling pathways.


Asunto(s)
Células Epiteliales/virología , Virus de la Influenza A/inmunología , Interferón gamma/farmacología , Janus Quinasa 1/metabolismo , Factor de Transcripción STAT1/metabolismo , Transducción de Señal/efectos de los fármacos , Western Blotting , Línea Celular Tumoral , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Proteínas de Unión al GTP/genética , Expresión Génica/efectos de los fármacos , Antígenos HLA-DR/genética , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Molécula 1 de Adhesión Intercelular/genética , Factor 1 Regulador del Interferón/genética , Factor 3 de Genes Estimulados por el Interferón/metabolismo , Interferón-alfa/farmacología , Janus Quinasa 1/genética , Microscopía Confocal , Modelos Biológicos , FN-kappa B/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosforilación/efectos de los fármacos , Receptores de Interferón/genética , Receptores de Interferón/metabolismo , Mucosa Respiratoria/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transactivadores/genética , Transactivadores/metabolismo , Receptor de Interferón gamma
19.
Oncol Rep ; 19(6): 1421-7, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18497946

RESUMEN

Molecular studies of cylindromas, which arise from the eccrine or apocrine cells of the skin, have demonstrated frequent alterations at chromosome 16q12-13, recently found to house the cylindromatosis (CYLD) gene. CYLD, a tumor suppressor gene, has deubiquitinating enzyme activity and inhibits the activation of transcription factor NF-kappaB. Loss of the deubiquitinating activity of CYLD is correlated with tumorigenesis. It has been reported that the expression of CYLD is observed in various organs. We demonstrated previously that human salivary gland tumor (SGT) cell line, HSG spontaneously expresses CYLD and also found that adenoid cystic carcinoma (ACC) arising from the hard palate was distinctly positive for CYLD, immunohistochemically. However, it is unclear whether loss of CYLD is associated with development of SGTs. This study examined CYLD function in SGT cells and attempted to clarify whether CYLD is associated with development of SGTs. The expression of CYLD and NF-kappaB mRNAs in HSG cells was increased by TNF-alpha. Translocation of NF-kappaB protein from the cytoplasm to the nucleus in HSG cells peaked at 30 min after TNF-alpha stimulation, then decreased at 60 min, whereas that of CYLD protein increased gradually in a time-dependent manner. Luciferase reporter assay indicated that TNF-alpha induced a 5-fold increase of NF-kappaB-dependent transcription at 4 h, which was further enhanced by knockdown of CYLD using RNA interference. Taken together, these data demonstrated that the levels of both CYLD and NF-kappaB mRNAs accumulated in HSG cells during 24 h after TNF-alpha stimulation, although the NF-kappaB activity in the cells was at least negatively regulated by CYLD. Immunohistochemical examinations revealed that there are several correlations between the expression of CYLD and NF-kappaB-related factors in 17 cases of ACC tissues. These findings suggest that loss of CYLD is associated with development of SGTs.


Asunto(s)
Carcinoma Adenoide Quístico/metabolismo , Genes Supresores de Tumor/fisiología , Neoplasias de las Glándulas Salivales/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma Adenoide Quístico/patología , Enzima Desubiquitinante CYLD , Femenino , Humanos , Quinasa I-kappa B/metabolismo , Masculino , Persona de Mediana Edad , FN-kappa B/metabolismo , ARN Mensajero/metabolismo , ARN Interferente Pequeño/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Neoplasias de las Glándulas Salivales/patología , Factor de Transcripción ReIA/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
20.
Biochem Biophys Res Commun ; 350(2): 339-44, 2006 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-17010317

RESUMEN

Sulindac, a non-steroidal anti-inflammatory drug, has been shown to exert an anti-tumor effect on several types of cancer. To determine the effect of sulindac on intracellular signaling pathways in host immune cells such as macrophages, we investigated the effect of the drug on interferon gamma (IFNgamma)-induced expression of signal transducer and activator of transcription 1 (STAT1) and other genes in mouse macrophage-like cell line RAW264.7 cells. Sulindac, but not aspirin or sodium salicylate, inhibited IFNgamma-induced expression of the CXC ligand 9 (CXCL9) mRNA, a chemokine for activated T cells, whereas the interferon-induced expression of CXCL10 or IFN regulatory factor-1 was not affected by sulindac. Luciferase reporter assay demonstrated that sulindac inhibited IFNgamma-induced promoter activity of the CXCL9 gene. Surprisingly, sulindac had no inhibitory effect on IFNgamma-induced STAT1 activation; however, constitutive nuclear factor kappaB activity was suppressed by the drug. These results indicate that sulindac selectively inhibited IFNgamma-inducible gene expression without inhibiting STAT1 activation.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Quimiocinas CXC/genética , Interferón gamma/antagonistas & inhibidores , Macrófagos/efectos de los fármacos , Sulindac/farmacología , Animales , Línea Celular , Quimiocina CXCL9 , Quimiocinas CXC/biosíntesis , Regulación hacia Abajo , Macrófagos/inmunología , Ratones , FN-kappa B/metabolismo , Regiones Promotoras Genéticas , ARN Mensajero/metabolismo , Factor de Transcripción STAT1/metabolismo , Activación Transcripcional
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA