Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Neoplasia ; 45: 100938, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37778252

RESUMEN

TP63 (p63) is strongly expressed in lower-grade carcinomas of the head and neck, skin, breast, and urothelium to maintain a well-differentiated phenotype. TP63 has two transcription start sites at exons 1 and 3' that produce TAp63 and ΔNp63 isoforms, respectively. The major protein, ΔNp63α, epigenetically activates genes essential for epidermal/craniofacial differentiation, including ΔNp63 itself. To examine the specific role of weakly expressed TAp63, we disrupted exon 1 using CRISPR-Cas9 homology-directed repair in a head and neck squamous cell carcinoma (SCC) line. Surprisingly, TAp63 knockout cells having either monoallelic GFP cassette insertion paired with a frameshift deletion allele or biallelic GFP cassette insertion exhibited ΔNp63 silencing. Loss of keratinocyte-specific gene expression, switching of intermediate filament genes from KRT(s) to VIM, and suppression of cell-cell and cell-matrix adhesion components indicated the core events of epithelial-mesenchymal transition. Many of the positively and negatively affected genes, including ΔNp63, displayed local DNA methylation changes. Furthermore, ΔNp63 expression was partially rescued by transfection of the TAp63 knockout cells with TAp63α and application of DNA methyltransferase inhibitor zebularine. These results suggest that TAp63, a minor part of the TP63 gene, may be involved in the auto-activation mechanism of ΔNp63 by which the keratinocyte-specific epigenome is maintained in SCC.


Asunto(s)
Carcinoma de Células Escamosas , Transactivadores , Humanos , Transactivadores/genética , Transición Epitelial-Mesenquimal/genética , Metilación de ADN , Edición Génica , Fosfoproteínas/genética , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
2.
Biomed Res ; 44(1): 1-7, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36682796

RESUMEN

G protein-coupled receptor class C group 5 member B (GPRC5B) is involved in extracellular glucose sensing, glucose metabolism, and insulin resistance. Many cancers require glucose at high concentrations to survive and grow. We have investigated the association between tumour GPRC5B expression and the prognosis for patients with cancer, including head-and-neck squamous cell carcinoma (HNSCC), using data from The Human Protein Atlas. The 5-year survival rate was significantly reduced in patients with HNSCC, gastric, pancreatic, colorectal, and breast cancers if their tumours exhibited high levels of GPRC5B expression. The role of GPRC5B in glucose metabolism was assessed using six HNSCC cell lines with varying levels of GPRC5B expression. High levels of GPRC5B expression were found to favour rapid cell growth. The viability of an HNSCC cell line with normal and transfected GPRC5B expression was also assessed and no differences were observed under standard culture conditions. However, under glucose-deficient culture conditions, GPRC5B-overexpressing cells exhibited increased viability and reduced apoptosis. The results highlight the association between high GPRC5B expression and poor 5-year survival rates in patients with various cancers, including HNSCC. Furthermore, we have demonstrated that GPRC5B supports cancer cell survival under glucose-depleted conditions and could be a target molecule for cancer therapy.


Asunto(s)
Glucosa , Neoplasias de Cabeza y Cuello , Humanos , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Glucosa/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Apoptosis/genética , Neoplasias de Cabeza y Cuello/genética , Línea Celular Tumoral
3.
Proc Natl Acad Sci U S A ; 119(25): e2116027119, 2022 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-35704759

RESUMEN

The epidermis is the outermost layer of the skin and the body's primary barrier to external pathogens; however, the early epidermal immune response remains to be mechanistically understood. We show that the chemokine CXCL14, produced by epidermal keratinocytes, exhibits robust circadian fluctuations and initiates innate immunity. Clearance of the skin pathogen Staphylococcus aureus in nocturnal mice was associated with CXCL14 expression, which was high during subjective daytime and low at night. In contrast, in marmosets, a diurnal primate, circadian CXCL14 expression was reversed. Rhythmically expressed CXCL14 binds to S. aureus DNA and induces inflammatory cytokine production by activating Toll-like receptor (TLR)9-dependent innate pathways in dendritic cells and macrophages underneath the epidermis. CXCL14 also promoted phagocytosis by macrophages in a TLR9-independent manner. These data indicate that circadian production of the epidermal chemokine CXCL14 rhythmically suppresses skin bacterial proliferation in mammals by activating the innate immune system.


Asunto(s)
Epidermis , Inmunidad Innata , Enfermedades Cutáneas Bacterianas , Animales , Quimiocinas CXC/genética , Quimiocinas CXC/inmunología , Relojes Circadianos/inmunología , Epidermis/inmunología , Inmunidad Innata/genética , Inmunidad Innata/inmunología , Queratinocitos/inmunología , Mamíferos , Ratones , Enfermedades Cutáneas Bacterianas/inmunología , Enfermedades Cutáneas Bacterianas/metabolismo , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus/inmunología
4.
Int J Mol Sci ; 20(8)2019 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-31014014

RESUMEN

Cancer is a leading cause of death and disease worldwide, with a tremendous financial impact. Thus, the development of cost-effective novel approaches for suppressing tumor growth and progression is essential. In an attempt to identify the mechanisms responsible for tumor suppression, we screened for molecules downregulated in a cancer progression model and found that the chemokine CXCL14, also called BRAK, was the most significantly downregulated. Increasing the production of CXCL14 protein by transfecting tumor cells with a CXCL14 expression vector and transplanting the cells into the back skin of immunodeficient mice suppressed tumor cell growth compared with that of parental tumor cells, suggesting that CXCL14 suppressed tumor growth in vivo. However, some studies have reported that over-expression of CXCL14, especially in stromal cells, stimulated the progression of tumor formation. Transgenic mice expressing 10-fold more CXCL14 protein than wild-type C57BL/6 mice showed reduced rates of chemical carcinogenesis, transplanted tumor growth, and metastasis without apparent side effects. CXCL14 also acts as an antimicrobial molecule. In this review, we highlight recent studies involving the identification and characterization of CXCL14 in cancer progression and discuss the reasons for the context-dependent effects of CXCL14 on tumor formation.


Asunto(s)
Quimiocinas CXC/metabolismo , Neoplasias/patología , Antiinfecciosos/metabolismo , Antiinfecciosos/farmacología , Antineoplásicos Inmunológicos/uso terapéutico , Candida/efectos de los fármacos , Cetuximab/uso terapéutico , Quimiocinas CXC/genética , Quimiocinas CXC/farmacología , Bacterias Grampositivas/efectos de los fármacos , Humanos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo
5.
Neoplasia ; 21(5): 494-503, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30986748

RESUMEN

TP63 (p63), a member of the tumor suppressor TP53 (p53) gene family, is essential for ectodermal tissue development and suppresses malignant progression of carcinomas. The most abundant isoform, ΔNp63α (referred to as p63), lacks the N-terminal transactivation (TA) domain, and was originally characterized as a dominant-negative type suppressor against p53 family proteins. It also binds to TCF/LEF to inhibit ß-catenin. Nevertheless, transcriptional activation by p63 has also been observed in varied systems. To understand the puzzling results, we analyzed the structure-function relationship of p63 in the control of ß-catenin-dependent transcription. p63 acted as a suppressor of moderately induced ß-catenin. However, when nuclear targeted S33Y ß-catenin was applied to cause the maximum enhancer activation, p63 displayed a ß-catenin-coactivating function. The DNA-binding domain of p63 and the target sequence facilitated it. Importantly, we newly found that, despite the absence of TA domain, p63 was associated with p300, a general adaptor protein and chromatin modifier causing transcriptional activation. C-terminal α domain of p63 was essential for p300-binding and for the coactivator function. These results were related to endogenous p63-p300 complex formation and Wnt/ß-catenin-responsive gene regulation by p63 in squamous cell carcinoma lines. The novel p63-p300 interaction may be involved in positive regulation of gene expression in tissue development and carcinogenesis.


Asunto(s)
Neoplasias Óseas/patología , Proteína p300 Asociada a E1A/metabolismo , Regulación Neoplásica de la Expresión Génica , Osteosarcoma/patología , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo , beta Catenina/metabolismo , Neoplasias Óseas/genética , Neoplasias Óseas/metabolismo , Proteína p300 Asociada a E1A/genética , Humanos , Osteosarcoma/genética , Osteosarcoma/metabolismo , Dominios y Motivos de Interacción de Proteínas , Factores de Transcripción/genética , Transcripción Genética , Activación Transcripcional , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/genética , beta Catenina/genética
6.
Oncotarget ; 8(45): 78312-78326, 2017 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-29108231

RESUMEN

Extracellular acidity is a hallmark of solid tumors and is associated with metastasis in the tumor microenvironment. Acidic extracellular pH (pH e ) has been found to increase intracellular Ca2+ and matrix metalloproteinase-9 (MMP-9) expression by activating NF-κB in the mouse B16 melanoma model. The present study assessed whether TRPM5, an intracellular Ca2+-dependent monovalent cation channel, is associated with acidic pH e signaling and induction of MMP-9 expression in this mouse melanoma model. Treatment of B16 cells with Trpm5 siRNA reduced acidic pH e -induced MMP-9 expression. Enforced expression of Trpm5 increased the rate of acidic pH e -induced MMP-9 expression, as well as increasing experimental lung metastasis. This genetic manipulation did not alter the pH e critical for MMP-9 induction but simply amplified the percentage of inducible MMP-9 at each pH e . Treatment of tumor bearing mice with triphenylphosphine oxide (TPPO), an inhibitor of TRPM5, significantly reduced spontaneous lung metastasis. In silico analysis of clinical samples showed that high TRPM5 mRNA expression correlated with poor overall survival rate in patients with melanoma and gastric cancer but not in patients with cancers of the ovary, lung, breast, and rectum. These results showed that TRPM5 amplifies acidic pH e signaling and may be a promising target for preventing metastasis of some types of tumor.

7.
Cell Cycle ; 15(5): 699-710, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26890356

RESUMEN

Submitted: TP63 (p63), a member of the tumor suppressor TP53 (p53) gene family, is expressed in keratinocyte stem cells and well-differentiated squamous cell carcinomas to maintain cellular potential for growth and differentiation. Controversially, activation of the Wnt/ß-catenin signaling by p63 (Patturajan M. et al., 2002, Cancer Cells) and inhibition of the target gene expression (Drewelus I. et al., 2010, Cell Cycle) have been reported. Upon p63 RNA-silencing in squamous cell carcinoma (SCC) lines, a few Wnt target gene expression substantially increased, while several target genes moderately decreased. Although ΔNp63α, the most abundant isoform of p63, appeared to interact with protein phosphatase PP2A, neither GSK-3ß phosphorylation nor ß-catenin nuclear localization was altered by the loss of p63. As reported earlier, ΔNp63α enhanced ß-catenin-dependent luc gene expression from pGL3-OT having 3 artificial Wnt response elements (WREs). However, this activation was detectable only in HEK293 cells examined so far, and involved a p53 family-related sequence 5' to the WREs. In Wnt3-expressing SAOS-2 cells, ΔNp63α rather strongly inhibited transcription of pGL3-OT. Importantly, ΔNp63α repressed WREs isolated from the regulatory regions of MMP7. ΔNp63α-TCF4 association occurred in their soluble forms in the nucleus. Furthermore, p63 and TCF4 coexisted at a WRE of MMP7 on the chromatin, where ß-catenin recruitment was attenuated. The combined results indicate that ΔNp63α serves as a repressor that regulates ß-catenin-mediated gene expression.


Asunto(s)
Silenciador del Gen , Elementos de Respuesta , Factores de Transcripción/fisiología , Proteínas Supresoras de Tumor/fisiología , Secuencia de Bases , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Sitios de Unión , Cateninas/fisiología , Línea Celular Tumoral , Cromatina/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Metaloproteinasa 7 de la Matriz/metabolismo , Proteína Fosfatasa 2/metabolismo , Factor de Transcripción 4 , Factores de Transcripción/metabolismo , Proteínas Wnt/fisiología , Vía de Señalización Wnt
9.
Sci Rep ; 5: 9083, 2015 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-25765541

RESUMEN

Cancer progression involves carcinogenesis, an increase in tumour size, and metastasis. Here, we investigated the effect of overexpressed CXC chemokine ligand 14 (CXCL14) on these processes by using CXCL14/BRAK (CXCL14) transgenic (Tg) mice. The rate of AOM/DSS-induced colorectal carcinogenesis in these mice was significantly lower compared with that for isogenic wild type C57BL/6 (Wt) mice. When tumour cells were injected into these mice, the size of the tumours that developed and the number of metastatic nodules in the lungs of the animals were always significantly lower in the Tg mice than in the Wt ones. Injection of anti-asialo-GM1 antibodies to the mice before and after injection of tumour cells attenuated the suppressing effects of CXCL14 on the tumor growth and metastasis, suggesting that NK cell activity played an important role during CXCL14-mediated suppression of tumour growth and metastasis. The importance of NK cells on the metastasis was also supported when CXCL14 was expressed in B16 melanoma cells. Further, the survival rates after tumour cell injection were significantly increased for the Tg mice. As these Tg mice showed no obvious abnormality, we propose that CXCL14 to be a promising molecular target for cancer suppression/prevention.


Asunto(s)
Transformación Celular Neoplásica/genética , Quimiocinas CXC/genética , Neoplasias Pulmonares/secundario , Neoplasias/genética , Neoplasias/patología , Animales , Antígenos Ly/inmunología , Autoanticuerpos/inmunología , Transformación Celular Neoplásica/inmunología , Transformación Celular Neoplásica/metabolismo , Quimiocinas CXC/metabolismo , Enfermedad Crónica , Colitis/complicaciones , Colitis/genética , Colitis/inmunología , Modelos Animales de Enfermedad , Femenino , Gangliósido G(M1)/inmunología , Galactosilceramidas/metabolismo , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Depleción Linfocítica , Melanoma Experimental , Ratones , Ratones Transgénicos , Subfamilia B de Receptores Similares a Lectina de Células NK/inmunología , Neoplasias/mortalidad , Carga Tumoral
10.
Biomed Res ; 35(6): 381-8, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25743344

RESUMEN

CXCL14/BRAK (BRAK) is a secreted chemokine with anti-tumor activity, and its expression is suppressed in tumor cells. We previously reported the anti-tumor activity of BRAK in cell lines of head and neck squamous cell carcinoma (HNSCC) and the suppression of BRAK secretion in these cells. BRAK secretion in fibrosarcoma cells is restored by Fasudil, which is a Rho-kinase (ROCK) inhibitor. In this study, we examined the anti-tumor effect of BRAK by evaluating its gene expression and protein secretion in HNSCC cell lines. We found that BRAK mediated the suppressive effect of Fasudil against HNSCC cells. Tumor development in female BALB/cAJclnu/nu mice was suppressed by Fasudil. Also secretion of BRAK protein by tumor cell lines in vitro was significantly stimulated by Fasudil treatment. Similarly, the production of BRAK protein was significantly increased by the addition of Fasudil to cultured tumor cells. Furthermore Fasudil significantly increased BRAK gene expression at the mRNA level in HNSCC cell line. Inhibition of the RhoA/ROCK pathway by siRNAs significantly stimulated BRAK gene expression. These results show that the tumor-suppressive effect of Fasudil was mediated by BRAK, suggesting that Fasudil may therefore be useful for the treatment of HNSCC.


Asunto(s)
1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/análogos & derivados , Carcinoma de Células Escamosas/patología , Quimiocinas CXC/genética , Regulación Neoplásica de la Expresión Génica/fisiología , Neoplasias de Cabeza y Cuello/patología , Inhibidores de Proteínas Quinasas/farmacología , Quinasas Asociadas a rho/antagonistas & inhibidores , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/farmacología , Animales , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Línea Celular Tumoral , Femenino , Neoplasias de Cabeza y Cuello/genética , Neoplasias de Cabeza y Cuello/metabolismo , Humanos , Ratones , Ratones Endogámicos BALB C , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Carcinoma de Células Escamosas de Cabeza y Cuello
11.
J Pharmacol Sci ; 120(3): 241-9, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23099322

RESUMEN

We previously reported that chemokine CXCL14/BRAK (BRAK) has antitumor activity in several carcinoma cells indicating that BRAK secretion suppresses carcinoma cells. Ras-homologous small GTPase (RhoA) and Rho-associated coiled-coil-containing protein kinase (ROCK) are important regulators of secretory processes, and activation of the RhoA/ROCK signaling pathway stimulates tumor invasion and metastasis. We investigated the effects of fasudil, a specific ROCK inhibitor, on BRAK secretion and tumor progression in mesenchymal fibrosarcoma cells (MC57). We demonstrated the antitumor activity of secreted BRAK using MC57 transplantation of BRAK in overexpressed transgenic mice. Further, to eliminate the influence of change in the mRNA expression of endogenous BRAK, we produced stable MC57 cell lines expressing BRAK (MC57-BRAK) or mock vector (MC57-MOCK). Fasudil significantly increased BRAK secretion by MC57-BRAK cells in a dose-dependent manner. To determine the effect of fasudil on tumor growth, MC57-BRAK and MC57-MOCK cells were transplanted into wild-type mice. Fasudil treatment suppressed tumor growth only in mice that had received MC57-BRAK cell transplants. These results indicate that fasudil inhibits fibrosarcoma growth by stimulating BRAK secretion and suggests that fasudil therapy might have clinical efficacy.


Asunto(s)
1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/análogos & derivados , Antineoplásicos/uso terapéutico , Quimiocinas CXC/metabolismo , Fibrosarcoma/tratamiento farmacológico , Proteínas de Neoplasias/metabolismo , Inhibidores de Proteínas Quinasas/uso terapéutico , Quinasas Asociadas a rho/antagonistas & inhibidores , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/farmacología , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/uso terapéutico , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Quimiocinas CXC/genética , Fibrosarcoma/metabolismo , Fibrosarcoma/patología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Trasplante de Neoplasias , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Recombinantes/metabolismo , Transducción de Señal/efectos de los fármacos , Carga Tumoral/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Proteína de Unión al GTP rhoA/metabolismo
12.
Biochem Biophys Res Commun ; 420(2): 217-22, 2012 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-22382027

RESUMEN

The chemokine BRAK/CXCL14 (BRAK) is expressed in normal squamous epithelium, but is not expressed or is expressed at negligible levels in head and neck squamous cell carcinoma. Malignant cells are known to be dedifferentiated compared with normal epithelial cells, suggesting a role for differentiation cues in the expression of BRAK. Thus, we examined the relationship between BRAK expression and stages of differentiation level in epithelial cells. Immunohistochemical analysis showed that BRAK protein was expressed in cells above the spinous cell layer in normal epithelia. In HSC-3 cells in culture, expression of BRAK mRNA was significantly upregulated by cell contact in a cell density-dependent manner, and mRNA expression of cell differentiation markers such as involucrin, cystatin-A, TGM1, TGM3, and TGM5 was concomitantly augmented. Furthermore, the upregulation of BRAK induced by cell contact was suppressed by chlorpromazine, a specific inhibitor of calmodulin. We previously reported that GC boxes and a TATA-like sequence in the BRAK promoter region are associated with the expression of BRAK. Using a promoter assay and ChIP, we demonstrated that binding of the stimulating protein-1 (SP1) transcription factor to a GC box upstream of the BRAK transcription start site was necessary for cell density-dependent upregulation of BRAK. These results indicated that upregulation of BRAK was accompanied by differentiation of epithelial cells induced by calcium/calmodulin signaling, and that SP1 binding to the BRAK promoter region played an important role in this signaling.


Asunto(s)
Calcio/metabolismo , Calmodulina/metabolismo , Diferenciación Celular/genética , Quimiocinas CXC/genética , Células Epiteliales/citología , Regulación de la Expresión Génica , Factor de Transcripción Sp1/metabolismo , Calmodulina/antagonistas & inhibidores , Comunicación Celular/genética , Recuento de Células , Clorpromazina/farmacología , Cistatina A/metabolismo , Células Epiteliales/metabolismo , Humanos , Regiones Promotoras Genéticas , Transducción de Señal , Transglutaminasas/metabolismo , Células Tumorales Cultivadas , Regulación hacia Arriba
13.
ISRN Otolaryngol ; 2012: 797619, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23762619

RESUMEN

In order to find a suppressor(s) of tumor progression in vivo for head and neck squamous cell carcinoma (HNSCC), we searched for molecules downregulated in HNSCC cells when the cells were treated with epidermal growth factor (EGF), whose receptor is frequently overactivated in HNSCC. The expression of BRAK, which is also known as CXC chemokine ligand 14 (CXCL14), was downregulated significantly by the treatment of HNSCC cells with EGF as observed by cDNA microarray analysis followed by reverse-transcriptase polymerase chain reaction analysis and western blotting. The EGF effect on the expression of CXCL14/BRAK was attenuated by the copresence of inhibitors of the EGF receptor, MEK, and ERK. The rate of tumor formation in vivo of BRAK-expressing vector-transfected tumor cells in athymic nude mice or SCID mice was significantly lower than that of mock vector-transfected ones. In addition tumors formed in vivo by the BRAK-expressing cells were significantly smaller than those of the mock-transfected ones. These results indicate that CXCL14/BRAK is a chemokine having suppressive activity toward tumor progression of HNSCC in vivo. Our approach will be useful to find new target molecules to suppress progression of tumors of various origins in addition to HNSCC.

15.
Neurosci Lett ; 487(3): 335-40, 2011 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-20974226

RESUMEN

We immunohistochemically investigated the distribution of CXCL14, also called BRAK protein in the rat hypothalamus using anti-human CXCL14 serum. CXCL14-immunoreactive somata were localized in the periventricular area and paraventricular and supraoptic hypothalamic nuclei. In the former, immunoreactive neuronal somata, confirmed by double staining with a neuronal marker, NeuN, contained diffuse CXCL14-like immunoreactivity in their perikarya. In contrast, immunoreactive somata in the latter contained immunoreactive puncta within their perikarya. Very dense immunoreactive fibers and puncta were seen in the median eminence. Dense immunoreactive fibers were seen in the arcuate nucleus and ventromedial hypothalamic nucleus. Other hypothalamic areas contained a few immunoreactive fibers and puncta. These results demonstrated for the first time that CXCL14 protein is present in a subset of hypothalamic neurons and suggest that CXCL14 participates in hypothalamic functions such as control of autonomic nervous systems and/or participates in immune cell recruitment via the median eminence.


Asunto(s)
Quimiocinas CXC/biosíntesis , Hipotálamo/metabolismo , Neuronas/metabolismo , Animales , Western Blotting , Hipotálamo/inmunología , Inmunohistoquímica , Masculino , Neuronas/inmunología , Ratas , Ratas Wistar
16.
Neoplasia ; 12(12): 969-79, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21170261

RESUMEN

The expression of p63 (TP63/p51) occurs in the basal cells of stratified epithelia and is strongly enhanced at the early stages of squamous cell carcinomas (SCCs) of the head and neck, skin, cervix, and others. We analyzed a promoter/enhancer region (2kΔN) that drives the predominant expression of ΔNp63 for sensitivity to Smad signaling pathways. Reporter assays in HepG2 cells showed a moderate activation of 2kΔN by Smad2 and IκB kinase α (IKKα), partners of the newly identified keratinocyte-specific transforming growth factor ß (TGF-ß) signaling, but not by other Smad molecules. In A431 cells, 2kΔN was activated by Smad2 and IKKα, for which a Smad binding element (SMD2) at -204 was essential. Binding of Smad2 to the chromosomal SMD2 site was detectable. The association of Smad2 with IKKα was evident in the nucleus of A431, accounting for the enhancement of ΔNp63 expression by TGF-ß. Moreover, both ΔNp63 and IKKα were necessary to maintain the noninvasive phenotype of this cell line. FaDu, an invasive, Smad4-deficient SCC, also allowed 2kΔN transactivation by transfected Smad2 in the presence of endogenous IKKα. Reflecting the lack of chromosomal SMD2-Smad2 association and the absence of nuclear IKKα, however, endogenous ΔNp63 was not controlled by TGF-ß or IKKα in FaDu. SCC tissue arrays showed nuclear accumulation of IKKα and p63 intensification in well-differentiated noninvasive lesions. This study indicates that p63 is a target gene of the proposed keratinocyte-specific TGF-ß signal pathway for suppression of the malignant conversion of SCC.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Quinasa I-kappa B/metabolismo , Regiones Promotoras Genéticas , Transducción de Señal , Proteína Smad2/metabolismo , Transactivadores/genética , Factor de Crecimiento Transformador beta/metabolismo , Proteínas Supresoras de Tumor/genética , Western Blotting , Carcinoma de Células Escamosas/genética , Técnica del Anticuerpo Fluorescente , Células Hep G2 , Humanos , Queratinocitos/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transactivadores/metabolismo , Factores de Transcripción , Factor de Crecimiento Transformador beta/genética , Proteínas Supresoras de Tumor/metabolismo
17.
Free Radic Res ; 44(8): 913-24, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20815772

RESUMEN

The present study investigated the effects of oxidative stress induced by reactive oxygen species (ROS), such as hydrogen peroxide (H(2)O(2)) and hydroxyl radical (HO(*)), on the expression of both BRAK , which is also known as non-ELR motif angiostatic CXC chemokine ligand 14 (CXCL14), in head and neck squamous cell carcinoma (HNSCC) cells. When HNSCC cells were cultured in the presence of ROS, the expression of BRAK was significantly decreased whereas that of IL-8 was increased. Interestingly, the effects on the expression of both genes in HNSCC cells were much greater with HO(blacksquare, square, filled) than with H(2)O(2). The effects of ROS on both BRAK and IL-8 expression were attenuated by pre-treatment with N-acetyl-L-cysteine (NAC), epidermal growth factor receptor (EGFR), and mitogen-activated protein kinase (MAPK) inhibitors. These results indicate that oxidative stress induced by H(2)O(2) or HO(*) stimulates angiogenesis and tumuor progression by altering the gene expression of BRAK and IL-8 via the EGFR/MEK/ERK pathway in human HNSCC cells.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Quimiocinas CXC/genética , Neoplasias de Cabeza y Cuello/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Supervivencia Celular/efectos de los fármacos , Quimiocinas CXC/metabolismo , Compuestos Ferrosos/farmacología , Neoplasias de Cabeza y Cuello/genética , Neoplasias de Cabeza y Cuello/patología , Humanos , Peróxido de Hidrógeno/metabolismo , Peróxido de Hidrógeno/farmacología , Radical Hidroxilo/análisis , Radical Hidroxilo/metabolismo , Interleucina-8/biosíntesis , Estrés Oxidativo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Células Tumorales Cultivadas
18.
Biomed Res ; 31(3): 199-206, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20622470

RESUMEN

We reported previously that the forced expression of the chemokine BRAK/CXCL14 in head and neck squamous cell carcinoma cells decreased the rate of tumor formation and size of tumor xenografts in athymic nude mice and SCID mice. In order to clarify the expression of BRAK/CXCL14 affected either the settlement of carcinoma cells in host tissues in vivo or proliferation of the colonized carcinoma cells or both, we prepared oral floor carcinoma-derived HSC-2 cells in which BRAK/CXCL14 expression was induced upon doxycycline treatment. Then 30 nude mice were separated into 3 groups composed of 10 mice per group: Group I, the control, in which the engineered cells were directly xenografted onto the back of the mice; Group II, the cells were xenografted and then the mice were treated with doxycycline; and Group III, the cells were pretreated with doxycycline during culture, and the host mice were also treated with the drug before and after xenografting. The effects of BRAK/CXCL14 expression were examined by measuring the tumor size. The order of the size of tumor xenografts was Group I > II > III, even though the growth rate of the engineered cells was the same whether or not the cells were cultured in the presence of the drug. In addition, the size of tumors was significantly down-regulated after xenografting the doxycycline-pretreated cells in Group III. These data indicate that BRAK/CXCL14 expression in oral floor carcinoma cells reduced both the rate of settlement and the proliferation of the cells in vivo after settlement of the cells.


Asunto(s)
Proliferación Celular , Quimiocinas CXC/metabolismo , Neoplasias de la Boca/metabolismo , Animales , Quimiocinas CXC/genética , Electroforesis en Gel de Poliacrilamida , Humanos , Inmunohistoquímica , Ratones , Ratones Desnudos , Ratones SCID , Neoplasias de la Boca/patología , Trasplante Heterólogo
19.
Biomed Res ; 31(2): 123-31, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20460740

RESUMEN

BRAK/CXCL14 is a chemokine that is expressed in many normal cells and tissues but is absent from or expressed at very low levels in transformed cells and cancerous tissues including head and neck squamous cell carcinoma (HNSCC). We reported previously that the forced expression of BRAK/CXCL14 in HNSCC cells decreased the rate of tumor formation and size of tumor xenografts in athymic nude mice and SCID mice, suggesting that expression level of the gene is important for tumor suppression. In order to study the regulatory mechanisms governing the expression of this gene, we determined the transcriptional start site and promoter motifs of the gene. The major transcriptional start site determined by 5'rapid amplification of cDNA end method was located 283 bp downstream of the first proposed site of the gene. Determination of luciferase activities of reporter gene constructs with various deletions or mutations showed that an atypical TATA-like sequence, TATTAA was essential for the transcription of the gene and that the AP-1 binding sequence and tandem GC box were necessary for stimulating the expression of the gene in human squamous epithelial cells. The human DNA region was highly homologous (95% base identity) to the mouse gene. In addition, okadaic acid, an inhibitor of serine/threonine phosphatases 1, 2A and 2B, stimulated TATTAA sequence and AP-1 binding-sequence dependent promoter activity as well as increased the level of BRAK/CXCL14 mRNA, indicating that these sequences are essential for the regulation of BRAK/CXCL14 gene expression in the cells.


Asunto(s)
Quimiocinas CXC/genética , Quimiocinas/genética , Regiones Promotoras Genéticas , Animales , Secuencia de Bases , Carcinoma de Células Escamosas/genética , ADN Complementario/genética , Genes Reporteros , Humanos , Masculino , Ratones , Ratones Desnudos , Ratones SCID , Neoplasias/genética , ARN Mensajero/genética , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/metabolismo
20.
Biochem Biophys Res Commun ; 396(4): 1060-4, 2010 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-20478268

RESUMEN

The mitogen-activated protein kinase (MAPK) family comprises ERK, JNK, p38 and ERK5 (big-MAPK, BMK1). UV irradiation of squamous cell carcinoma cells induced up-regulation of gene expression of chemokine BRAK/CXCL14, stimulated p38 phosphorylation, and down-regulated the phosphorylation of ERK. Human p38 MAPKs exist in 4 isoforms: p38 alpha, beta, gamma and delta. The UV stimulation of p38 phosphorylation was not inhibited by the presence of SB203580 or PD169316, inhibitors of p38 alpha and beta, suggesting p38 phosphorylation was not dependent on these 2 isoforms and that p38 gamma and/or delta was responsible for the phosphorylation. In fact, inhibition of each of these 4 p38 isoforms by the introduction of short hairpin (sh) RNAs for respective isoforms revealed that only shRNA for p38 delta attenuated the UV-induced up-regulation of BRAK/CXCL14 gene expression. In addition, over-expression of p38 isoforms in the cells showed the association of p38 delta with ERK1 and 2, concomitant with down-regulation of ERK phosphorylation. The usage of p38 delta isoform by UV irradiation is not merely due to the abundance of this p38 isoform in the cells. Because serum deprivation of the cells also induced an increase in BRAK/CXCL14 gene expression, and in this case p38 alpha and/or beta isoform is responsible for up-regulation of BRAK/CXCL14 gene expression. Taken together, the data indicate that the respective stress-dependent action of p38 isoforms is responsible for the up-regulation of the gene expression of the chemokine BRAK/CXCL14.


Asunto(s)
Quimiocinas CXC/genética , Regulación de la Expresión Génica , Expresión Génica/efectos de la radiación , Proteína Quinasa 13 Activada por Mitógenos/metabolismo , Rayos Ultravioleta , Línea Celular Tumoral , Humanos , Proteína Quinasa 13 Activada por Mitógenos/genética , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...