Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 160
Filtrar
1.
Int J Mol Sci ; 22(21)2021 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-34769248

RESUMEN

Bacteroides fragilis enterotoxin (BFT) produced by enterotoxigenic B. fragilis (ETBF) causes colonic inflammation. BFT initially contacts intestinal epithelial cells (IECs) and affects the intestinal barrier. Although molecular components of the gut epithelial barrier such as metalloproteinase-7 (MMP-7) and syndecan-2 are known to be associated with inflammation, little has been reported about MMP-7 expression and syndecan-2 shedding in response to ETBF infection. This study explores the role of BFT in MMP-7 induction and syndecan-2 release in IECs. Stimulating IECs with BFT led to the induction of MMP-7 and the activation of transcription factors such as NF-κB and AP-1. MMP-7 upregulation was not affected by NF-κB, but it was related to AP-1 activation. In BFT-exposed IECs, syndecan-2 release was observed in a time- and concentration-dependent manner. MMP-7 suppression was associated with a reduction in syndecan-2 release. In addition, suppression of ERK, one of the mitogen-activated protein kinases (MAPKs), inhibited AP-1 activity and MMP-7 expression. Furthermore, the suppression of AP-1 and ERK activity was related to the attenuation of syndecan-2 release. These results suggest that a signaling cascade comprising ERK and AP-1 activation in IECs is involved in MMP-7 upregulation and syndecan-2 release during exposure to BFT.


Asunto(s)
Bacteroides fragilis/química , Células Epiteliales/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Mucosa Intestinal/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Metaloproteinasa 7 de la Matriz/biosíntesis , Metaloendopeptidasas/toxicidad , Sindecano-2/metabolismo , Factor de Transcripción AP-1/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Células HCT116 , Humanos , Metaloendopeptidasas/química
2.
Medicine (Baltimore) ; 100(9): e24953, 2021 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-33655961

RESUMEN

ABSTRACT: Colon cancer is one of the most common cancers in the world. To identify the candidate genes in the carcinogenesis and progression of colon cancer, the microarray datasets GSE10950, GSE44861 and GSE74602 were downloaded from the Gene Expression Omnibus (GEO) database. The differentially expressed genes (DEGs) were identified, and functional enrichment analyses were performed. A total of 176 DEGs were identified, consisting of 55 genes upregulated and 121 genes downregulated in colon cancer tissues compared to non-cancerous tissues. The DEGs were mainly enriched in mineral absorption, nitrogen metabolism and complement and coagulation cascades. By using STRING database analysis, we constructed a coexpression network composed of 140 nodes and 280 edges for the DEGs with a combined score >0.4 and a significant interaction relation. Thirteen hub genes were identified, and poor OS of patients was only associated with high expression of Matrix Metallopeptidase 7 (MMP7), which may be involved in the carcinogenesis, invasion or recurrence of colon cancer. In conclusion, we propose that the DEGs and hub genes identified in the present study may be regarded as diagnostic biomarkers for colon cancer. Moreover, the overexpression of MMP7 may correlate with poor prognosis.


Asunto(s)
Neoplasias del Colon/genética , Biología Computacional/métodos , Regulación Neoplásica de la Expresión Génica , Metaloproteinasa 7 de la Matriz/genética , Biomarcadores de Tumor/biosíntesis , Biomarcadores de Tumor/genética , Neoplasias del Colon/diagnóstico , Neoplasias del Colon/metabolismo , ADN de Neoplasias/genética , Bases de Datos Genéticas , Progresión de la Enfermedad , Perfilación de la Expresión Génica/métodos , Humanos , Metaloproteinasa 7 de la Matriz/biosíntesis , Análisis por Micromatrices , Pronóstico
3.
Toxicology ; 452: 152717, 2021 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-33581214

RESUMEN

The effects of asbestos on immunocompetent cells have been investigated. In particular, attention was paid to regulatory T cell function, which was observed using the HTLV-1 immortalized human polyclonal T cell line MT-2. Exposure to asbestos (approximately more than 25 µg/mL for 1-3 day) induced apoptosis, and we observed an increase in regulatory T cell function and acceleration of the cell cycle with continuous exposure to low concentrations of asbestos (5-10 µg/mL for more than eight months). Furthermore, cDNA microarray analysis in this study revealed that expression of matrix metalloproteinase-7 (MMP-7) was markedly higher in exposed sublines compared to original MT-2 cells. It was determined that MMP-7 had no effect on Treg function, as determined by examination of sublines and by addition of recombinant MMP-7 and neutralizing antibodies or inhibitors of MMP-7. However, when examining melting of the extracellular matrix (an MMP-7-mediated event) or the extent to which the MT-2 parent strain or long-term exposed subline cells pass through a fibronectin-coated filter, more filter passes were observed for the subline. These results suggest that the effect of asbestos fibers on Treg cells results in excessive migration of the tumor microenvironment through hypersecretion of MMP-7 together with an increase in suppressive function and enhancement of cell cycle progression. Therefore, one possible way to prevent the development of asbestos-induced cancer is to reduce the function (including MMP-7 production) or amount of Treg cells by physiologically active substances or food ingredients. Alternatively, it may be possible to invoke immune checkpoint treatments when carcinogenesis occurs.


Asunto(s)
Amianto/toxicidad , Carcinógenos/toxicidad , Movimiento Celular/efectos de los fármacos , Metaloproteinasa 7 de la Matriz/biosíntesis , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/metabolismo , Línea Celular Transformada , Movimiento Celular/fisiología , Humanos
4.
Anticancer Res ; 40(6): 3097-3108, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32487604

RESUMEN

BACKGROUND/AIM: C-C motif chemokine ligand 18 (CCL18) is overexpressed in the microenvironment of tumors, promotes invasion and metastasis and is thus important for the therapeutic outcome of many tumor entities. The Gs-coupled seven-transmembrane receptor GPR30 is known as both a CCL18 and an estrogen receptor; its activation by estradiol leads to a transactivation of membrane-tethered pro-heparin-binding EGF-like growth factor and the MAPK/ERK pathway. We examined whether this signaling pathway remains the same under CCL18 stimulation, as opposed to estradiol stimulation. MATERIALS AND METHODS: We investigated the effects of CCL18 on the lung cancer cell line A549, that show low GPR30 expression and the breast cancer cell lines MCF-7, that has high GPR30 expression and MDA-MB-231. These cells were stimulated in different media with CCL18 and then analyzed by qPCR, In-Cell Western®, western blot and ELISA. RESULTS: Many similarities on the effect of CCL18 on the already known estradiol-activated signaling pathway via the G protein-coupled estrogen receptor GPR30 were identified. GPR30 is involved in the expression of matrix metalloproteinases (MMPs), which may play a role in the transactivation of ERK-1/-2 via the cleavage of membrane-bound HB-EGF, via Src-related tyrosine kinases and Gßγ-subunits. With increasing CCL18 concentration, the expression of MMP7 decreased in A549 cells. With decreasing estrogen content of the medium, there was an increasing effect of CCL18 on the inhibition of the relative expression of MMP7. Inhibition of GPR30 with G15 also resulted in a decrease in the relative expression of MMP7, irrespective of the subsequent stimulation with CCL18. This is a rather unexpected result, because the estrogen estradiol and CCL18 both activate GPR30. MCF-7 cells which express more GPR30 did not show any dependence of the relative MMP7 expression on CCL18 except in estrogen-free FCS medium. CCL18 induced an increased relative ERK activation in In-Cell western (ICW) at A549 cells. Stimulation with CCL18 caused decreased ERK activation with simultaneous inhibition of adenylate cyclase in MCF-7. However, stimulation with CCL18 and simultaneous inhibition of cyclooxygenase in MCF-7 resulted in increased ERK activation. In A549, stimulation with CCL18 and co-incubation with dbcAMP resulted in decreased ERK activation in both ICW and Western blot. CONCLUSION: In summary, the Gs-coupled receptor GPR30 plays an important role in the signaling pathway of CCL18. CCL18 and estradiol may not lead to the same signaling pathway after activating GPR30.


Asunto(s)
Quimiocinas CC/metabolismo , Estradiol/metabolismo , Metaloproteinasa 7 de la Matriz/metabolismo , Receptores de Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Células A549 , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Quimiocinas CC/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Gliceraldehído-3-Fosfato Deshidrogenasa (Fosforilante)/metabolismo , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Células MCF-7 , Metaloproteinasa 7 de la Matriz/biosíntesis , Metaloproteinasa 7 de la Matriz/genética , Fosforilación , ARN Largo no Codificante/biosíntesis , ARN Largo no Codificante/genética , Receptores de Estrógenos/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Transducción de Señal
5.
J Virol ; 94(6)2020 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-31827001

RESUMEN

Herpes simplex virus 1 (HSV-1) can infect virtually all cell types in vitro An important reason lies in its ability to exploit heparan sulfate (HS) for attachment to cells. HS is a ubiquitous glycosaminoglycan located on the cell surface and tethered to proteoglycans such as syndecan-1. Previously, we have shown that heparanase (HPSE) facilitates the release of viral particles by cleaving HS. Here, we demonstrate that HPSE is a master regulator where, in addition to directly enabling viral release via HS removal, it also facilitates cleavage of HS-containing ectodomains of syndecan-1, thereby further enhancing HSV-1 egress from infected cells. Syndecan-1 cleavage is mediated by upregulation of matrix metalloproteases (MMPs) that accompanies higher HPSE expression in infected cells. By overexpressing HPSE, we have identified MMP-3 and MMP-7 as important sheddases of syndecan-1 shedding in corneal epithelial cells, which are natural targets of HSV-1 infection. MMP-3 and MMP-7 were also naturally upregulated during HSV-1 infection. Altogether, this paper shows a new connection between HSV-1 release and syndecan-1 shedding, a phenomenon that is regulated by HPSE and executed by the MMPs. Our results also identify new molecular markers for HSV-1 infection and new targets for future interventions.IMPORTANCE HSV-1 is a common cause of recurrent viral infections in humans. The virus can cause a range of mucosal pathologies. Efficient viral egress from infected cells is an important step for HSV-1 transmission and virus-associated pathologies. Host mechanisms that contribute to HSV-1 egress from infected cells are poorly understood. Syndecan-1 is a common heparan sulfate proteoglycan expressed by many natural target cells. Despite its known connection with heparanase, a recently identified mediator of HSV-1 release, syndecan-1 has not been previously investigated in HSV-1 release. In this study, we demonstrate that the shedding of syndecan-1 by MMP-3 and MMP-7 supports viral egress. We show that the mechanism behind the activation of these MMPs is mediated by heparanase, which is upregulated upon HSV-1 infection. Our study elucidates a new connection between HSV-1 egress, heparanase, and matrix metallopeptidases; identifies new molecular markers of infection; and provides potential new targets for therapeutic interventions.


Asunto(s)
Glucuronidasa/metabolismo , Herpesvirus Humano 1/metabolismo , Sindecano-1/metabolismo , Liberación del Virus , Esparcimiento de Virus , Línea Celular , Regulación Enzimológica de la Expresión Génica , Glucuronidasa/genética , Humanos , Metaloproteinasa 3 de la Matriz/biosíntesis , Metaloproteinasa 3 de la Matriz/genética , Metaloproteinasa 7 de la Matriz/biosíntesis , Metaloproteinasa 7 de la Matriz/genética , Sindecano-1/genética , Regulación hacia Arriba
6.
Cell Host Microbe ; 25(5): 706-718.e7, 2019 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-31053533

RESUMEN

Metabolic programs and host defense are highly integrated to ensure proper immune responses during stress. Central to these responses, mTOR regulates immune functions by sensing and integrating environmental cues, yet how these systems are coordinated at the intestinal surface remains undefined. We show that the antimicrobial peptide α-defensin is functionally sustained during nutrient deprivation because of regulation of the defensin-processing enzyme MMP7 by microbiota- and host-derived factors. Unlike other antimicrobial peptides, the MMP7-α-defensin axis remains active during nutrient fluctuations, providing essential protection against enteric pathogens. Sustained Mmp7 expression requires the microbiota and is mediated by de-repression of the transcription activator Atoh1 upon attenuation of the transcriptional repressor Hes1 in intestinal epithelial cells. Hes1 levels are regulated via mTOR and controlled translationally, constituting a metabolism-translation-transcription loop. Disrupting this loop by supplying nutrients paradoxically compromises antibacterial defense. Together, these results uncover a regulatory circuit that couples host nutrient status to epithelial antimicrobial immunity.


Asunto(s)
Células Epiteliales/inmunología , Regulación de la Expresión Génica , Inmunidad Mucosa , Metaloproteinasa 7 de la Matriz/biosíntesis , Nutrientes/metabolismo , Factor de Transcripción HES-1/metabolismo , alfa-Defensinas/biosíntesis , Animales , Línea Celular , Células Epiteliales/efectos de los fármacos , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/inmunología , Ratones Endogámicos C57BL
7.
J Biol Chem ; 294(21): 8529-8542, 2019 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-30967475

RESUMEN

The farnesoid X receptor (FXR) is a member of the nuclear receptor superfamily of bile acid-activated transcription factors and an important regulator of cell proliferation, apoptosis, and Wnt signaling. Down-regulated expression of FXR plays an important role in some malignancies such as colon cancer, and in rodent models of intestinal neoplasia, FXR knockout increases the size and number of colon tumors. These previous observations implicate FXR as a tumor suppressor, but the underlying molecular mechanisms are unclear. Employing complementary experimental approaches and using human colon cancer specimens, human and murine colon cancer cell lines, and FXR transgenic mice, here we identified an additional, potentially important role for FXR. We observed an inverse relationship between the expression of FXR and matrix metalloproteinase-7 (MMP7), a collagenase and signaling molecule consistently associated with colon cancer progression. We noted that FXR gene ablation increases MMP7 expression. Consistent with this finding, FXR overexpression and a dominant-negative FXR mutation reduced and augmented, respectively, MMP7 expression. Of note, MMP7 was the only MMP gene family member whose expression was down-regulated after FXR activation. FXR-mediated regulation of MMP7 transcription did not require heterodimerization with the retinoid X receptor (RXR), indicating that FXR represses MMP7 expression independently of RXR. Last, we uncovered that FXR suppresses MMP7 transcription by binding to a negative FXR-responsive element in the 5' MMP7 promoter, an event that inhibited colon cancer cell proliferation and invasion. These findings identify the FXR-MMP7 axis as a potential therapeutic target for managing colon cancer.


Asunto(s)
Neoplasias del Colon/metabolismo , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Metaloproteinasa 7 de la Matriz/biosíntesis , Proteínas de Neoplasias/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Células CACO-2 , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Células HCT116 , Células HT29 , Humanos , Metaloproteinasa 7 de la Matriz/genética , Ratones , Ratones Noqueados , Proteínas de Neoplasias/genética , Receptores Citoplasmáticos y Nucleares/genética , Elementos de Respuesta
8.
EMBO Mol Med ; 10(8)2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29941541

RESUMEN

The clinical management of pancreatic ductal adenocarcinoma (PDAC) is hampered by the lack of reliable biomarkers. This study investigated the value of soluble stroma-related molecules as PDAC biomarkers. In the first exploratory phase, 12 out of 38 molecules were associated with PDAC in a cohort of 25 PDAC patients and 16 healthy subjects. A second confirmatory phase on an independent cohort of 131 PDAC patients, 30 chronic pancreatitis patients, and 131 healthy subjects confirmed the PDAC association for MMP7, CCN2, IGFBP2, TSP2, sICAM1, TIMP1, and PLG Multivariable logistic regression model identified biomarker panels discriminating respectively PDAC versus healthy subjects (MMP7 + CA19.9, AUC = 0.99, 99% CI = 0.98-1.00) (CCN2 + CA19.9, AUC = 0.96, 99% CI = 0.92-0.99) and PDAC versus chronic pancreatitis (CCN2 + PLG+FN+Col4 + CA19.9, AUC = 0.94, 99% CI = 0.88-0.99). Five molecules were associated with PanIN development in two GEM models of PDAC (PdxCre/LSL-KrasG12D and PdxCre/LSL-KrasG12D/+/LSL-Trp53R172H/+), suggesting their potential for detecting early disease. These markers were also elevated in patient-derived orthotopic PDAC xenografts and associated with response to chemotherapy. The identified stroma-related soluble biomarkers represent potential tools for PDAC diagnosis and for monitoring treatment response of PDAC patients.


Asunto(s)
Biomarcadores de Tumor/sangre , Carcinoma Ductal Pancreático/diagnóstico , Neoplasias Pancreáticas/diagnóstico , Adulto , Anciano , Anciano de 80 o más Años , Animales , Carcinoma Ductal Pancreático/sangre , Carcinoma Ductal Pancreático/tratamiento farmacológico , Estudios de Cohortes , Factor de Crecimiento del Tejido Conjuntivo/biosíntesis , Factor de Crecimiento del Tejido Conjuntivo/sangre , Femenino , Humanos , Molécula 1 de Adhesión Intercelular/biosíntesis , Molécula 1 de Adhesión Intercelular/sangre , Masculino , Metaloproteinasa 7 de la Matriz/biosíntesis , Metaloproteinasa 7 de la Matriz/sangre , Persona de Mediana Edad , Neoplasias Pancreáticas/sangre , Neoplasias Pancreáticas/tratamiento farmacológico , Pronóstico , Solubilidad , Células del Estroma/metabolismo , Trombospondinas/biosíntesis , Trombospondinas/sangre , Inhibidor Tisular de Metaloproteinasa-1/biosíntesis , Inhibidor Tisular de Metaloproteinasa-1/sangre , Microambiente Tumoral/fisiología
9.
Anticancer Drugs ; 29(6): 530-538, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29553945

RESUMEN

Colorectal cancer (CRC) is one of the most frequent malignant tumors. Signaling by the PI3K/AKT pathway is crucial for CRC development and progression, including proliferation and migration. Celastrol has an anticancer effect, but its mechanism needs to be determined. Here, we showed that celastrol suppressed CRC cell proliferation and migration. Celastrol treatment also decreased the PI3K/AKT pathway components, and MMP3 and MMP7 expression levels. In addition, knockdown of AKT, not mTOR, inhibited MMP3 and MMP7 expression levels and AKT silencing promoted the celastrol-induced effects on CRC cell proliferation and migration. Taken together, these findings indicated that the celastrol-induced antitumor effects were mediated through MMP3 and MMP7 by the PI3K/AKT signaling pathway.


Asunto(s)
Neoplasias Colorrectales/tratamiento farmacológico , Metaloproteinasa 3 de la Matriz/metabolismo , Metaloproteinasa 7 de la Matriz/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Triterpenos/farmacología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Neoplasias Colorrectales/enzimología , Neoplasias Colorrectales/patología , Células HCT116 , Humanos , Metaloproteinasa 3 de la Matriz/biosíntesis , Metaloproteinasa 3 de la Matriz/genética , Metaloproteinasa 7 de la Matriz/biosíntesis , Metaloproteinasa 7 de la Matriz/genética , Inhibidores de la Metaloproteinasa de la Matriz/farmacología , Triterpenos Pentacíclicos , Fosfatidilinositol 3-Quinasas/biosíntesis , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/biosíntesis , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/biosíntesis , Regulación hacia Arriba/efectos de los fármacos
10.
In Vivo ; 32(1): 133-138, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29275310

RESUMEN

BACKGROUND/AIM: Matrix metalloproteinase 7 (MMP7) expression is highly associated with colorectal cancer and modulates tumour growth and invasion. Radiation injury induces inflammation with increases in MMP7 and in transforming growth factor beta (TGFß). The aim of this study was to investigate the effect on MMP7 and TGFß. expression in patients with rectal cancer undergoing different regimens of neoadjuvant radiotherapy (RT). PATIENTS AND METHODS: We studied 53 patients in three RT treatment groups receiving RT of 25 Gy, long-term RT 50 Gy and controls receiving no RT. Three biopsies were obtained from each patient during the treatments: before RT, after RT and after surgery. Tissue samples were formalin fixed, paraffin embedded and tissue microarrays were constructed and stained for MMP7 and TGFß. Mann-Whitney U-tests and Wilcoxon Z-tests were used to determine differences between patients before and after RT, and after surgery, as well as between the RT groups. RESULTS: In all three patient groups, increases of MMP7 and TGFß expression were observed after surgery. MMP7 expression was significantly increased in patients receiving short-term RT but TGFß expression was not affected by RT. CONCLUSION: 50 Gy Irradiation of rectal cancer gives less tumour activation of MMP7, whilst it is up-regulated by 25 Gy and surgery regardless of RT.


Asunto(s)
Metaloproteinasa 7 de la Matriz/biosíntesis , Neoplasias del Recto/radioterapia , Recto/efectos de la radiación , Anciano , Relación Dosis-Respuesta en la Radiación , Femenino , Humanos , Inmunohistoquímica , Masculino , Terapia Neoadyuvante/métodos , Radioterapia Adyuvante/métodos , Neoplasias del Recto/metabolismo , Neoplasias del Recto/patología , Recto/metabolismo , Recto/patología , Factores de Tiempo , Análisis de Matrices Tisulares , Factor de Crecimiento Transformador beta/biosíntesis
11.
Tumour Biol ; 39(6): 1010428317706054, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28635400

RESUMEN

Long noncoding RNAs play an important role in various biological processes, including tumorigenesis. FOXC1 (Forkhead box C1) is a member of the Forkhead box family of transcription factors and plays a crucial role in nasopharyngeal carcinoma. In this study, a novel long noncoding RNA (FOXCUT) located upstream of FOXC1 was investigated in 42 nasopharyngeal carcinoma patients. Our analysis revealed that the expression levels of FOXCUT and FOXC1 in nasopharyngeal carcinoma tissues were significantly higher than those observed in chronic nasopharyngitis tissues and that FOXCUT expression was positively correlated with FOXC1 expression. Additionally, knockdown of FOXCUT significantly inhibited proliferation and migration of nasopharyngeal carcinoma cell lines and resulted in downregulated expression of the matrix metalloproteinase 7 and matrix metalloproteinase 9, as well as vascular endothelial growth factor A and ß-catenin. Our findings suggested that FOXCUT expression contributed to the development and progression of nasopharyngeal carcinoma by targeting FOXC1 and that FOXCUT might be useful as a potential nasopharyngeal carcinoma biomarker and therapeutic target.


Asunto(s)
Biomarcadores de Tumor/genética , Carcinoma/genética , Factores de Transcripción Forkhead/genética , Neoplasias Nasofaríngeas/genética , ARN Largo no Codificante/genética , Adulto , Biomarcadores de Tumor/biosíntesis , Carcinoma/patología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Femenino , Factores de Transcripción Forkhead/biosíntesis , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Metaloproteinasa 7 de la Matriz/biosíntesis , Metaloproteinasa 9 de la Matriz/biosíntesis , Persona de Mediana Edad , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/patología , ARN Largo no Codificante/biosíntesis , Factor A de Crecimiento Endotelial Vascular/biosíntesis , beta Catenina/biosíntesis
12.
Sci Rep ; 7(1): 3869, 2017 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-28634370

RESUMEN

Metalloproteinases (MMPs) contribute to tissue remodeling and acute inflammation not only by degrading extracellular matrix proteins but also by controlling the influx of chemokines through the regulation and shedding of syndecans. B-lymphocytes, in addition to their well-known function as antibody producing cells, participate in the innate immune response by secreting inflammatory cytokines and chemokines. However, there is little information about the role of B-lymphocytes in the regulation of MMPs; consequently, herein we investigated whether activated human circulating B-lymphocytes contributed to the secretion of MMPs. We demonstrate that B-lymphocytes activated by un-methylated CpG motifs, found in bacterial DNA, and ß-glucans, found in the cell wall of fungi, both induced MMP-7. Interestingly, while CpG-stimulated cells activated the mTOR pathway via TLR9 receptor to induced MMP-7, ß-glucan-stimulated cells were mTOR-independent and used Dectin-1 receptor. B-lymphocytes did not seem to have a major role in the secretion of tissue inhibitors of metalloproteinases (TIMPs). However, secreted MMP-7 participated in the shedding of Syndecan-4 from the surface of B-lymphocytes. In conclusion, circulating human B-lymphocytes contribute to the regulation of the innate immune system by participating in the secretion of MMP-7 which in turn is important for the shedding of Syndecan-4 in response to infectious stimuli.


Asunto(s)
Antígenos/inmunología , Linfocitos B/inmunología , Linfocitos B/metabolismo , Metaloproteinasa 7 de la Matriz/biosíntesis , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Humanos , Leucocitos/inmunología , Leucocitos/metabolismo , Metaloproteinasas de la Matriz/metabolismo , Receptores de Antígenos de Linfocitos B/metabolismo , Inhibidores Tisulares de Metaloproteinasas/metabolismo , Receptores Toll-Like/metabolismo , beta-Glucanos/metabolismo
13.
Acta Biochim Pol ; 64(2): 295-299, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28510611

RESUMEN

INTRODUCTION: Tissue inhibitors of metalloproteinases (TIMP) and the matrix metalloproteinases (MMP) are involved in the spread of cancer. METHODS: We have evaluated the matrix metalloproteinases' (MMP-10, MMP-7) and their inhibitors' (tissue inhibitors of metalloproteinases - TIMP-1, TIMP-2) mRNA expression in 61 esophageal cancer samples from patients who had undergone surgery, by using real-time quantitative RT-PCR, and correlated the results with the patient clinicopathologic features. RESULTS: MMP-10, MMP-7, TIMP-1, TIMP-2 were overexpressed in 73%, 85%, 55% and 42% of esophageal cancer samples, respectively. The expression of MMP-10, TIMP-1, and TIMP-2 correlated with the tumor size. The MMP-7 overexpression was associated with the tumour stage (I, II vs III, p=0.05) and lymph node metastasis (N0 vs N1, p=0.037). CONCLUSIONS: We conclude that in the resected esophageal cancer an increased mRNA expression of MMP-7, MMP-10 and TIMP-1 correlated with clinicopathologic features. We suggest that these genes may play a role during progression of the disease.


Asunto(s)
Neoplasias Esofágicas/genética , Metaloproteinasa 10 de la Matriz/biosíntesis , Metaloproteinasa 7 de la Matriz/biosíntesis , Inhibidor Tisular de Metaloproteinasa-1/biosíntesis , Inhibidor Tisular de Metaloproteinasa-2/biosíntesis , Anciano , Neoplasias Esofágicas/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Metástasis Linfática , Masculino , Metaloproteinasa 10 de la Matriz/genética , Metaloproteinasa 7 de la Matriz/genética , Persona de Mediana Edad , Inhibidor Tisular de Metaloproteinasa-1/genética , Inhibidor Tisular de Metaloproteinasa-2/genética
14.
Minerva Med ; 108(1): 13-19, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27827523

RESUMEN

BACKGROUND: MicroRNAs (miRNAs) play critical roles in the development and progression of human malignancy. The levels of miR-143 microRNA are lower in malignant tumors - including colorectal cancer (CRC) - as it is a tumor suppressor. However, the potential mechanism of miR-143 in CRC remains largely unknown. METHODS: Target prediction programs and luciferase reporter assay was used to predict the targets of miR-143. Following overexpression of miR-143 in CRC cells, target gene matrix metalloproteinase 7 (MMP7) expression was detected by quantitative real-time PCR (qRT-PCR) and western blot. In addition, the expression of MMP7 was quantified in CRC tissues and cell lines. Moreover, we determined the effect of MMP7 on CRC cell proliferation and invasion. RESULTS: In the present study, TargetScan predicted that miR-143 could directly bind to 3'-UTR of MMP7 mRNA, and luciferase reporter assay further supported the hypothesis that MMP7 might act as a direct target gene of miR-143. Our data showed that increased expression of miR-143 repressed MMP7 expression in CRC cells both in mRNA and protein levels. Furthermore, qRT-PCR showed that the expression of MMP7 was increased in CRC tissues and cell lines, and inversely correlated with miR-143 expression in CRC tissues. Finally, our results indicated that increased expression of MMP7 reversed the potential influence of miR-143 on CRC cell proliferation and invasion ability. CONCLUSIONS: Our results indicated that miR-143 might act as a tumor suppressor by targeting MMP7 during the development of CRC.


Asunto(s)
Neoplasias Colorrectales/patología , Metaloproteinasa 7 de la Matriz/fisiología , MicroARNs/genética , Proteínas de Neoplasias/fisiología , ARN Neoplásico/genética , Regiones no Traducidas 3'/genética , División Celular , Línea Celular Tumoral , Neoplasias Colorrectales/enzimología , Neoplasias Colorrectales/genética , Regulación Neoplásica de la Expresión Génica , Genes Reporteros , Humanos , Metaloproteinasa 7 de la Matriz/biosíntesis , Metaloproteinasa 7 de la Matriz/genética , Invasividad Neoplásica , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética
15.
Tumour Biol ; 37(4): 5225-30, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26553362

RESUMEN

Isocitrate dehydrogenase 2 (IDH2) is a mitochondrial NADP-dependent isocitrate dehydrogenase and has been found to be a tumor suppressor in several types of tumors. However, the roles of IDH2 in hepatocellular carcinoma (GC) as well as underlying mechanisms remain unknown. Here, the IDH2 and matrix metalloproteinase 7 (MMP7) levels in the specimens from 30 GC patients were investigated by Western blot and ELISA, respectively. Their relationship was examined by correlation analyses. Patient survival with high IDH2 levels and low IDH2 levels was compared. IDH2 levels, and MMP7 levels were modified in a human GC cell line. The effects of IDH2 or MMP7 modulation on the expression of each other were analyzed. The dependence of nuclear factor κB (NF-κB) signaling was examined using a specific inhibitor. We found that the IDH2 levels significantly decreased in GC, and were even lower in GC with metastases, compared to those without metastases. IDH2 levels inversely correlated with MMP7 levels in GC. GC patients with low IDH2 had lower 5-year survival. MMP7 levels did not regulate IDH2 levels, while IDH2 inhibited MMP7 levels in GC cells, in a NF-κB signaling dependent manner. Together, these data suggest that IDH2 may be a tumor suppressor in that its loss may promote malignant progression of GC via NF-κB-dependent increases in MMP7 activity.


Asunto(s)
Carcinogénesis/genética , Isocitrato Deshidrogenasa/biosíntesis , Metaloproteinasa 7 de la Matriz/genética , Neoplasias Gástricas/genética , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Isocitrato Deshidrogenasa/genética , Estimación de Kaplan-Meier , Masculino , Metaloproteinasa 7 de la Matriz/biosíntesis , FN-kappa B/genética , Invasividad Neoplásica/genética , Metástasis de la Neoplasia , Transducción de Señal/genética , Neoplasias Gástricas/patología
16.
Mol Med Rep ; 13(2): 1263-8, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26648518

RESUMEN

During progression of gastric cancer, degradation of the extracellular matrix by matrix metalloproteinases (MMPs) has been associated with poor prognosis. Tanshinone IIA (Tan-IIA) exerts antitumor activity in a variety of human cancer cells. It is extracted from Danshen (Salviae miltiorrhizae radix), and induces apoptosis and inhibits the proliferation of gastric cancer cells. However, the molecular mechanisms underlying the inhibition of migration in gastric cancer by Tan-IIA have not been fully elucidated. In the present study, AGS cell migration ability was evaluated using a wound-healing assay. The protein expression levels of nuclear factor (NF)-κB-p65, cyclooxygenase (COX)-2, MMP-2, -7, and -9 and ß-actin in AGS cells were measured by western blotting. The results demonstrated that AGS cells treated with Tan-IIA exhibit decreased protein expression levels of NF-κB-p65, COX-2, and MMP-2, -7 and -9. The results also indicate that Tan-IIA inhibits migration ability in a dose- and time-dependent manner. These findings demonstrate that Tan-IIA inhibits the migration ability of AGS human gastric cancer cells and that decreasing the protein expression of NF-κB-p65, COX-2, and MMP-2, -7 and -9 may be an underlying molecular mechanism.


Asunto(s)
Abietanos/administración & dosificación , Ciclooxigenasa 2/biosíntesis , Metaloproteinasa 2 de la Matriz/biosíntesis , Metaloproteinasa 7 de la Matriz/biosíntesis , Metaloproteinasa 9 de la Matriz/biosíntesis , Neoplasias Gástricas/tratamiento farmacológico , Factor de Transcripción ReIA/biosíntesis , Abietanos/química , Actinas/biosíntesis , Actinas/genética , Antineoplásicos Fitogénicos/administración & dosificación , Antineoplásicos Fitogénicos/química , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Ciclooxigenasa 2/genética , Medicamentos Herbarios Chinos/administración & dosificación , Medicamentos Herbarios Chinos/química , Regulación Neoplásica de la Expresión Génica , Humanos , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 7 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/genética , Salvia miltiorrhiza , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Factor de Transcripción ReIA/genética
17.
Tumour Biol ; 37(4): 5001-11, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26537584

RESUMEN

Wnt/ß-catenin signaling pathway plays a major role in the cancer metastasis. Several microRNAs (miRNAs) are contributed to the inhibition of breast cancer metastasis. Here, we attempted to find novel targets and mechanisms of microRNA-100 (miR-100) in regulating the migration and invasion of breast cancer cells. In this study, we found that miR-100 expression was downregulated in human breast cancer tissues and cell lines. The overexpression of miR-100 inhibited the migration and invasion of MDA-MB-231 breast cancer cells. Inversely, the downregulation of miR-100 increased the migration and invasion of MCF-7 breast cancer cells. Furthermore, FZD-8, a receptor of Wnt/ß-catenin signaling pathway, was demonstrated a direct target of miR-100. The overexpression of miR-100 decreased the expression levels not only FZD-8 but also the key components of Wnt/ß-catenin pathway, including ß-catenin, metalloproteniase-7 (MMP-7), T-cell factor-4 (TCF-4), and lymphoid enhancing factor-1 (LEF-1), and increased the protein expression levels of GSK-3ß and p-GSK-3ß in MDA-MB-231 cells, and the transfection of miR-100 inhibitor in MCF-7 cells showed the opposite effects. In addition, the expression of miR-100 was negatively correlated with the FZD-8 expression in human breast cancer tissues. Overall, these findings suggest that miR-100 suppresses the migration and invasion of breast cancer cells by targeting FZD-8 and inhibiting Wnt/ß-catenin signaling pathway and manipulation of miR-100 may provide a promoting therapeutic strategy for cancer breast treatment.


Asunto(s)
Neoplasias de la Mama/genética , MicroARNs/genética , Invasividad Neoplásica/genética , Receptores de Superficie Celular/biosíntesis , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/biosíntesis , Neoplasias de la Mama/patología , Movimiento Celular/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Glucógeno Sintasa Quinasa 3 beta/biosíntesis , Humanos , Células MCF-7 , Metaloproteinasa 7 de la Matriz/biosíntesis , MicroARNs/biosíntesis , Invasividad Neoplásica/patología , Receptores de Superficie Celular/genética , Factor de Transcripción 4 , Factores de Transcripción/biosíntesis , Vía de Señalización Wnt , beta Catenina/genética
18.
BMC Cancer ; 15: 333, 2015 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-25925261

RESUMEN

BACKGROUND: Cancer-associated fibroblasts (CAFs), which reside around tumor cells, are suggested to play a pivotal role in tumor progression. Here we performed microarray analyses to compare gene expression profiles between CAFs and non-cancerous gastric fibroblasts (NGFs) from a patient with gastric cancer and found that fibroblast growth factor 9 (FGF9) was a novel growth factor overexpressed in CAFs. We then examined the biological effects of FGF9 during progression of gastric cancer. METHODS: Expression of FGF9 in CAFs and NGFs, and their secreted products, were examined by Western blotting. The effects of FGF9 on AGS and MKN28 gastric cancer cells in terms of proliferation, invasion and anti-apoptosis were assessed by WST-1 assay, invasion chamber assay and FACS, respectively. Furthermore, the intracellular signaling by which FGF9 exerts its biological roles was examined in vitro. RESULTS: FGF9 was strongly expressed in CAFs in comparison with NGFs, being compatible with microarray data indicating that FGF9 was a novel growth factor overexpressed in CAFs. Treatment with FGF9 promoted invasion and anti-apoptosis through activation of the ERK and Akt signaling pathways in AGS and MKN28 cells, whereas these effects were attenuated by treatment with anti-FGF9 neutralizing antibody. In addition, FGF9 treatment significantly enhanced the expression of matrix metalloproteinase 7 (MMP7) in both cell lines. CONCLUSIONS: FGF9 is a possible mediator secreted by CAFs that promotes the anti-apoptosis and invasive capability of gastric cancer cells.


Asunto(s)
Apoptosis/genética , Factor 9 de Crecimiento de Fibroblastos/biosíntesis , Metaloproteinasa 7 de la Matriz/biosíntesis , Neoplasias Gástricas/genética , Anticuerpos Neutralizantes/administración & dosificación , Apoptosis/efectos de los fármacos , Biomarcadores de Tumor/biosíntesis , Biomarcadores de Tumor/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Factor 9 de Crecimiento de Fibroblastos/administración & dosificación , Factor 9 de Crecimiento de Fibroblastos/genética , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Metaloproteinasa 7 de la Matriz/genética , Invasividad Neoplásica/genética , Transducción de Señal/genética , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/patología
19.
J Pharm Pharmacol ; 67(8): 1143-55, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25880237

RESUMEN

OBJECTIVES: Shikonin is an active naphthoquinone pigment isolated from the root of Lithospermum erythrorhizon. This study was designed to explore the inhibition of Shikonin on cell viability, adhesion, migration and invasion ability of gastric cancer (GC) and its possible mechanism. METHODS: 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay was performed for cell viability and adhesion ability of MGC-803 cells. Cell scratch repair experiments were conducted for the determination of migration ability while transwell assay for cell invasion ability. Western blot analysis and real-time polymerase chain reaction assay were used for the detection of protein and mRNA expressions. KEY FINDINGS: Fifty per cent inhibitory concentration of Shikonin on MGC-803 cells was 1.854 µm. Shikonin (1 µm) inhibited significantly the adhesion, invasion and migratory ability of MGC-803 cells. Interestingly, Shikonin in the presence or absence of anti-Toll-like receptor 2 (TLR2) antibody (2 µg) and nuclear factor-kappa B (NF-κB) inhibitor MG-132 (10 µm) could decrease these ability of MGC-803 cells markedly, as well as the expression levels of matrix metalloproteinases (MMP)-2, MMP-7, TLR2 and p65 NF-κB. In addition, the co-incubation of Shikonin and anti-TLR2/MG-132 has a significant stronger activity than anti-TLR2 or MG-132 alone. CONCLUSIONS: The results indicated that Shikonin could suppress the cell viability, adhesion, invasion and migratory ability of MGC-803 cells through TLR2- or NF-κB-mediated pathway. Our findings provide novel information for the treatment of Shikonin on GC.


Asunto(s)
Antineoplásicos/farmacología , FN-kappa B/metabolismo , Naftoquinonas/farmacología , Neoplasias Gástricas/tratamiento farmacológico , Receptor Toll-Like 2/metabolismo , Adhesión Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Humanos , Concentración 50 Inhibidora , Lithospermum , Metaloproteinasa 2 de la Matriz/biosíntesis , Metaloproteinasa 7 de la Matriz/biosíntesis , Reacción en Cadena en Tiempo Real de la Polimerasa , Neoplasias Gástricas/fisiopatología
20.
Oncotarget ; 6(15): 13402-15, 2015 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-25596746

RESUMEN

Recurrence of colorectal cancer (CRC) following a potentially curative resection is a challenging clinical problem. Matrix metalloproteinase-7 (MMP-7) is over-expressed by CRC cells and supposed to play a major role in CRC cell diffusion and metastasis. MMP-7 RNA expression was assessed by real-time PCR using specific primers in peritoneal washing fluid obtained during surgical procedure. After surgery, patients underwent a regular follow up for assessing recurrence. transcripts for MMP-7 were detected in 31/57 samples (54%). Patients were followed-up (range 20-48 months) for recurrence prevention. Recurrence was diagnosed in 6 out of 55 patients (11%) and two patients eventually died because of this. Notably, all the six patients who had relapsed were positive for MMP-7. Sensitivity and specificity of the test were 100% and 49% respectively. Data from patients have also been corroborated by computational approaches. Public available coloncarcinoma datasets have been employed to confirm MMP7 clinical impact on the disease. Interestingly, MMP-7 expression appeared correlated to Tgfb-1, and correlation of the two factors represented a poor prognostic factor. This study proposes positivity of MMP-7 in peritoneal cavity as a novel biomarker for predicting disease recurrence in patients with CRC.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Metaloproteinasa 7 de la Matriz/biosíntesis , Recurrencia Local de Neoplasia/metabolismo , Peritoneo/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/biosíntesis , Biomarcadores de Tumor/genética , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/cirugía , Femenino , Humanos , Masculino , Metaloproteinasa 7 de la Matriz/genética , Persona de Mediana Edad , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/patología , Periodo Posoperatorio , Pronóstico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA