Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Int J Nanomedicine ; 17: 3711-3722, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36051351

RESUMEN

Purpose: Magnetic particle imaging (MPI) is an emerging radiation-free, non-invasive three-dimensional tomographic technology that can visualize the concentrations of superparamagnetic iron oxide nanoparticles (SPIONs). To verify the applicability of the previously proposed point-of-care testing MPI (PoCT-MPI) in medical diagnosis and therapeutics, we imaged SPIONs in animal tumor models. Methods: CT26 or MC38 mouse colon carcinoma cells (2 × 106 cells) were subcutaneously injected into the right flank of BALB/c mice. SPIONs were either injected directly into the tumor lesions in the intratumoral group or through tail veins in the intravenous group. CT26 and MC38 tumor models were examined both intratumorally and intravenously to confirm the biological availability of SPIONs using PoCT-MPI. Results: Signals were observed in the tumor lesions from day 1 to day 7. This is the first study to successfully image the pathological region and show the biodistribution of SPIONs in CT26 tumor models using the recently developed PoCT-MPI technology. Furthermore, MC38 tumor models were examined, resulting in similar images to those of the CT26 tumor model in both intratumoral and intravenous groups. Conclusion: The present study demonstrates the biological applicability of PoCT-MPI, which promises to be a powerful diagnostic and therapeutic technique in biomedical imaging.


Asunto(s)
Nanopartículas de Magnetita , Neoplasias , Animales , Nanopartículas Magnéticas de Óxido de Hierro , Fenómenos Magnéticos , Imagen por Resonancia Magnética , Ratones , Distribución Tisular , Tomografía
2.
Front Oncol ; 12: 976407, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36176383

RESUMEN

Extracellular vesicles (EVs) derived from urine are promising tools for the diagnosis of urogenital cancers. Urinary EVs (uEVs) are considered potential biomarkers for bladder cancer (BC) because urine is in direct contact with the BC tumor microenvironment and thus reflects the current state of the disease. However, challenges associated with the effective isolation and analysis of uEVs complicate the clinical detection of uEV-associated protein biomarkers. Herein, we identified uEV-derived alpha-2-macroglobulin (a2M) as a novel diagnostic biomarker for BC through comparative analysis of uEVs obtained from patients with BC pre- and post-operation using an antibody array. Furthermore, enzyme-linked immunosorbent assay of uEVs isolated from patients with BC (n=60) and non-cancer control subjects (n=23) validated the significant upregulation of a2M expression in patient uEVs (p<0.0001). There was no significant difference in whole urine a2M levels between patients with BC and controls (p=0.317). We observed that compared to classical differential centrifugation, ExoDisc, a centrifugal microfluidic tangential flow filtration device, was a significantly more effective separation method for uEV protein analysis. We expect that our approach for EV analysis will provide an efficient route for the identification of clinically meaningful uEV-based biomarkers for cancer diagnosis.

3.
Mol Cells ; 44(10): 710-722, 2021 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-34711689

RESUMEN

Hypoxia, or low oxygen tension, is a hallmark of the tumor microenvironment. The hypoxia-inducible factor-1α (HIF-1α) subunit plays a critical role in the adaptive cellular response of hypoxic tumor cells to low oxygen tension by activating gene-expression programs that control cancer cell metabolism, angiogenesis, and therapy resistance. Phosphorylation is involved in the stabilization and regulation of HIF-1α transcriptional activity. HIF-1α is activated by several factors, including the mitogen-activated protein kinase (MAPK) superfamily. MAPK phosphatase 3 (MKP-3) is a cytoplasmic dual-specificity phosphatase specific for extracellular signal-regulated kinase 1/2 (Erk1/2). Recent evidence indicates that hypoxia increases the endogenous levels of both MKP-3 mRNA and protein. However, its role in the response of cells to hypoxia is poorly understood. Herein, we demonstrated that small-interfering RNA (siRNA)-mediated knockdown of MKP-3 enhanced HIF-1α (not HIF-2α) levels. Conversely, MKP-3 overexpression suppressed HIF-1α (not HIF-2α) levels, as well as the expression levels of hypoxia-responsive genes (LDHA, CA9, GLUT-1, and VEGF), in hypoxic colon cancer cells. These findings indicated that MKP-3, induced by HIF-1α in hypoxia, negatively regulates HIF-1α protein levels and hypoxia-responsive genes. However, we also found that long-term hypoxia (>12 h) induced proteasomal degradation of MKP-3 in a lactic acid-dependent manner. Taken together, MKP-3 expression is modulated by the hypoxic conditions prevailing in colon cancer, and plays a role in cellular adaptation to tumor hypoxia and tumor progression. Thus, MKP-3 may serve as a potential therapeutic target for colon cancer treatment.


Asunto(s)
Hipoxia de la Célula/genética , Neoplasias del Colon/genética , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/metabolismo , Animales , Línea Celular Tumoral , Neoplasias del Colon/patología , Humanos , Masculino , Ratones , Transfección , Microambiente Tumoral
4.
Cell Rep ; 37(1): 109798, 2021 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-34587481

RESUMEN

Despite the worldwide effect of the coronavirus disease 2019 (COVID-19) pandemic, the underlying mechanisms of fatal viral pneumonia remain elusive. Here, we show that critical COVID-19 is associated with enhanced eosinophil-mediated inflammation when compared to non-critical cases. In addition, we confirm increased T helper (Th)2-biased adaptive immune responses, accompanying overt complement activation, in the critical group. Moreover, enhanced antibody responses and complement activation are associated with disease pathogenesis as evidenced by formation of immune complexes and membrane attack complexes in airways and vasculature of lung biopsies from six fatal cases, as well as by enhanced hallmark gene set signatures of Fcγ receptor (FcγR) signaling and complement activation in myeloid cells of respiratory specimens from critical COVID-19 patients. These results suggest that SARS-CoV-2 infection may drive specific innate immune responses, including eosinophil-mediated inflammation, and subsequent pulmonary pathogenesis via enhanced Th2-biased immune responses, which might be crucial drivers of critical disease in COVID-19 patients.


Asunto(s)
Anticuerpos Antivirales/inmunología , COVID-19/inmunología , Proteínas del Sistema Complemento/inmunología , Eosinófilos/inmunología , Inflamación/inmunología , Neumonía Viral/inmunología , SARS-CoV-2/inmunología , Inmunidad Adaptativa , Adulto , Anciano , Anciano de 80 o más Años , Complejo Antígeno-Anticuerpo/metabolismo , COVID-19/metabolismo , COVID-19/virología , Activación de Complemento , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Eosinófilos/virología , Femenino , Humanos , Inflamación/metabolismo , Inflamación/virología , Lesión Pulmonar/inmunología , Lesión Pulmonar/patología , Lesión Pulmonar/virología , Masculino , Persona de Mediana Edad , Neumonía Viral/metabolismo , Receptores de IgG/inmunología , Receptores de IgG/metabolismo , Índice de Severidad de la Enfermedad , Transducción de Señal , Células Th2/inmunología , Carga Viral , Adulto Joven
5.
Int J Mol Sci ; 21(2)2020 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-31963761

RESUMEN

The demyelinating diseases of the central nervous system involve myelin abnormalities, oligodendrocyte damage, and consequent glia activation. Neurotoxicant cuprizone (CPZ) was used to establish a mouse model of demyelination. However, the effects of CPZ on microRNA (miRNA) expression and behavior have not been clearly reported. We analyzed the behavior of mice administered a diet containing 0.2% CPZ for 6 weeks, followed by 6 weeks of recovery. Rotarod analysis demonstrated that the treated group had poorer motor coordination than control animals. This effect was reversed after 6 weeks of CPZ withdrawal. Open-field tests showed that CPZ-treated mice exhibited significantly increased anxiety and decreased exploratory behavior. CPZ-induced demyelination was observed to be alleviated after 4 weeks of CPZ treatment, according to luxol fast blue (LFB) staining and myelin basic protein (MBP) expression. miRNA expression profiling showed that the expression of 240 miRNAs was significantly changed in CPZ-fed mice compared with controls. Furthermore, miR-155-5p and miR-20a-5p upregulations enhanced NgR induction through Smad 2 and Smad 4 suppression in demyelination. Taken together, our results demonstrate that CPZ-mediated demyelination induces behavioral deficits with apparent alterations in miRNA expression, suggesting that differences in miRNA expression in vivo may be new potential therapeutic targets for remyelination.


Asunto(s)
Cuprizona/efectos adversos , Enfermedades Desmielinizantes/psicología , Conducta Exploratoria/efectos de los fármacos , Redes Reguladoras de Genes/efectos de los fármacos , Animales , Enfermedades Desmielinizantes/inducido químicamente , Enfermedades Desmielinizantes/genética , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/efectos de los fármacos , MicroARNs/genética , Prueba de Desempeño de Rotación con Aceleración Constante
6.
Neurotox Res ; 37(3): 732-742, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31916130

RESUMEN

Myelin abnormalities, oligodendrocyte damage, and concomitant glia activation are common characteristics of demyelinating diseases of the central nervous system. Administration of the neurotoxicant cuprizone (CPZ) has been extensively used to establish a reproducible mouse model of demyelination. However, its effects on myelin-related gene expression have not been sufficiently explored. In the present study, we used the Affymetrix Mouse Gene 2.0 ST Array to analyze the changes in gene expression profiles. Comparative gene expression profiling in age-matched C57BL/6 mice administered with 0.2% CPZ and with normal diet, revealed that the expression of 1655 genes was significantly changed in CPZ-fed mice with a fold change ≥ 1.5. Our results demonstrated that CPZ-induced demyelination induces apparent alterations in the expression of most of the myelin-related genes, including the 6 key genes MBP, MAG, and MOG and GFAP, CXCR4, and NgR, which were observed to be downregulated and upregulated, respectively, suggesting that the differences in gene expression in vivo could serve as potential therapeutic targets for remyelination.


Asunto(s)
Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Cuprizona/toxicidad , Enfermedades Desmielinizantes/inducido químicamente , Enfermedades Desmielinizantes/genética , Expresión Génica , Vaina de Mielina/genética , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Masculino , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Microglía/metabolismo , Vaina de Mielina/efectos de los fármacos , Oligodendroglía/efectos de los fármacos , Oligodendroglía/metabolismo , Transcriptoma
7.
Exp Mol Med ; 51(10): 1-15, 2019 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-31649250

RESUMEN

Nogo receptor (NgR) has been shown to inhibit the migration and invasion of human glioma cells. However, little is known regarding the regulatory mechanisms of NgR in glioblastoma multiforme (GBM). In this study, we propose a novel mechanism that regulates the maturation process of NgR through an interaction with vimentin. The inhibition of TGFß1 activity by LY2109761 attenuated the migration/invasion of GBM cells by upregulating cell-surface NgR. Conversely, the treatment of GBM cells with TGFß1 suppressed NgR maturation. We showed that NgR and vimentin interact, which could be a possible mechanism for the suppression of NgR maturation. The knockdown of vimentin suppressed the migration/invasion of GBM cells through the increased maturation of NgR. Finally, TCGA (The Cancer Genome Atlas) analysis also supported the association of NgR and vimentin. The maturation of NgR is regulated by the interaction of vimentin and NgR, which attenuates the invasive activity of GBM, and might be a potential therapeutic target for brain cancer.


Asunto(s)
Proteínas de la Matriz Extracelular/genética , Glioblastoma/genética , Receptor Nogo 1/genética , Factor de Crecimiento Transformador beta/genética , Vimentina/genética , Biomarcadores de Tumor/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica/genética , Glioblastoma/patología , Humanos , Invasividad Neoplásica/genética
8.
Oncol Rep ; 41(6): 3464-3474, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31002343

RESUMEN

The EF­hand calcium binding protein tescalcin (TESC) is highly expressed in various human and mouse cancer tissues and is therefore considered a potential oncogene. However, the underlying mechanism that governs TESC expression remains unclear. Emerging evidence suggests that TESC expression is under epigenetic regulation. In the present study, the relationship between the epigenetic modification and gene expression of TESC in gastric cancer was investigated. To evaluate the relationship between the methylation and expression of TESC in gastric cancer, the methylation status of CpG sites in the TESC promoter was analyzed using microarray with the Illumina Human Methylation27 BeadChip (HumanMethylation27_270596_v.1.2), gene profiles from the NCBI Dataset that revealed demethylated status were acquired, and real­time methylation­specific PCR (MSP) in gastric cancer cells was conducted. In the present study, it was demonstrated that the hypermethylation of TESC led to the downregulation of TESC mRNA/protein expression. In addition, 5­aza­2c­deoxycytidine (5'­aza­dC) restored TESC expression in the tested gastric cancer cells except for SNU­620 cells. ChIP assay further revealed that the methylation of the TESC promoter was associated with methyl­CpG binding domain protein (MBD)1, histone deacetylase (HDAC)2, and Oct­1 and that treatment with 5'­aza­dC facilitated the dissociation of MBD1, HDAC2, and Oct­1 from the promoter of TESC. Moreover, silencing of TESC increased MBD1 expression and decreased the H3K4me2/3 level, thereby causing transcriptional repression and suppression of cell survival in NCI­N87 cells; conversely, overexpression of TESC downregulated MBD1 expression and upregulated the H3K4me2 level associated with active transcription in SNU­638 cells. These results indicated that the differential expression of TESC via the modification status of the promoter and histone methylation controled cell survival in gastric cancer cells. Overall, the present study provided a novel therapeutic strategy for gastric cancer.


Asunto(s)
Azacitidina/farmacología , Proteínas de Unión al Calcio/genética , Metilación de ADN/genética , Neoplasias Gástricas/genética , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Metilación de ADN/efectos de los fármacos , Proteínas de Unión al ADN/genética , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Código de Histonas/genética , Histona Desacetilasa 2/genética , Humanos , Análisis por Micromatrices , Factor 1 de Transcripción de Unión a Octámeros/genética , Regiones Promotoras Genéticas/efectos de los fármacos , Neoplasias Gástricas/patología , Factores de Transcripción/genética
9.
Sci Rep ; 8(1): 5415, 2018 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-29615744

RESUMEN

There is increasing evidence that the complement system is activated in various cancer tissues. Besides being involved in innate immunity against pathogens, the complement system also participates in inflammation and the modulation of tumor microenvironment. Recent studies suggest that complement activation promotes tumor progression in various ways. Among some cancer cell lines, we found that human bone osteosarcoma epithelial cells (U2-OS) can activate the alternative pathway of the complement system by pooled normal human serum. Interestingly, U2-OS cells showed less expression of complement regulatory proteins, compared to other cancer cell lines. Furthermore, the activated complement system enhanced the production of growth factors, which promoted angiogenesis of human endothelial cells. Our results demonstrated a direct linkage between the complement system and angiogenesis using the in vitro model, which suggest the complement system and related mechanisms might be potential targets for cancer treatment.


Asunto(s)
Neoplasias Óseas/patología , Proteínas del Sistema Complemento/metabolismo , Factor 1 de Crecimiento de Fibroblastos/metabolismo , Neovascularización Patológica/metabolismo , Osteosarcoma/patología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Neoplasias Óseas/irrigación sanguínea , Neoplasias Óseas/metabolismo , Línea Celular Tumoral , Células Endoteliales/patología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Osteosarcoma/irrigación sanguínea , Osteosarcoma/metabolismo , Fosforilación
10.
Tumour Biol ; 37(10): 13843-13853, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27485112

RESUMEN

We reported previously that tescalcin (TESC) levels were higher in tissue and serum from colorectal cancer (CRC) patients and suggested that TESC was a potential oncotarget in CRC. The aim of this study was to investigate the function of TESC in CRC invasion and metastatic potential. TESC expression was knocked down in CRC cells using small interfering RNA (siRNA). The expression of TESC siRNA reduced cell migration and invasion by inhibiting matrix metalloprotease (MMP) and the epithelial-mesenchymal transition (EMT) pathway. RT-PCR and Western blot analysis showed that TESC siRNA induced E-cadherin. Consistently, TESC overexpression in HCT116 (HCT/TESC) cells enhanced cell migration and invasion by activating MMP and the EMT pathway and reducing E-cadherin. The formation of liver metastatic nodules in vivo was strongly increased in mice injected with HCT/TESC cells compared with that in mice injected with HCT/mock cells. This study demonstrates that TESC is involved in cell migration, invasion, and EMT during CRC tumor invasion. These results implicate TESC as a metastatic mediator and provide a biological rationale for the adverse prognosis associated with elevated TESC expression in human CRC.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Proteínas de Unión al Calcio/metabolismo , Movimiento Celular , Neoplasias Colorrectales/patología , Neoplasias Hepáticas/secundario , Animales , Apoptosis , Biomarcadores de Tumor/genética , Western Blotting , Proteínas de Unión al Calcio/genética , Estudios de Casos y Controles , Proliferación Celular , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/cirugía , Femenino , Estudios de Seguimiento , Humanos , Técnicas para Inmunoenzimas , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/cirugía , Ratones , Ratones Desnudos , Invasividad Neoplásica , Metástasis de la Neoplasia , Estadificación de Neoplasias , Pronóstico , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Oncotarget ; 5(8): 2149-60, 2014 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-24811141

RESUMEN

Tescalcin (TESC) is an EF-hand calcium binding protein that is differentially expressed in several tissues, however it is not reported that the expression and functional roles of TESC in colorectal cancer. Levels of messenger RNA (mRNA) and protein expression of TESC in colorectal cancer tissues were assessed using RT-PCR, real time PCR, immunohistochemistry, and clinicopathologic analyses. Quantitative analysis of TESC levels in serum specimens was performed using sandwich ELISA. Colorectal cancer cells transfected with TESC small interfering RNA and short hairpin RNA were examined in cell proliferation assays, phospho-MAPK array, and mouse xenograft models. Here we demonstrated that TESC is overexpressed in colorectal cancer (CRC), but was not expressed in normal mucosa and premalignant dysplastic lesions. Furthermore, serum TESC levels were elevated in patients with CRC. Knockdown of TESC inhibited the Akt-dependent NF-κB pathway and decreased cell survival in vitro. Depletion of TESC reduced tumor growth in a CRC xenograft model. Thus, TESC is a potential diagnostic marker and oncotarget in colorectal cancer.


Asunto(s)
Adenocarcinoma/metabolismo , Biomarcadores de Tumor/análisis , Proteínas de Unión al Calcio/metabolismo , Neoplasias Colorrectales/metabolismo , Adenocarcinoma/mortalidad , Adenocarcinoma/patología , Adulto , Anciano , Animales , Western Blotting , Neoplasias Colorrectales/mortalidad , Neoplasias Colorrectales/patología , Ensayo de Inmunoadsorción Enzimática , Femenino , Xenoinjertos , Humanos , Inmunohistoquímica , Inmunoprecipitación , Estimación de Kaplan-Meier , Masculino , Ratones , Ratones Desnudos , Microscopía Confocal , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Regulación hacia Arriba
12.
Cell Signal ; 24(10): 1940-9, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22735811

RESUMEN

In our previous study, we reported that endothelial cell specific molecule-1 (ESM-1) was increased in tissue and serum from colorectal cancer patients and suggested that ESM-1 can be used as a potential serum marker for early detection of colorectal cancer. The aim of this study was to evaluate the role of ESM-1 as an intracellular molecule in colorectal cancer. ESM-1 expression was knocked down by small interfering RNA (siRNA) in colorectal cancer cells. Expression of ESM-1 siRNA decreased cell survival through the Akt-dependent inhibition of NF-κB/IκB pathway and an interconnected reduction in phospho-Akt, -p38, -ERK1, -RSK1, -GSK-3α/ß and -HSP27, as determined by a phospho-MAPK array. ESM-1 silencing induced G(1) phase cell cycle arrest by induction of PTEN, resulting in the inhibition of cyclin D1 and inhibited cell migration and invasion of COLO205 cells. Consistently, ESM-1 overexpression in HCT-116 cells enhanced cell proliferation through the Akt-dependent activation of NF-κB pathway. In addition, ESM-1 interacted with NF-κB and activated NF-κB promoter. This study demonstrates that ESM-1 is involved in cell survival, cell cycle progression, migration, invasion and EMT during tumor invasion in colorectal cancer. Based on our results, ESM-1 may be a useful therapeutic target for colorectal cancer.


Asunto(s)
Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/secundario , FN-kappa B/inmunología , Metástasis de la Neoplasia/inmunología , Proteínas de Neoplasias/inmunología , Proteoglicanos/inmunología , Puntos de Control del Ciclo Celular , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Ciclina D1/inmunología , Regulación Neoplásica de la Expresión Génica , Humanos , Metaloproteinasas de la Matriz/genética , Metaloproteinasas de la Matriz/inmunología , Metástasis de la Neoplasia/genética , Metástasis de la Neoplasia/patología , Proteínas de Neoplasias/genética , Fosfohidrolasa PTEN/inmunología , Proteoglicanos/genética , Proteínas Proto-Oncogénicas c-akt/inmunología , Interferencia de ARN , ARN Interferente Pequeño/genética , Transducción de Señal
13.
Cancer Lett ; 318(2): 226-33, 2012 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-22198481

RESUMEN

IL-32 is a newly discovered cytokine. Recently, various reports suggest that it plays a role as a proinflammatory mediator and may be involved in several cancer carcinogenesis. However, IL-32 expression in hepatocellular carcinoma (HCC) remains unclear. In this study, we investigated the expression and role of IL-32α in hepatocellular carcinoma, because IL-32 was identified as an upregulated gene in hepatocellular carcinoma tissues compared to nontumorous regions using DNA microarray. IL-32α was overexpressed in tissue and serum from patients with HCC and localized in the cytoplasm and nucleus of hepatocellular carcinoma tumor cells. Moreover, secreted IL-32α concentration in the serum of patients with hepatocellular carcinoma was elevated as compared with those in the normal serum using a developed sandwich ELISA. Furthermore, IL-32α suppression in hepatocellular carcinoma decreased expression of phospho-p38 MAPK, NF-κB, and antiapoptotic protein Bcl-2 and induced expression of proapoptotic proteins as well as p53 and PUMA resulting in the suppression of cell growth and induction of intrinsic apoptosis. Based on our results, we suggest that IL-32α is involved in the progression of hepatocellular carcinoma and may be a useful biomarker for diagnosis and therapeutic target of hepatocellular carcinoma.


Asunto(s)
Apoptosis/fisiología , Biomarcadores de Tumor/fisiología , Carcinoma Hepatocelular/metabolismo , División Celular/fisiología , Interleucinas/fisiología , Neoplasias Hepáticas/metabolismo , FN-kappa B/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Secuencia de Bases , Biomarcadores de Tumor/sangre , Western Blotting , Carcinoma Hepatocelular/patología , Cartilla de ADN , Ensayo de Inmunoadsorción Enzimática , Humanos , Inmunohistoquímica , Interleucinas/sangre , Neoplasias Hepáticas/patología , Reacción en Cadena de la Polimerasa
14.
Mol Pain ; 5: 50, 2009 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-19725970

RESUMEN

Group I mGluRs (mGluR1 and 5) pre- and/or postsynaptically regulate synaptic transmission at glutamatergic synapses. By recording spontaneous EPSCs (sEPSCs) in the spinal trigeminal subnucleus oralis (Vo), we here investigated the regulation of glutamatergic transmission through the activation of group I mGluRs. Bath-applied DHPG (10 microM/5 min), activating the group I mGluRs, increased sEPSCs both in frequency and amplitude; particularly, the increased amplitude was long-lasting. The DHPG-induced increases of sEPSC frequency and amplitude were not NMDA receptor-dependent. The DHPG-induced increase in the frequency of sEPSCs, the presynaptic effect being further confirmed by the DHPG effect on paired-pulse ratio of trigeminal tract-evoked EPSCs, an index of presynaptic modulation, was significantly but partially reduced by blockades of voltage-dependent sodium channel, mGluR1 or mGluR5. Interestingly, PKC inhibition markedly enhanced the DHPG-induced increase of sEPSC frequency, which was mainly accomplished through mGluR1, indicating an inhibitory role of PKC. In contrast, the DHPG-induced increase of sEPSC amplitude was not affected by mGluR1 or mGluR5 antagonists although the long-lasting property of the increase was disappeared; however, the increase was completely inhibited by blocking both mGluR1 and mGluR5. Further study of signal transduction mechanisms revealed that PLC and CaMKII mediated the increases of sEPSC in both frequency and amplitude by DHPG, while IP3 receptor, NO and ERK only that of amplitude during DHPG application. Altogether, these results indicate that the activation of group I mGluRs and their signal transduction pathways differentially regulate glutamate release and synaptic responses in Vo, thereby contributing to the processing of somatosensory signals from orofacial region.


Asunto(s)
Receptores de Glutamato Metabotrópico/fisiología , Transducción de Señal/fisiología , Animales , Femenino , Inmunohistoquímica , Masculino , Metoxihidroxifenilglicol/análogos & derivados , Metoxihidroxifenilglicol/farmacología , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley , Receptor del Glutamato Metabotropico 5 , Receptores de Glutamato Metabotrópico/agonistas , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA