Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Death Differ ; 29(11): 2137-2150, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35462559

RESUMEN

Protein S-nitrosylation is known to regulate enzymatic function. Here, we report that nitric oxide (NO)-related species can contribute to Alzheimer's disease (AD) by S-nitrosylating the lysosomal protease cathepsin B (forming SNO-CTSB), thereby inhibiting CTSB activity. This posttranslational modification inhibited autophagic flux, increased autolysosomal vesicles, and led to accumulation of protein aggregates. CA-074Me, a CTSB chemical inhibitor, also inhibited autophagic flux and resulted in accumulation of protein aggregates similar to the effect of SNO-CTSB. Inhibition of CTSB activity also induced caspase-dependent neuronal apoptosis in mouse cerebrocortical cultures. To examine which cysteine residue(s) in CTSB are S-nitrosylated, we mutated candidate cysteines and found that three cysteines were susceptible to S-nitrosylation. Finally, we observed an increase in SNO-CTSB in both 5XFAD transgenic mouse and flash-frozen postmortem human AD brains. These results suggest that S-nitrosylation of CTSB inhibits enzymatic activity, blocks autophagic flux, and thus contributes to AD pathogenesis.


Asunto(s)
Enfermedad de Alzheimer , Enfermedades Neurodegenerativas , Animales , Humanos , Ratones , Catepsina B , Agregado de Proteínas , Enfermedades Neurodegenerativas/genética , Proteínas/metabolismo , Enfermedad de Alzheimer/metabolismo , Cisteína , Óxido Nítrico
2.
Biochem Biophys Res Commun ; 578: 91-96, 2021 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-34547629

RESUMEN

The SARS-CoV-2 variant is rapidly spreading across the world and causes to resurge infections. We previously reported that CT-P59 presented its in vivo potency against Beta variants, despite its reduced activity in cell experiments. Yet, it remains uncertain to exert the antiviral effect of CT-P59 on Gamma, Delta and its associated variants (L452R). To tackle this question, we carried out cell tests and animal studies. CT-P59 showed neutralization against Gamma, Delta, Epsilon, and Kappa variants in cells, with reduced susceptibility. The mouse challenge experiments with Gamma and Delta variants substantiated in vivo potency of CT-P59 showing symptom remission and virus abrogation in the respiratory tract. Collectively, cell and animal studies showed that CT-P59 is effective against Gamma and Delta variants infection, hinting that CT-P59 has therapeutic potential for patients infected with Gamma, Delta and its associated variants.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Neutralizantes/farmacología , Tratamiento Farmacológico de COVID-19 , Modelos Animales de Enfermedad , Inmunoglobulina G/farmacología , SARS-CoV-2/efectos de los fármacos , Animales , Antivirales/farmacología , Peso Corporal/efectos de los fármacos , COVID-19/virología , Femenino , Humanos , Ratones Transgénicos , SARS-CoV-2/genética , SARS-CoV-2/fisiología , Análisis de Supervivencia
3.
Int J Mol Sci ; 21(5)2020 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-32131547

RESUMEN

Tumor cell resistance to anti-cancer drugs is a major obstacle in tumor therapy. In this study, we investigated the mechanism of cordycepin-mediated resensitization to cisplatin in T24R2 cells, a T24-derived cell line. Treatment with cordycepin or cisplatin (2 µg/mL) alone failed to induce cell death in T24R2 cells, but combination treatment with these drugs significantly induced apoptosis through mitochondrial pathways, including depolarization of mitochondrial membranes, decrease in anti-apoptotic proteins Bcl-2, Bcl-xL, and Mcl-1, and increase in pro-apoptotic proteins Bak and Bax. High expression levels of MDR1 were the cause of cisplatin resistance in T24R2 cells, and cordycepin significantly reduced MDR1 expression through inhibition of MDR1 promoter activity. MDR1 promoter activity was dependent on transcription factor Ets-1 in T24R2 cells. Although correlation exists between MDR1 and Ets-1 expression in bladder cancer patients, active Ets-1, Thr38 phosphorylated form (pThr38), was critical to induce MDR1 expression. Cordycepin decreased pThr-38 Ets-1 levels and reduced MDR1 transcription, probably through its effects on PI3K signaling, inducing the resensitization of T24R2 cells to cisplatin. The results suggest that cordycepin effectively resensitizes cisplatin-resistant bladder cancer cells to cisplatin, thus serving as a potential strategy for treatment of cancer in patients with resistance to anti-cancer drugs.


Asunto(s)
Antineoplásicos/farmacología , Cisplatino/farmacología , Desoxiadenosinas/farmacología , Resistencia a Antineoplásicos , Neoplasias de la Vejiga Urinaria/metabolismo , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Interacciones Farmacológicas , Humanos , Proteína Proto-Oncogénica c-ets-1/metabolismo
4.
Cells ; 8(5)2019 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-31121982

RESUMEN

N-Myc downstream-regulated gene 2 (NDRG2) was characterized as a tumor suppressor, inducing anti-metastatic and anti-proliferative effects in several tumor cells. However, NDRG2 functions on anticancer drug sensitivity, and its molecular mechanisms are yet to be fully investigated. In this study, we investigated the mechanism of NDRG2-induced sensitization to As2O3 in the U937 cell line, which is one of the most frequently used cells in the field of resistance to As2O3. NDRG2-overexpressing U937 cells (U937-NDRG2) showed a higher sensitivity to As2O3 than mock control U937 cell (U937-Mock). The higher sensitivity to As2O3 in U937-NDRG2 was associated with Mcl-1 degradation through glycogen synthase kinase 3ß (GSK3ß) activation. Inhibitory phosphorylation of GSK3ß was significantly reduced in U937-NDRG2, and the reduction was diminished by okadaic acid, a protein phosphatase inhibitor. NDRG2 mediated the interaction between GSK3ß and protein phosphatase 2A (PP2A), inducing dephosphorylation of GSK3ß at S9 by PP2A. Although the C-terminal deletion mutant of NDRG2 (ΔC NDRG2), which could not interact with PP2A, interacted with GSK3ß, the mutant failed to dephosphorylate GSK3ß at S9 and increased sensitivity to As2O3. Our findings suggest that NDRG2 is a kind of adaptor protein mediating the interaction between GSK3ß and PP2A, inducing GSK3ß activation through dephosphorylation at S9 by PP2A, which increases sensitivity to As2O3 in U937 cells.


Asunto(s)
Antineoplásicos/uso terapéutico , Trióxido de Arsénico/uso terapéutico , Leucemia Promielocítica Aguda/tratamiento farmacológico , Leucemia Promielocítica Aguda/metabolismo , Proteínas Supresoras de Tumor/fisiología , Hidrolasas de Éster Carboxílico/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Humanos , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Células U937
5.
Cell Death Dis ; 9(2): 30, 2018 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-29348517

RESUMEN

The downregulation of N-Myc downstream-regulated gene 2 (NDRG2) is known to be associated with the progression and poor prognosis of several cancers. Sensitivity to anti-cancer may be associated with a good prognosis in cancer patients, and NDRG2, which is induced by p53, sensitizes the cells to chemotherapy. However, the unique function of NDRG2 as an inducer of apoptosis under chemotreatment has not been sufficiently studied. In this study, we investigated the role of NDRG2 in chemo-sensitivity, focusing on cisplatin in U937 histiocytic lymphoma, which has the loss-of-functional mutation in p53. NDRG2 promoted the sensitivity to cisplatin through the modulation of the BAK-to-Mcl-1 ratio. The degradation of Mcl-1 and increase in BAK were mediated by JNK activation and the eIF2α/p-eIF2α pathway, respectively, which depended on PKR activation in NDRG2-overexpressed U937 (U937-NDRG2) cells. NOX5 was highly expressed in U937-NDRG2 cells and contributed to ROS production after cisplatin treatment. ROS scavenging or NOX5-knockdown successfully inhibited the sensitivity of U937-NDRG2 cells to cisplatin. Taken together, these findings indicate that NDRG2 contributed to the increased sensitivity to ciplatin through the modulation of Bak-to-Mcl-1 ratio regulated by NOX5-ROS-PKR pathway; therefore, we suggest that NDRG2 may be a molecular target for improving the efficacy of drug treatment in cancer patients.


Asunto(s)
Antineoplásicos/uso terapéutico , Cisplatino/uso terapéutico , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proteína Destructora del Antagonista Homólogo bcl-2/metabolismo , Antineoplásicos/farmacología , Apoptosis , Cisplatino/farmacología , Humanos
6.
Cell Death Dis ; 8(4): e2729, 2017 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-28383558

RESUMEN

Cystatin SN (CST1), a known inhibitor of cathepsin B (CatB), has important roles in tumor development. Paradoxically, CatB is a member of the cysteine cathepsin family that acts in cellular processes, such as tumor development and invasion. However, the relationship between CST1 and CatB, and their roles in tumor development are poorly understood. In this study, we observed that the knockdown of CST1 induced the activity of senescence-associated ß-galactosidase, a marker of cellular senescence, and expression of senescence-associated secretory phenotype genes, including interleukin-6 and chemokine (C-C motif) ligand 20, in MDA-MB-231 and SW480 cancer cells. Furthermore, CST1 knockdown decreased extracellular CatB activity, and direct CatB inhibition, using specific inhibitors or shCatB, induced cellular senescence. Reconstitution of CST1 restored CatB activity and inhibited cellular senescence in CST1 knockdown cells. CST1 knockdown or CatB inhibition increased glycogen synthase (GS) kinase 3ß phosphorylation at serine 9, resulting in the activation of GS and the induction of glycogen accumulation associated with cellular senescence. Importantly, CST1 knockdown suppressed cancer cell proliferation, soft agar colony growth and tumor growth in a xenograft model. These results indicate that CST1-mediated extracellular CatB activity enhances tumor development by preventing cellular senescence. Our findings suggest that antagonists of CST1 or inhibitors of CatB are potential anticancer agents.


Asunto(s)
Catepsina B/metabolismo , Proliferación Celular , Glucógeno Sintasa/metabolismo , Glucógeno/biosíntesis , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Cistatinas Salivales/metabolismo , Animales , Catepsina B/genética , Senescencia Celular/genética , Técnicas de Silenciamiento del Gen , Glucógeno/genética , Glucógeno Sintasa/genética , Células HEK293 , Xenoinjertos , Humanos , Células MCF-7 , Ratones , Proteínas de Neoplasias/genética , Trasplante de Neoplasias , Neoplasias/genética , Cistatinas Salivales/genética
7.
Exp Dermatol ; 26(7): 585-591, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-27714857

RESUMEN

Melanogenesis is a key pathway for the regulation of skin pigmentation and the development of skin-lightening/skin-whitening drugs or cosmetics. In this study, we found that ß-mangostin from seedcases of Garcinia mangostana inhibited α-melanocyte-stimulating hormone (α-MSH)-mediated melanogenesis in B16F10 melanoma cells and a three-dimensional human skin model. ß-Mangostin significantly inhibited the protein level of tyrosinase induced by α-MSH in UPS (ubiquitin proteasome system)-independent and lysosome-dependent manner. The inhibition of autophagy by 3-methyladenine treatment or ATG5 knockdown effectively recovered premelanosome protein as well as tyrosinase degraded by the ß-mangostin treatment. However, rapamycin, a representative non-selective autophagy inducer, triggered autophagy in α-MSH-stimulated cells, which was characterized by a considerable decrease in p62, but it was unable to inhibit melanogenesis. Melanosome-engulfing autophagosomes were observed using transmission electron microscopy. Furthermore, previously formed melanin could be degraded effectively in an autophagy-dependent manner in ß-mangostin-treated cells. Taken together, our results suggest that ß-mangostin inhibits the melanogenesis induced by α-MSH via an autophagy-dependent mechanism, and thus, the depigmentation effect of ß-mangostin may depend on autophagy targeted at the melanosome rather than non-selective autophagy.


Asunto(s)
Melanoma/metabolismo , Neoplasias Cutáneas/metabolismo , Xantonas/farmacología , alfa-MSH/metabolismo , Adenina/análogos & derivados , Adenina/farmacología , Animales , Autofagia , Supervivencia Celular , Garcinia mangostana , Humanos , Inflamación , Melaninas/metabolismo , Melanocitos/citología , Melanoma Experimental , Melanosomas/metabolismo , Ratones , Microscopía Electrónica de Transmisión , Monofenol Monooxigenasa/metabolismo , Pigmentación , Extractos Vegetales/farmacología , Complejo de la Endopetidasa Proteasomal/metabolismo , Semillas/química , Piel/metabolismo , Ubiquitina/metabolismo
8.
Phytomedicine ; 23(12): 1462-1468, 2016 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-27765366

RESUMEN

BACKGROUND: Broussonetia papyrifera (B. papyrifera), also known as paper mulberry, has been used as a traditional medicine for the treatment of several diseases, including ophthalmic disorders and impotency. However, the biological activity of kazinol A (1) among flavonols isolated from B. papyrifera has not been identified. PURPOSE: We identified a candidate metabolite for anti-human bladder cancer treatment from B. papyrifera and investigated the possible molecular mechanisms underlying its cytotoxic effects in T24 and cisplatin-resistant T24R2 human bladder cancer cells. METHODS: T24 and T24R2 cells were treated with five flavonols from B. papyrifera and their cytotoxic effects were determined using MTT assay, cell cycle analysis, mitochondrial membrane potential, and propidium iodide staining. Autophagy rate was calculated by counting LC3-GFP dots in the cells. All related protein expressions were analyzed by immunoblotting. RESULTS: Compound 1 showed relatively higher cytotoxicity in the human bladder cancer cells, T24 and T24R2, rather than other tissues-originated cancer cells. Compound 1 significantly attenuated cell growth through G0/1 arrest mediated by a decrease in cyclin D1 and an increase of p21. Apoptosis and autophagy induced by compound 1 treatment was accompanied by a modulation of the AKT-BAD pathway and AMPK-mTOR pathway, respectively. CONCLUSIONS: Our results suggest that compound 1 induces cytotoxic effects in human bladder cancer cells, including the cisplatin-resistant T24R2. Compound 1 may be a candidate for the development of effective anti-cancer drug on human urinary bladder cancer.


Asunto(s)
Antineoplásicos Fitogénicos/uso terapéutico , Broussonetia/química , Resistencia a Antineoplásicos/efectos de los fármacos , Fitoterapia , Extractos Vegetales/uso terapéutico , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Vejiga Urinaria/efectos de los fármacos , Antineoplásicos Fitogénicos/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cisplatino/farmacología , Humanos , Extractos Vegetales/farmacología , Vejiga Urinaria/patología
9.
Nucleic Acid Ther ; 25(2): 95-102, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25692533

RESUMEN

Although oligodeoxynucleotides containing CpG motifs (CpG-ODN) are potent immune stimulators, the use of natural CpG-ODN--phosphodiester-backbone CpG--has been limited due to its instability by nuclease in vivo. The aim of this study is to investigate the anticancer efficiency of CpG-ODN capsulated using liposome, which enhances the stability of CpG-ODN. We formulated lipoplex, encapsulated natural CpG-ODN from Mycobacterium bovis with liposome, and tested its immune stimulatory activity in vitro and in vivo. The lipoplex induced a systemic innate immune response in vivo and stimulated dendritic cells, but not macrophages, to stimulate proinflammatory cytokines such as tumor necrosis factor alpha and interleukin-6 in vitro. As expected, the lipoplex effectively mediated the prolonged cancer-therapeutic activity against B16 melanoma, which was dependent on natural killer and CD8(+) T cells. The therapeutic activity was observed after only intratumoral administration of lipoplex among several treatment routes. Intratumoral treatment of lipoplex significantly increased the populations of natural killer and CD8(+) T cells and reduced regulatory CD4(+) T cell recruitment, which was correlated with expression profiles of chemokines (CCL1, CCL3, CXCL1, CXCL10, and CCL22). The antitumor therapeutic effect of lipoplex was dependent on the altered lymphocyte population that might be developed by the profile of intratumoral chemokine expression.


Asunto(s)
Antineoplásicos/administración & dosificación , Islas de CpG , Liposomas , Linfocitos Infiltrantes de Tumor/patología , Melanoma Experimental/tratamiento farmacológico , Animales , Linfocitos T CD8-positivos/patología , Vías de Administración de Medicamentos , Femenino , Células Asesinas Naturales/patología , Ratones , Ratones Endogámicos C57BL
10.
Biomol Ther (Seoul) ; 23(1): 71-6, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25593646

RESUMEN

Peroxisome proliferator-activated receptor gamma (PPARγ) was identified as a cell-intrinsic regulator of Th17 cell differentiation. Th17 cells have been associated with several autoimmune diseases, including experimental autoimmune encephalomyelitis (EAE), inflammatory bowel disease (IBD), and collagen-induced arthritis. In this study, we confirmed PPARγ-mediated inhibition of Th17 cell differentiation and cytokine production at an early stage. Treatment with ciglitazone, a PPARγ ligand, reduced both IL-1ß-mediated enhancement of Th17 differentiation and activation of Th17 cells after polarization. For Th17 cell differentiation, we found that ciglitazone-treated cells had a relatively low proliferative activity and produced a lower amount of cytokines, regardless of the presence of IL-1ß. The inhibitory activity of ciglitazone might be due to decrease of CCNB1 expression, which regulates the cell cycle in T cells. Hence, we postulate that a pharmaceutical PPARγ activator might be a potent candidate for treatment of Th17-mediated autoimmune disease patients.

11.
Oncotarget ; 5(2): 519-29, 2014 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-24504172

RESUMEN

Collagen triple helix repeat-containing 1 (CTHRC1) is known to be aberrantly upregulated in most human solid tumors, although the functional roles of CTHRC1 in colorectal cancer remain unclear. In this study, we investigated the occurrence of CTHRC1 upregulation and its role in vivo and in vitro. The expression profile and clinical importance of CTHRC1 were examined by reverse transcription-polymerase chain reaction and immunohistochemical analyses in normal and tumor patient samples. CTHRC1 was detectable in normal tissues, but also was highly expressed in tumor specimens. CTHRC1 upregulation was significantly associated with demethylation of the CTHRC1 promoter in colon cancer cell lines and tumor tissues. Clinicopathologic analyses showed that nodal status and expression of CTHRC1 (95% CI 0.999-3.984, p=0.05) were significant prognostic factors for disease-free survival. Promoter CpG methylation and hypermethylation status were measured by bisulfite sequencing and pyrosequencing analysis. Furthermore, we showed that overexpression of CTHRC1 in the SW480 and HT-29 cell lines increased invasiveness, an effect mediated by extracellular signal-regulated kinase (ERK)-dependent upregulation of matrix metalloproteinase 9 (MMP9). Consistent with this, we found that knockdown of CTHRC1 attenuated ERK activation and cancer cell invasivity. These results demonstrate that CTHRC1 expression is elevated in human colon cancer cell lines and clinical specimens, and promotes cancer cell invasivity through ERK-dependent induction of MMP9 expression. Our results further suggest that high levels of CTHRC1 expression are associated with poor clinical outcomes.


Asunto(s)
Adenocarcinoma/metabolismo , Neoplasias Colorrectales/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Metaloproteinasa 9 de la Matriz/biosíntesis , Adenocarcinoma/genética , Adenocarcinoma/patología , Adulto , Anciano , Anciano de 80 o más Años , Movimiento Celular/fisiología , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Supervivencia sin Enfermedad , Inducción Enzimática , Proteínas de la Matriz Extracelular/biosíntesis , Proteínas de la Matriz Extracelular/genética , Femenino , Células HT29 , Humanos , Masculino , Persona de Mediana Edad , Transfección
12.
Pathol Oncol Res ; 19(4): 731-7, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23658133

RESUMEN

CTHRC1 has been known as a regulator of collagen expression and cell migration. The aim of this research was to clarify the clinicopathologic significance of CTHRC1 expression in human breast cancer. 22 cases of breast cancer tissues, randomly selected from clinically diagnosed patients, showed a significant increase of CTHRC1 mRNA expression compared to the normal tissue from the same patients using RT-PCR and real-time PCR. Additionally we investigated breast cancers from 189 patients by immunohistochemistry (IHC). A high level of CTHRC1 expression was observed in 111 (58.7 %) out of 189 breast cancer patients and the expression was significantly correlated with histologic grade (P = 0.026), nodal status (P < 0.001), and TNM pathologic stage (P = 0.002). High CTHRC1 expression was associated with a shorter recurrence free survival (P = 0.008). Taken together, the results showed that CTHRC1 over-expression was significantly associated with clinicopathological factors of poor prognosis in invasive ductal carcinoma. CTHRC1 could be used as a supplementary prognostic biomarker and a potential therapeutic target in breast cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Carcinoma Ductal de Mama/metabolismo , Carcinoma Ductal de Mama/patología , Proteínas de la Matriz Extracelular/biosíntesis , Neoplasias de la Mama/genética , Carcinoma Ductal de Mama/genética , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Femenino , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Persona de Mediana Edad , Metástasis de la Neoplasia , Pronóstico
13.
Food Chem Toxicol ; 58: 273-9, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23659910

RESUMEN

Chelidonium majus L. is an herbal plant that is commonly used in Western phytotherapy and traditional Chinese medicine for diuretic, antitussive, eye-regenerative, anti-osteoporotic, and radioprotective purposes. In this study, we purified 6-acetonyl-5,6-dihydrosanguinarine (ADS) from C. majus and investigated its immune-stimulatory effect. We found that ADS has the potential to induce the inflammatory cytokines TNF-α, IL-6, and IL-8 in macrophages and dendritic cells (DCs), that NFκB activation is a critical mediator of ADS-induced cytokine production, and that the activation of NFκB was dependent on reactive oxygen species (ROS). ADS induced phosphorylation of ERK and JNK, which was also associated with NFκB activation; phosphorylarion and cytokine production were inhibited by ROS scavenger and by specific MAPK inhibitors. Taken together, the results suggest that ADS from C. majus, as a positive immune modulator, induces inflammatory cytokines that might improve immunity, via the ROS-ERK/JNK-NFκB pathway.


Asunto(s)
Benzodioxoles/farmacología , Benzofenantridinas/farmacología , Chelidonium/metabolismo , Citocinas/biosíntesis , Mediadores de Inflamación/metabolismo , Sistema de Señalización de MAP Quinasas , FN-kappa B/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Animales , Línea Celular , Ratones
14.
Carcinogenesis ; 33(10): 1882-8, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22696597

RESUMEN

N-myc downstream-regulated gene 2 (NDRG2) has been studied for its inhibitory effects against growth and metastasis of many tumor cell types. In this study, we showed NDRG2 expression was correlated with favorable recurrence-free survival of patients with breast cancer and inhibited metastasis of breast cancer cells (4T1). NDRG2 expression was examined in 189 breast carcinoma tissues and paired normal breast tissues using immunohistochemistry. Histological and clinicopathological data were correlated using Pearson's chi-square test of independence. NDRG2 expression in human breast cancer tissues was inversely associated with lymph node metastasis and pTNM stage. Furthermore, patients with breast cancer with a high level of NDRG2 expression showed favorable recurrence-free survival (P = 0.038). To study the effect of NDRG2 on metastasis in vivo, we established an NDRG2-overexpressing mouse breast cancer cell line (4T1-NDRG2) and measured the metastasis and survival of 4T1-NDRG2 tumor-bearing mice. To test whether transforming growth factor ß (TGF-ß)- mediated metastasis of 4T1 was inhibited by NDRG2 expression, TGF-Smad-binding element (SBE)-luciferase activity and/or measurement of active TGF-ß were performed in cell or tumor tissue level. 4T1-NDRG2 cells grew gradually and showed less metastatic activity in vivo and low invasiveness in vitro. 4T1-NDRG2 cells showed lower SBE-luciferase activity and lower level of active autocrine TGF-ß than 4T1-Mock did. Correctly, our data show that NDRG2 significantly suppress tumor metastasis by attenuating active autocrine TGF-ß production, and the attenuation might be typically associated with the favorable recurrence-free survival of patients clinically.


Asunto(s)
Supervivencia sin Enfermedad , Proteínas/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Línea Celular Tumoral , Femenino , Humanos , Metástasis Linfática , Ratones , Invasividad Neoplásica , Metástasis de la Neoplasia/genética , Recurrencia
15.
Int J Mol Med ; 29(6): 1153-7, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22426464

RESUMEN

Type I interferons (IFNs), including IFN-ß, are known to enhance antigen (Ag) presentation and to promote the expansion, survival and effector function of CD8+ cytotoxic lymphocytes (CTL) during viral infections. Furthermore, IFN-ß is a potent candidate for antitumor drugs; however, recombinant IFN-ß is too unstable for use in tumor therapy in vivo. In this study, we therefore examined the efficacy and mechanism of exogenous IFN-ß as a biomolecule for tumor therapy, using adenovirus encoding IFN-ß (Ad-IFNß) as a therapeutic agent in a mouse model. Ag104Ld and 4T1 tumor cells exposed to Ad-IFNß showed growth retardation and cell death in vitro, and tumor growth as well as tumor metastasis was inhibited in vivo. The Ad-IFNß-mediated antitumor effect was dependent on CD8+ T cells in vivo, rather than on a direct cytotoxic effect of Ad-IFNß. Transient T lymphocyte depletion was observed in tumor tissue after intratumoral injection with Ad-IFNß. Despite the T lymphocyte depletion, the proliferation of Ag-specific CD8+ T cells was increased in Ad-IFNß-treated mice compared to control virus-treated mice. These results suggest that IFN-ß might contribute to the inhibition of tumor growth by depleting Ag-nonspecific T lymphocytes and enhancing proliferation of Ag-specific CD8+ T cells.


Asunto(s)
Adenoviridae/genética , Antineoplásicos/uso terapéutico , Epítopos/inmunología , Interferón beta/genética , Interferón beta/uso terapéutico , Neoplasias/tratamiento farmacológico , Linfocitos T/inmunología , Adenoviridae/efectos de los fármacos , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Proliferación Celular/efectos de los fármacos , Epítopos/efectos de los fármacos , Terapia Genética , Humanos , Depleción Linfocítica , Ratones , Neoplasias/inmunología , Neoplasias/patología , Inducción de Remisión , Linfocitos T/efectos de los fármacos , Linfocitos T/patología , Carga Tumoral/inmunología , Microambiente Tumoral
16.
Biosci Biotechnol Biochem ; 75(11): 2138-41, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22056434

RESUMEN

IL-17-producing T lymphocytes have been found to comprise a distinct lineage of T helper cells (Th17 cells) that are major causes of autoimmune diseases such as EAE, RA, and IBD. In this study, we found that activin receptor type-2A (Acvr2a) is a gene induced during the differentiation of this effector cell lineage as compared with naïve T cells. The transcript of Acvr2a was not induced in Th1 and Th2 cells, and both TGF-ß and IL-6 were required for the induction of Acvr2a. When the differentiation of Th17 cells was inhibited by all tarans retinoic acid (ATRA) which induces regulatory T cell (Treg) differentiation under Th17 differentiation conditions, expression of Acvr2a was also inhibited. Hence we propose that Acvr2a is a Th17 specific gene making Th17 cells distinct from other helper T cells, Th1, Th2, and Treg.


Asunto(s)
Receptores de Activinas Tipo II/metabolismo , Interleucina-6/metabolismo , Células Th17/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Tretinoina/farmacología , Receptores de Activinas Tipo II/genética , Receptores de Activinas Tipo II/inmunología , Animales , Diferenciación Celular , Linaje de la Célula , Femenino , Regulación de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Linfocitos T Reguladores/inmunología , Células TH1/metabolismo , Células Th17/inmunología , Células Th2/metabolismo
17.
Biochem Biophys Res Commun ; 396(3): 684-90, 2010 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-20438703

RESUMEN

N-myc downstream-regulated gene 2 (NDRG2) implicated in cellular growth and differentiation was previously reported as it is specifically expressed in primary and in vitro-differentiated dendritic cells (DCs) from monocytes and CD34(+) progenitor cells. However, its function has yet to be investigated in DCs. Here, the novel NDRG2 function about modulation of cytokines in DC was observed in this study. The secretion of IL-10 was not found in the monocyte-derived DC cells with high level of NDRG2 expression, but IL-10 was abundantly secreted up to 1ng/ml in the monocyte-derived macrophages with low level of NDRG2 expression, and further confirmed that the expression of IL-10 was dramatically increased in NDRG2-silenced DCs under presence of LPS, and significantly reduced in the NDRG2-overexpressed U937 cells under stimulation of PMA. The secretion of IL-12p70 was significantly reduced in the siNDRG2 introduced DC cells. The intracellular signaling of IL-10 secretion was markedly inhibited by SB203580, inhibitor of p38 MAPK, in the LPS-activated DCs and phosphorylation of p38 MAPK was decreased in the NDRG2 introduced U937 cells under PMA-stimulation. Taken together, NDRG2 might have a pivotal role as one of intrinsic factors for the modulation of p38 MAPK phosphorylation, and subsequently involve in controlling of IL-10 production.


Asunto(s)
Interleucina-10/biosíntesis , Células Mieloides/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Células Cultivadas , Células Dendríticas/metabolismo , Humanos , Imidazoles/farmacología , Interleucina-10/genética , Lipopolisacáridos , Macrófagos/metabolismo , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Piridinas/farmacología , ARN Interferente Pequeño/genética , Proteínas Supresoras de Tumor/genética , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...