Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Rheumatology (Oxford) ; 59(4): 742-753, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-31377809

RESUMEN

OBJECTIVES: Xerostomia in SS patients has been associated with low quality and quantity of salivary mucins, which are fundamental for the hydration and protection of the oral mucosa. The aim of this study was to evaluate if cytokines induce aberrant mucin expression and whether tauroursodeoxycholic acid (TUDCA) is able to counteract such an anomaly. METHODS: Labial salivary glands from 16 SS patients and 15 control subjects, as well as 3D acini or human submandibular gland cells stimulated with TNF-α or IFN-γ and co-incubated with TUDCA, were analysed. mRNA and protein levels of Mucin 1 (MUC1) and MUC7 were determined by RT-qPCR and western blot, respectively. Co-immunoprecipitation and immunofluorescence assays for mucins and GRP78 [an endoplasmic reticulum (ER)-resident protein] were also performed. mRNA levels of RelA/p65 (nuclear factor-κB subunit), TNF-α, IL-1ß, IL-6, SEL1L and EDEM1 were determined by RT-qPCR, and RelA/p65 localization was evaluated by immunofluorescence. RESULTS: MUC1 is overexpressed and accumulated in the ER of labial salivary gland from SS patients, while MUC7 accumulates throughout the cytoplasm of acinar cells; however, MUC1, but not MUC7, co-precipitated with GRP78. TUDCA diminished the overexpression and aberrant accumulation of MUC1 induced by TNF-α and IFN-γ, as well as the nuclear translocation of RelA/p65, together with the expression of inflammatory and ER stress markers in 3D acini. CONCLUSION: Chronic inflammation alters the secretory process of MUC1, inducing ER stress and affecting the quality of saliva in SS patients. TUDCA showed anti-inflammatory properties decreasing aberrant MUC1 accumulation. Further studies are necessary to evaluate the potential therapeutic effect of TUDCA in restoring glandular homeostasis in SS patients.


Asunto(s)
Células Acinares/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Mucina-1/efectos de los fármacos , Glándulas Salivales Menores/efectos de los fármacos , Síndrome de Sjögren/metabolismo , Glándula Submandibular/efectos de los fármacos , Ácido Tauroquenodesoxicólico/farmacología , Xerostomía/metabolismo , Células Acinares/metabolismo , Adulto , Anciano , Estudios de Casos y Controles , Células Cultivadas , Chaperón BiP del Retículo Endoplásmico , Estrés del Retículo Endoplásmico/genética , Femenino , Proteínas de Choque Térmico/efectos de los fármacos , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Humanos , Inmunoprecipitación , Técnicas In Vitro , Interferón gamma/farmacología , Interleucina-1beta/efectos de los fármacos , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Masculino , Proteínas de la Membrana/efectos de los fármacos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Persona de Mediana Edad , Mucina-1/genética , Mucina-1/metabolismo , Mucinas/efectos de los fármacos , Mucinas/genética , Mucinas/metabolismo , Proteínas/efectos de los fármacos , Proteínas/genética , Proteínas/metabolismo , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Glándulas Salivales Menores/metabolismo , Proteínas y Péptidos Salivales/efectos de los fármacos , Proteínas y Péptidos Salivales/genética , Proteínas y Péptidos Salivales/metabolismo , Síndrome de Sjögren/genética , Glándula Submandibular/citología , Glándula Submandibular/metabolismo , Factor de Transcripción ReIA/efectos de los fármacos , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/metabolismo , Factor de Necrosis Tumoral alfa/efectos de los fármacos , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Xerostomía/genética
2.
J Cell Physiol ; 234(7): 12019-12028, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30523643

RESUMEN

Esophageal squamous cell carcinoma (ESCC) is one of the most common digestive tumors worldwide. The Mucin 1 (MUC1) heterodimeric protein has been confirmed that is overexpressed in ESCC and induced adverse outcomes. However, the detailed mechanism(s) remained challenging. So, we investigated the relationship between MUC1-C and metabolism in ESCC cells. In the results, TP53-induced glycolysis and apoptosis regulator (TIGAR) was overexpressed and correlative with MUC1-C positively in ESCC tissue. Targeting MUC1-C inhibits AKT-mTORC-S6K1 signaling and blocks TIGAR translation. We found that the inhibitory effect of GO-203 on TIGAR was mediated by inhibition of AKT-mTOR-S6K1 pathway. The findings also demonstrated that the suppressive effect of GO-203 on TIGAR is related to the decrease of glutathione level, the increase of reactive oxygen species and the loss of mitochondrial transmembrane membrane potential. In xenograft tissues, GO-203 inhibited the growth of ESCC cells and lead to the low expression of transmembrane C-terminal subunit (MUC1-C) and TIGAR. This evidence supports the contention that MUC1-C is significant for metabolism in ESCC and indicated that MUC1-C is a potential target for the treatment of ESCC.


Asunto(s)
Carcinoma de Células Escamosas de Esófago/tratamiento farmacológico , Mucina-1/efectos de los fármacos , Péptidos/farmacología , Proteínas Proto-Oncogénicas c-akt/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Neoplasias Esofágicas/tratamiento farmacológico , Neoplasias Esofágicas/metabolismo , Femenino , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones Endogámicos BALB C , Ratones Desnudos , Mucina-1/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Especies Reactivas de Oxígeno/metabolismo
3.
Br J Haematol ; 178(6): 914-926, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28643330

RESUMEN

Lenalidomide (LEN) acts directly on multiple myeloma (MM) cells by inducing cereblon-mediated degradation of interferon regulatory factor 4, Ikaros (IKZF)1 and IKZF3, transcription factors that are essential for MM cell survival. The mucin 1 (MUC1) C-terminal transmembrane subunit (MUC1-C) oncoprotein is aberrantly expressed by MM cells and protects against reactive oxygen species (ROS)-mediated MM cell death. The present studies demonstrate that targeting MUC1-C with GO-203, a cell-penetrating peptide inhibitor of MUC1-C homodimerization, is more than additive with LEN in downregulating the WNT/ß-catenin pathway, suppressing MYC, and inducing late apoptosis/necrosis. We show that the GO-203/LEN combination acts by synergistically increasing ROS and, in turn, suppressing ß-catenin. LEN resistance has been linked to activation of the WNT/ß-catenin→CD44 pathway. In this regard, our results further demonstrate that targeting MUC1-C is effective against LEN-resistant MM cells. Moreover, GO-203 resensitized LEN-resistant MM cells to LEN treatment in association with suppression of ß-catenin and CD44. Targeting MUC1-C also resulted in downregulation of CD44 on the surface of primary MM cells. These findings, and the demonstration that expression of MUC1 and CD44 significantly correlate in microarrays from primary MM cells, provide support for combining GO-203 with LEN in the treatment of MM and in LEN-resistance.


Asunto(s)
Mucina-1/efectos de los fármacos , Mieloma Múltiple/patología , Talidomida/análogos & derivados , Antineoplásicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Apoptosis/efectos de los fármacos , Biología Computacional/métodos , Resistencia a Antineoplásicos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Receptores de Hialuranos/metabolismo , Lenalidomida , Terapia Molecular Dirigida/métodos , Mucina-1/metabolismo , Mieloma Múltiple/metabolismo , Oxidación-Reducción/efectos de los fármacos , Péptidos/administración & dosificación , Péptidos/farmacología , Especies Reactivas de Oxígeno/metabolismo , Talidomida/administración & dosificación , Talidomida/farmacología , Células Tumorales Cultivadas/efectos de los fármacos
4.
Am J Pathol ; 186(12): 3146-3159, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27765636

RESUMEN

Lung cancer is the leading cause of cancer-related death, and 87% of these deaths are directly attributable to smoking. Using three-dimensional cultures of primary human bronchial epithelial cells, we demonstrated that loss of adherens junction protein, epithelial cadherin, and the aberrant interaction of its adherens junction binding partner, p120-catenin (p120ctn), with the cytoplasmic tail of apical mucin-1 (MUC1-CT) represent initiating steps in the epithelial-to-mesenchymal transition. Smoke provoked the rapid nuclear entry of p120ctn in complex with MUC1-CT that was inhibited using the MUC1-CT inhibitory peptides, PMIP and GO-201. Nuclear entry of p120ctn promoted its interaction with transcriptional repressor kaiso and the rapid shuttling of kaiso to the cytoplasm. Nuclear exit of kaiso permitted the up-regulation of oncogenic transcription factors Fos/phospho-Ser32 Fos, FosB, Fra1/phospho-Ser265 Fra1, which was inhibited through suppression of p120ctn's nuclear export using leptomycin-B. These data indicated that smoke-induced nuclear-to-cytoplasmic translocation of kaiso depends on the nuclear import of p120ctn in complex with MUC1-CT and the nuclear export of kaiso in complex with p120ctn. The presence of MUC1-CT/p120ctn and p120ctn/kaiso complexes in lung squamous cell carcinoma and adenocarcinoma specimens from human patients confirms the clinical relevance of these events. Thus, enhancing kaiso's suppressor role of protumor genes by sequestering kaiso in the nucleus of a smoker's airway epithelium may represent a novel approach of treating lung cancer.


Asunto(s)
Cateninas/metabolismo , Neoplasias Pulmonares/etiología , Mucina-1/metabolismo , Fumar/efectos adversos , Factores de Transcripción/metabolismo , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/etiología , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Anciano , Antígenos CD , Cadherinas/metabolismo , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/etiología , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Movimiento Celular , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Células Epiteliales/metabolismo , Transición Epitelial-Mesenquimal , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Masculino , Persona de Mediana Edad , Mucina-1/efectos de los fármacos , Péptidos/farmacología , Transporte de Proteínas , Regulación hacia Arriba , Catenina delta
5.
Eye Contact Lens ; 39(6): 394-9, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24113462

RESUMEN

OBJECTIVE: The purpose of this study was first to evaluate the effect of multipurpose contact lens care solutions (MPSs) on the expression of membrane-associated mucins (MUC1 and MUC16) in SV40-transformed human corneal epithelial (HCE-T) cells and in vivo rat cornea. The second aim of this study was to determine the role of the common MPS additive boric acid in reducing mucin expression and release. METHODS: The HCE-T cells were exposed to different concentrations of MPS-F, MPS-G, MPS-H, MPS-I, and MPS-J with 100% treatment for 30 minutes and 10% treatment for 24 hours. MUC1 and MUC16 expressions were subsequently analyzed by Western blotting. Wister rats were also subjected to MPS-A, MPS-B, MPS-C, MPS-D, and MPS-E and received phosphate-buffered saline exposure (1 drop in the right eye every 10 minutes for 1 hour). The left eye was used as control. Cornea sections and lysates were used for the immunohistochemical assay of MUC1 and MUC16 expressions. Conditioned media from treated HCE-T cells were also analyzed using Western blotting. RESULTS: The MPSs containing boric acid downregulated MUC1 and MUC16 in the rat cornea, whereas MPSs without boric acid had no effect as demonstrated by the Western blotting and immunohistochemical analysis. Conditioned media from MPS-containing boric acid revealed some trace of MUC16. CONCLUSIONS: The clinical use of MPSs containing boric acid that reduce MUC1 and MUC16 availability should be avoided. Additionally, the presence of MUC16 in the conditioned media suggests that boric acid may have enhanced cleavage of MUC16 at the cell membrane surface.


Asunto(s)
Antígeno Ca-125/efectos de los fármacos , Soluciones para Lentes de Contacto/farmacología , Epitelio Corneal/efectos de los fármacos , Proteínas de la Membrana/efectos de los fármacos , Mucina-1/efectos de los fármacos , Análisis de Varianza , Animales , Western Blotting , Ácidos Bóricos/farmacología , Antígeno Ca-125/metabolismo , Células Cultivadas , Córnea/efectos de los fármacos , Córnea/metabolismo , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Epitelio Corneal/metabolismo , Humanos , Inmunohistoquímica , Proteínas de la Membrana/metabolismo , Mucina-1/metabolismo , Ratas , Ratas Wistar
6.
Mol Pharmacol ; 79(5): 886-93, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21346142

RESUMEN

Mucin 1 (MUC1) is a heterodimeric protein that is overexpressed in diverse human carcinomas. The oncogenic function of the MUC1 C-terminal subunit (MUC1-C) subunit is dependent on the formation of dimers through its cytoplasmic domain; however, it is not known whether MUC1-C can be targeted with small-molecule inhibitors. In the present work, an assay using the MUC1-C cytoplasmic domain (MUC1-CD) was established to screen small-molecule libraries for compounds that block its dimerization. Using this approach, the flavone apigenin was identified as an inhibitor of MUC1-CD dimerization in vitro and in cells. By contrast, the structurally related flavone baicalein was ineffective in blocking the formation of MUC1-CD dimers. In concert with these results, apigenin, and not baicalein, blocked the localization of MUC1-C to the nucleus. MUC1-C activates MUC1 gene expression in an autoinductive loop, and apigenin, but not baicalein, treatment was associated with down-regulation of MUC1 mRNA levels and MUC1-C protein. The results also demonstrate that apigenin-induced suppression of MUC1-C expression is associated with apoptotic cell death and loss of clonogenic survival. These findings represent the first demonstration that the MUC1-C cytoplasmic domain is a target for the development of small-molecule inhibitors.


Asunto(s)
Mucina-1/efectos de los fármacos , Proteínas Oncogénicas/antagonistas & inhibidores , Secuencia de Aminoácidos , Apigenina/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Dimerización , Regulación hacia Abajo/efectos de los fármacos , Humanos , Datos de Secuencia Molecular , Mucina-1/química , Mucina-1/genética , Proteínas Oncogénicas/química , Proteínas Oncogénicas/genética , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
7.
Med Oral Patol Oral Cir Bucal ; 16(1): e29-32, 2011 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-20711149

RESUMEN

OBJECTIVES: Oral contraceptives (OCP) are highly effective, safe and widely used. Higher exposure to endogenous and exogenous estrogens is generally thought to increase the risk of breast cancer. Therefore, this study was conducted to determine if oral contraceptive use affected the expression of CA 15-3, CEA and C-erb B-2 in the saliva of healthy women. STUDY DESIGN: The participants consisted of 87 healthy women (43 controls and 44 using oral contraceptives) ranging in age from 20 to 54 years. The volunteers participated by giving one - time stimulated whole saliva samples. Then the samples were analysed for CA 15-3, CEA and C-erb B-2 concentrations. RESULTS: The student t-test was used to compare group means for variables with comparable variability. The mean of C-erb B-2, CEA, and CA 15-3 concentrations (in the case and control groups) was (1.93, 1.70), (34.46, 31.62) and (12.58, 16.19) respectively. These differences were not statistically significant. CONCLUSIONS: Our findings suggest that the levels of the cancer biomarkers C-erb B-2, CEA and CA 15-3 were not affected by increased levels of estrogens in the body.


Asunto(s)
Antígeno Carcinoembrionario/análisis , Antígeno Carcinoembrionario/biosíntesis , Anticonceptivos Orales/farmacología , Mucina-1/análisis , Mucina-1/biosíntesis , Receptor ErbB-2/análisis , Receptor ErbB-2/biosíntesis , Saliva/química , Adulto , Antígeno Carcinoembrionario/efectos de los fármacos , Estudios de Casos y Controles , Femenino , Humanos , Persona de Mediana Edad , Mucina-1/efectos de los fármacos , Receptor ErbB-2/efectos de los fármacos , Adulto Joven
8.
JNCI Cancer Spectr ; 5(5)2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34485814

RESUMEN

Background: Circulating levels of cancer antigen (CA) 15-3, a tumor marker and regulator of cellular metabolism, were reduced by metformin in a nonrandomized neoadjuvant study. We examined the effects of metformin (vs placebo) on CA 15-3 in participants of MA.32, a phase III randomized trial in early-stage breast cancer. Methods: A total of 3649 patients with T1-3, N0-3, M0 breast cancer were randomly assigned; pretreatment and 6-month on-treatment fasting plasma were centrally assayed for CA 15-3. Genomic DNA was analyzed for the rs11212617 single nucleotide polymorphism. Absolute and relative change of CA 15-3 (metformin vs placebo) were compared using Wilcoxon rank and t tests. Regression models adjusted for baseline differences and assessed key interactions. All statistical tests were 2-sided. Results: Mean (SD) age was 52.4 (10.0) years. The majority of patients had T2/3, node-positive, hormone receptor-positive, HER2-negative breast cancer treated with (neo)adjuvant chemotherapy and hormone therapy. Mean (SD) baseline CA 15-3 was 17.7 (7.6) and 18.0 (8.1 U/mL). At 6 months, CA 15-3 was statistically significantly reduced in metformin vs placebo arms (absolute geometric mean reduction in CA 15-3 = 7.7% vs 2.0%, P < .001; relative metformin: placebo level of CA 15-3 [adjusted for age, baseline body mass index, and baseline CA 15-3] = 0.94, 95% confidence interval = 0.92 to 0.96). This reduction was independent of tumor characteristics, perioperative systemic therapy, baseline body mass index, insulin, and the single nucleotide polymorphism status (all Ps > .11). Conclusions: Our observation that metformin reduces CA 15-3 by approximately 6% was corroborated in a large placebo-controlled randomized trial. The clinical implications of this reduction in CA 15-3 will be explored in upcoming efficacy analyses of breast cancer outcomes in MA.32.


Asunto(s)
Neoplasias de la Mama/sangre , Metformina/uso terapéutico , Mucina-1/sangre , Índice de Masa Corporal , Neoplasias de la Mama/química , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Quimioterapia Adyuvante , Ayuno/sangre , Femenino , Humanos , Persona de Mediana Edad , Mucina-1/efectos de los fármacos , Placebos/uso terapéutico , Polimorfismo de Nucleótido Simple
9.
Tissue Cell ; 70: 101503, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33556842

RESUMEN

To elucidate the role of sialomucin in friction reduction, we investigated the sliding friction of pleural mesothelial cells monolayers cultured on fibrine gel. These measurements were performed on normal (4/4 RM-4) and on tumor (CARM-L1 TG3) cell lines. The effect of treatment with neuraminidase, which removes sialic acid from sialomucin, and of dexamethasone, which has shown to increase sialomucin expression, were also assessed. Furthermore, the expression of the main form of cell-surface-associated mucin (MUC1) present in the mesothelium, was assessed by western blot and immunofluorescence, under different experimental conditions. Expression of MUC1 was not significantly different in the two cell lines. Moreover, dexamethasone did not increase the expression of MUC1. Coefficient of kinetic friction (µ) was significantly higher in tumor cells than in normal cells. Neuraminidase increased µ in both cell lines. These results suggest that sialomucin may play a role in reducing the friction of pleural mesothelial cells.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Epitelio , Lubrificación , Mucina-1 , Sialomucinas , Línea Celular Tumoral , Células Cultivadas , Fricción/efectos de los fármacos , Humanos , Mucina-1/efectos de los fármacos , Mucina-1/metabolismo , Pleura/citología , Sialomucinas/metabolismo , Sialomucinas/farmacología
10.
Mol Immunol ; 46(4): 738-42, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18952292

RESUMEN

Mucin-1 (MUC1) is a transmembrane glycoprotein that is upregulated upon maturation of dendritic cells (DC) in vitro or in vivo. One of the proposed functions of surface expressed MUC1 is its involvement in migration of cells. We hypothesized that MUC1 is involved in DC migration since mature DC (mDC) are highly migratory cells and MUC1 is upregulated on the surface of DC upon maturation. In this study we cultured DC using two maturation cocktails, one cocktail containing IL-4, GM-CSF, TNFalpha, PGE2, IL-1 beta and IL-6 (TP1,6-DC) and the other IL-13, GM-CSF, Ribomunyl and IFN-gamma (RI-DC). Both maturation cocktails render DC with a similar surface phenotype including CCR7 expression, but only the former induces a migratory capacity of DC to a CCL19 gradient. To analyze the role of surface-expression of MUC1 on TP1,6-DC, that are capable of migration, expression of MUC1 was prevented by adding an anti-MUC1 antibody (Ab) during the maturation process. Compared with matured DC in the absence of the Ab, no difference was observed in chemokine-induced migratory behaviour between the MUC1+ and MUC1- DC populations in a standard Transwell chemotaxis assay, nor in organotypic cultures. Our data clearly demonstrate that surface MUC1 on DC does not influence intrinsic cell-motility, nor is it involved in cell-cell and cell-matrix dependent migration.


Asunto(s)
Movimiento Celular/inmunología , Células Dendríticas/inmunología , Mucina-1/inmunología , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Citocinas/farmacología , Células Dendríticas/efectos de los fármacos , Humanos , Molécula 1 de Adhesión Intercelular/inmunología , Molécula 1 de Adhesión Intercelular/metabolismo , Mucina-1/efectos de los fármacos
11.
Curr Med Chem ; 26(3): 554-563, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-28820070

RESUMEN

In the course of studying human mucin MUC1, the attitude towards this molecule has been changing time and again. Initially, the list of presumable functions of MUC1 was restricted to protecting and lubricating epithelium. To date, it is assumed to play an important role in cell signaling as well as in all stages of oncogenesis, from malignant cell transformation to tumor dissemination. The story of MUC1 is full of hopes and disappointments. However, the scientific interest to MUC1 has never waned, and the more profoundly it has been investigated, the clearer its hidden potential turned to be disclosed. The therapeutic potential of mucin MUC1 has already been noted by various scientific groups at the early stages of research. Over forty years ago, the first insights into MUC1 functions became a strong ground for considering this molecule as potential target for anticancer therapy. Therefore, this direction of research has always been of particular interest and practical importance. More than 200 papers on MUC1 were published in 2016; the majority of them are dedicated to MUC1-related anticancer diagnostics and therapeutics. Here we review the history of MUC1 studies from the very first attempts to reveal its functions to the ongoing renaissance.


Asunto(s)
Mucina-1/fisiología , Animales , Antineoplásicos/farmacología , Sistemas de Liberación de Medicamentos , Humanos , Mucina-1/efectos de los fármacos
12.
Eur J Pharmacol ; 859: 172499, 2019 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-31242439

RESUMEN

The leading causes of death in breast cancer patients are disease recurrence and metastasis. Growing evidence has suggested that metastasis possibly originates from cancer stem-like cells (CSCs). Previous studies indicated dopamine decreased CSC frequency through activating dopamine D1 receptor pathway. Hence, this study explored the efficacy of two dopamine D1 receptor agonists in lung metastasis of breast cancer and the preliminary mechanism. The two dopamine D1 receptor agonists, fenoldopam (FEN) and l-stepholidine (l-SPD), performed well in decreasing lung metastasis in 4T1 breast cancer model. And the cGMP in the primary tumor was significantly elevated while cAMP mildly elevated in FEN and l-SPD dosing groups. CSC markers (CD44+/CD24- and ALDH+) and MMP2 in 4T1 primary tumor were repressed after dopamine D1 receptor agonist administration while E-cadherin up-regulated. FEN and l-SPD also inhibited cancer stemness and cell motility in vitro, and the inhibitory effects could be reversed by dopamine D1 receptor antagonist SCH23390. Besides, FEN impacted the white blood cell increase caused by breast cancer disease showing decreased neutrophils but increased lymphocytes. Drug safety was verified in aspects of body weight, organ index and tissue section. In conclusion, dopamine D1 receptor agonists FEN and l-SPD showed efficacy in inhibiting metastasis along with good safety in breast cancer, thus providing an alternative for anti-metastasis therapy in the future. Furthermore, this study also indicates that dopamine D1 receptor may be a possible target for metastatic breast cancer treatment and even other cancers at a late stage.


Asunto(s)
Berberina/análogos & derivados , Neoplasias de la Mama/patología , Agonistas de Dopamina/farmacología , Fenoldopam/farmacología , Neoplasias Pulmonares/secundario , Células Madre Neoplásicas/patología , Receptores de Dopamina D1/metabolismo , Animales , Berberina/farmacología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Relación Dosis-Respuesta a Droga , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/prevención & control , Ratones , Mucina-1/efectos de los fármacos , Células Madre Neoplásicas/efectos de los fármacos
13.
Oncol Rep ; 19(1): 123-8, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18097585

RESUMEN

The ether lipid 1-O-octadecyl-2-O-methyl-3-glycero-phosphocholine (ET-18-OMe) inhibits cell-cell adhesion and induces invasiveness of breast cancer cells. Previously, we showed that a loss of cell-cell adhesion was due to sterical hindrance of E-cadherin by the anti-adhesive properties of the cell surface mucin episialin. Here, we demonstrated that the ether lipid ET-18-OMe induced the translocation of E-cadherin and episialin to membrane microdomains, enriched in glycosphingolipids, known to be involved in cell-cell adhesion and cell signaling. In addition, it was found that E-cadherin and clusters of episialin colocalized and associated with the glycosphingolipid, MSGb5, upon treatment with ET-18-OMe. Together, these results suggest that ET-18-OMe inhibits cell-cell adhesion by inducing the translocation of E-cadherin and episialin into MSGb5-enriched membrane microdomains, which leads to clustering and colocalization of the pro-adhesive E-cadherin and the anti-adhesive episialin thereby inhibiting cell-cell adhesion.


Asunto(s)
Antineoplásicos/farmacología , Cadherinas/efectos de los fármacos , Adhesión Celular/efectos de los fármacos , Microdominios de Membrana/efectos de los fármacos , Mucina-1/efectos de los fármacos , Éteres Fosfolípidos/farmacología , Cadherinas/metabolismo , Comunicación Celular/efectos de los fármacos , Línea Celular Tumoral , Técnica del Anticuerpo Fluorescente , Glicoesfingolípidos/metabolismo , Humanos , Inmunoprecipitación , Microdominios de Membrana/metabolismo , Mucina-1/metabolismo , Transporte de Proteínas/efectos de los fármacos
14.
Clin Cancer Res ; 13(15 Pt 2): s4652-4, 2007 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-17671159

RESUMEN

MUC1 is a mucinous glycoprotein which is overexpressed and under or aberrantly glycosylated in many human malignancies. MUC1 is associated with cellular transformation and can confer resistance to genotoxic agents. L-BLP25 is a peptide vaccine strategy that targets the exposed core peptide of MUC1. In preclinical studies, L-BLP25 induced a cellular immune response characterized by T-cell proliferation in response to MUC1 and production of IFN-gamma. Phase I and II trials have established the dose and schedule of the vaccine as well as its excellent safety profile. A randomized phase II trial of maintenance L-BLP25 versus best supportive care in patients with stage IIIB/IV non-small cell lung cancer who experienced clinical benefit from initial therapy has been reported. Updated survival analysis of this trial continues to show a strong survival trend in favor of L-BLP25 (median survival, 30.6 versus 13.3 months) in a subgroup of patients with locoregional stage IIIB disease. These promising results will be tested in a phase III trial of L-BLP25 versus placebo in patients with stage III non-small cell lung cancer after response to primary chemoradiotherapy.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/terapia , Neoplasias Pulmonares/terapia , Glicoproteínas de Membrana/uso terapéutico , Animales , Ensayos Clínicos como Asunto , Humanos , Mucina-1/efectos de los fármacos
15.
Cancer Biol Ther ; 6(4): 481-6, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18027437

RESUMEN

MUC1 is a mucin family protein, overexpressed in more than 90% of breast cancers in an underglycosylated form, exposing the core peptides of the extracellular domain that act as a potential target for antibody-mediated therapy. The highly conserved repeats of 20 amino acids VNTR varies between 20 and 125 depending on the allele. Each tandem repeat contains five potential O-glycosylation sites that have been exploited in designing cancer therapeutics. The core peptides in the tandem repeated VNTR domain are masked in normal cells and become exposed in the cancer cells associated mucins. The characteristics of the MUCI epitopes constituted with the tandem repeats and the carbohydrates present on MUC1 induce immune responses that favor targeted immunotherapy. In addition, aberrant glycosylation also plays an important role in enhancing the internalization of MUC1 into the cytoplasm making MUC1 a very attractive cytoplasmic delivery system of drugs and other therapeutic agents. MUC1 being present on most of the cancers of glandular epithelial origin, appears as a potential target for therapeutic interventions in these cancers.


Asunto(s)
Neoplasias de la Mama/terapia , Inmunoterapia , Mucina-1/efectos de los fármacos , Mucina-1/metabolismo , Secuencia de Aminoácidos , Anticuerpos Monoclonales/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Femenino , Glicosilación , Humanos , Datos de Secuencia Molecular , Mucina-1/inmunología
16.
Mol Cancer Ther ; 16(10): 2304-2314, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28729399

RESUMEN

Cutaneous T-cell lymphoma (CTCL) is a heterogeneous neoplasm and patients with relapsed/refractory disease exhibit resistance to standard therapies. We have previously demonstrated that the Mucin 1 C-terminal subunit (MUC1-C) plays a critical role in protection from oxidative stress in CTCL cells. Targeting of MUC1-C with a pharmacologic inhibitor, GO-203, was associated with apoptosis in CTCL. However, disease responses were incomplete underscoring the need for combinatorial strategies that could exploit the vulnerability of CTCL cells to oxidative signals. Cell lines, primary samples, and xenograft models of CTCL were used to assess synergy of GO-203 with decitabine, a hypomethylating agent. Present studies demonstrate that exposure of CTCL cells to decitabine in combination with GO-203, increased the generation of reactive oxygen species (ROS) levels and decreased levels of scavenger molecules, NADP, NADPH, glutathione, and TIGAR, critical to intracellular redox homeostasis. Dual exposure to GO-203 and decitabine resulted in marked downregulation of DNA methyl transferases demonstrating significant synergy of these agents in inducing global and gene specific hypomethylation. Accordingly, treatment with decitabine and GO-203 upregulated the ROS generating enzymes, NADPH oxidase 4 and dual oxidase 2 potentially due to their effect on epigenomic regulation of these proteins. In concert with these findings, exposure to decitabine and GO-203 resulted in heightened apoptotic death in CTCL cell lines, patient-derived primary samples and in a murine xenograft model. These findings indicate that decitabine intensifies MUC1-C inhibition induced redox imbalance and provides a novel combination of targeted and epigenetic agents for patients with CTCL. Mol Cancer Ther; 16(10); 2304-14. ©2017 AACR.


Asunto(s)
Azacitidina/análogos & derivados , Linfoma Cutáneo de Células T/tratamiento farmacológico , Mucina-1/genética , Estrés Oxidativo/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Azacitidina/administración & dosificación , Línea Celular Tumoral , Decitabina , Regulación Neoplásica de la Expresión Génica , Humanos , Linfoma Cutáneo de Células T/genética , Linfoma Cutáneo de Células T/patología , Ratones , Mucina-1/efectos de los fármacos , Oxidación-Reducción/efectos de los fármacos , Estrés Oxidativo/genética , Especies Reactivas de Oxígeno/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Cancer Res ; 58(18): 4079-81, 1998 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-9751614

RESUMEN

MUC1 mucin is expressed by normal and malignant epithelial cells and is thought to function through cell-cell interactions and transmembrane signal transduction events. Secreted cancer-associated MUC1 is immunosuppressive and inhibits human T-cell proliferation. We report here that newly synthesized MUC1 is expressed on the surface of mitogen-activated human T cells and is also found in soluble form in the supernatants from cultures of mitogen-activated human T cells. After removal of the mitogenic stimulus from the T-cell cultures, MUC1 expression is downregulated. The addition of anti-MUC1 monoclonal antibody to mitogen-activated cultures partially inhibits the T-cell proliferative response. These data suggest that MUC1 serves an immunodulatory function for human T lymphocytes.


Asunto(s)
Mucina-1/inmunología , Linfocitos T/inmunología , Anticuerpos Monoclonales , Regulación hacia Abajo , Humanos , Sistema Inmunológico/fisiología , Activación de Linfocitos , Mucina-1/efectos de los fármacos , Mucina-1/metabolismo , Fitohemaglutininas/farmacología , Reacción en Cadena de la Polimerasa , Linfocitos T/efectos de los fármacos
19.
Int J Mol Med ; 13(3): 459-64, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-14767580

RESUMEN

We have studied how benzyl-N-acetyl-alpha-D-galactosaminide, O-glycosylation inhibitor, affects the polymorphism and shedding of membrane-bound MUC1 mucin, and change in adhesive properties of cancer cells. In endometrial adenocarcinoma cells (Ishikawa line), high molecular weight MUC1 mucin was shed from cellular membrane and could be detected in culture medium 24 h after [14C]threonine labelling. Short-time (2 days) exposure of these cells to benzyl-N-acetyl-alpha-D-galactosaminide was associated with a reduction in sialic acid level and increase in T antigen content in cellular MUC1 mucin. These changes could be inverted after removal of the inhibitor. A longer, 6-day action of the inhibitor induced a decrease in sialic acid and T antigen levels in cellular MUC1 mucin. Benzyl-N-acetyl-alpha-D-galactosaminide treatment caused the occurrence of a few incompletely glycosylated glycoforms of MUC1 in cells, but not in culture medium. Adhesion of endometrial cells to ECM compounds (type I collagen) was increased by benzyl-N-acetyl-alpha-D-galactosaminide treatment, indicating that glycosylation of extracellular domain of MUC1 can modulate adhesive properties of cells.


Asunto(s)
Acetilgalactosamina/análogos & derivados , Neoplasias Endometriales/metabolismo , Mucina-1/metabolismo , Acetilgalactosamina/farmacología , Compuestos de Bencilo/farmacología , Adhesión Celular/efectos de los fármacos , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Colágeno Tipo I/metabolismo , Neoplasias Endometriales/patología , Femenino , Glicosilación/efectos de los fármacos , Humanos , Mucina-1/química , Mucina-1/efectos de los fármacos
20.
Neoplasma ; 49(2): 104-9, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12088101

RESUMEN

Increased expression of the epithelial mucin MUC1 has been linked to tumor aggressiveness in human breast carcinoma. In the present study, we have investigated if the factors affecting cells proliferation could influence MUC1 mucin biosynthesis and shedding from cell surface into the culture medium in two human breast cancer cell lines: MCF-7 (ER+) and MDA-MB-231 (ER-). Using MCF-7 line we found that estradiol at a concentration of 10(-7) M increased [3H]glucosamine incorporation into mucin in cell lysate approximately twofold in comparison with control cultures, and a similar increase was observed in the culture medium. The selective estrogen receptor modulator, tamoxifen (at concentrations of 10(-6) M and 10(-5) M) had a little inhibitory effect. MDA-MB-231 cells in culture were stimulated with phorbol ester PMA, the protein kinase C activator. We noted that PMA greatly stimulated MUC1 synthesis and its shedding to culture medium and that this effect was abolished by protein kinase C specific inhibitor--bisindolylmaleimide.


Asunto(s)
Neoplasias de la Mama/inmunología , Mucina-1/biosíntesis , Antígenos CD/biosíntesis , Autorradiografía , Neoplasias de la Mama/patología , División Celular , Electroforesis en Gel de Poliacrilamida , Estradiol/farmacología , Femenino , Glucosamina/metabolismo , Humanos , Mucina-1/efectos de los fármacos , Mucina-1/aislamiento & purificación , Tamoxifeno/farmacología , Tritio , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA