Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Cancer Cell Int ; 24(1): 59, 2024 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-38321552

RESUMEN

Peroxiredoxin 3 (PRDX3), a mitochondrial hydrogen peroxide scavenger, is known to be upregulated during tumorigenesis and cancer progression. In this study, we provide evidence for the first time that PRDX3 could regulate cellular signaling pathways associated with Matrix Metalloproteinase-1 (MMP-1) expression and activity in breast cancer progression. We show that shRNA-mediated gene silencing of PRDX3 inhibits cell migration and invasion in two triple-negative breast cancer cell lines. Reciprocal experiments show that PRDX3 overexpression promotes invasion and migration of the cancer cells, processes which are important in the metastatic cascade. Notably, this phenomenon may be attributed to the activation of MMP-1, which is observed to be upregulated by PRDX3 in the breast cancer cells. Moreover, immunohistochemical staining of breast cancer tissues revealed a positive correlation between PRDX3 and MMP-1 expression in both epithelial and stromal parts of the tissues. Further pathway reporter array and luciferase assay demonstrated that activation of ERK signaling is responsible for the transcriptional activation of MMP-1 in PRDX3-overexpressed cells. These findings suggest that PRDX3 could mediate cancer spread via ERK-mediated activation of MMP-1. Targeted inhibition of ERK signaling may be able to inhibit tumor metastasis in triple-negative breast cancer.

2.
Breast Cancer Res Treat ; 186(3): 655-665, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33625616

RESUMEN

PURPOSE: Breast cancer is the most common type of cancer affecting women worldwide. Phosphoglycerate dehydrogenase (PHGDH) is an oxidoreductase in the serine biosynthesis pathway. Although it has been reported to affect growth of various tumors, its role in breast cancer is largely unknown. This study aimed to analyze the expression of PHGDH in breast cancer tissue samples and to determine if PHGDH regulates breast cancer cell proliferation. METHODS: Tissue microarrays consisting of 305 cases of breast invasive ductal carcinoma were used for immunohistochemical evaluation of PHGDH expression. The role of PHGDH in breast cancer was investigated in vitro by knocking down its expression and determining the effect on cell proliferation and cell cycling, and in ovo by using a chorioallantoic membrane (CAM) assay. RESULTS: Immunohistochemical examination showed that PHGDH is mainly localized in the cytoplasm of breast cancer cells and significantly associated with higher cancer grade, larger tumor size, increased PCNA expression, and lymph node positivity. Analysis of the GOBO dataset of 737 patients demonstrated that increased PHGDH expression was associated with poorer overall survival. Knockdown of PHGDH expression in breast cancer cells in vitro resulted in a decrease in cell proliferation, reduction in cells entering the S phase of the cell cycle, and downregulation of various cell cycle regulatory genes. The volume of breast tumor in an in ovo CAM assay was found to be smaller when PHGDH was silenced. CONCLUSION: The findings suggest that PHGDH has a regulatory role in breast cancer cell proliferation and may be a potential prognostic marker and therapeutic target in breast cancer.


Asunto(s)
Neoplasias de la Mama , Fosfoglicerato-Deshidrogenasa , Neoplasias de la Mama/genética , Línea Celular Tumoral , Proliferación Celular , Femenino , Humanos , Fosfoglicerato-Deshidrogenasa/genética , Pronóstico , Serina
3.
Anal Chem ; 90(10): 6193-6198, 2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29608843

RESUMEN

Cell surface receptors play an important role in mediating cell communication and are used as disease biomarkers and therapeutic targets. We present a one-pot molecular toolbox, which we term the split proximity circuit (SPC), for the autonomous detection and visualization of cell surface receptor clusters. Detection was powered by antibody recognition and a series of autonomous DNA hybridization to achieve localized, enzyme-free signal amplification. The system under study was the human epidermal growth factor receptor (HER) family, that is, HER2:HER2 homodimer and HER2:HER3 heterodimer, both in cell lysate and in situ on fixed whole cells. The detection and imaging of receptors were carried out using standard microplate scans and confocal microscopy, respectively. The circuit operated specifically with minimal leakages and successfully captured the receptor expression profiles on three cell types without any intermediate washing steps.


Asunto(s)
ADN/química , Receptor ErbB-2/análisis , Receptor ErbB-3/análisis , Ensayo de Inmunoadsorción Enzimática , Humanos , Microscopía Confocal , Células Tumorales Cultivadas
4.
Exp Mol Pathol ; 105(3): 364-370, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30385176

RESUMEN

A-kinase anchor protein 12 (AKAP12) also known as Gravin and SSeCKS, is a novel potent scaffold protein for many key signaling factors, such as protein kinase C (PKC), PKA, cyclins as well as F-actin. AKAP12 expression is known to be suppressed in several human malignancies including breast, prostate, gastric and colon cancers. In this study, we evaluated the role of AKAP12 in the migration of breast cancer cells, an important cellular process in cancer progression. AKAP12 gene expression was analyzed in human breast cancer tissues using the Gene expression-based Outcome for Breast cancer Online (GOBO) database and TissueScan array, followed by relapse free survival (RFS) analysis with the Kaplan-Meier Plotter. AKAP12 protein was then analyzed in normal MCF10A breast cell line and six different breast cancer cell lines (AU565, Hs578T, MCF7, MDA-MB-231, T47D and ZR751). After which, siRNA-mediated knockdown of AKAP12 was carried out in MCF10A, MDA-MB-231 and Hs578T cells, followed by phenotypic assays. AKAP12 was observed to be reduced in breast cancer tissues as analyzed by GOBO and TissueScan array. Kaplan Meier survival analysis revealed that patients with AKAP12 gene expression had a higher RFS survival. There was also decreased AKAP12 protein expression in breast cancer cell lines compared to MCF10A normal epithelial breast cell line. Knockdown of AKAP12 in both MCF10A cells and Hs578T cells induced cell migration but did not alter cell proliferation. Moreover, siAKAP12 in aggressive MDA-MB-231 breast cancer cells led to an increase in cell migration. Immunofluorescence analysis of AKAP12 depleted MCF10A cells also revealed formation of thick stress fibers which could affect cell migration. Hence, the findings in this study suggest that AKAP12 is a potential metastasis suppressor in breast cancer.


Asunto(s)
Proteínas de Anclaje a la Quinasa A/genética , Proteínas de Anclaje a la Quinasa A/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Movimiento Celular , Bases de Datos como Asunto , Regulación hacia Abajo , Femenino , Perfilación de la Expresión Génica , Humanos , Estimación de Kaplan-Meier , Transcriptoma
5.
Breast Cancer Res Treat ; 151(1): 99-111, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25868865

RESUMEN

Triple-negative breast cancers (TNBCs) are clinically aggressive tumors with limited treatment options. We examined the clinicopathological associations and prognostic implications of FGFR1 and FGFR2 expression in TNBCs. Tissue microarrays constructed from TNBCs were immunostained with FGFR1 and FGFR2, and scored by intensity and percentage of tumor cells stained per intensity for each subcellular compartment, which were correlated with clinicopathological parameters and survival. Cell migration following siRNA-mediated silencing of the FGFR1 gene in TNBC cell lines was also performed. 714 cases were informative for FGFR1 and FGFR2 immunostaining. Thresholds were defined as at least 1 % of cells stained and H-score of 100 or more. Proportions positive by each threshold were, respectively, 89.9 %, 7.1 % for FGFR1 (cytoplasm); 36.8 %, 7.8 % for FGFR2 (cytoplasm); and 33.5 %, 5.2 % for FGFR2 (membrane). Significant associations included FGFR1 and FGFR2 immunostaining for lobular subtype, FGFR2 immunostaining with lower grade, and more basal-like cancers with H-scores of 100 or more FGFR1 immunostaining. Multivariate Cox regression analysis showed FGFR1 expression in TNBCs to be independently prognostic for overall survival (OS) at both thresholds. Cases completely negative (less than 1 % staining) for FGFR1 immunostaining showed improved OS, while those with H-score of 100 or more immunostaining had the worst OS. Cell line studies revealed up-regulation of the FGFR1 gene in the MDA-MB-231 and Hs578T TNBC cells, and specific knockdown of FGFR1 expression significantly reduced cell migration in MDA-MB-231 cell line. In conclusion, FGFR1 expression in TNBCs is independently prognostic of OS, and H-score of 100 or more FGFR1 immunostaining may define tumors that have treatment potential via FGFR signaling inhibition.


Asunto(s)
Invasividad Neoplásica/genética , Pronóstico , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/biosíntesis , Neoplasias de la Mama Triple Negativas/genética , Adulto , Anciano , Anciano de 80 o más Años , Línea Celular Tumoral , Movimiento Celular/genética , Supervivencia sin Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Persona de Mediana Edad , Invasividad Neoplásica/patología , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Análisis de Matrices Tisulares , Neoplasias de la Mama Triple Negativas/patología
6.
Histopathology ; 67(5): 709-18, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25817822

RESUMEN

AIMS: Identification of prognostic and predictive biomarkers for breast cancer is essential to better stratify patients for treatment and evaluate patient outcome. AT-rich interactive domain-containing protein 1B (ARID1B) is implicated in cell proliferation, but its role in tumorigenesis remains unclear. METHODS AND RESULTS: Immunohistochemical analysis of ARID1B expression using breast cancer tissue microarrays containing 156 breast invasive ductal carcinoma patient samples and subsequent statistical data analysis based on ARID1B immunoreactivity score were performed to examine the correlation between clinicopathological parameters in breast cancer and ARID1B expression. In-vitro assays were also performed to study the role of ARID1B in cell cycle progression. Univariate analysis revealed that high ARID1B expression is correlated closely with histological grade (P = 0.045) and size (P = 0.043) of invasive breast cancer. These findings were confirmed by multivariate analysis. Notably, increased ARID1B expression was frequently detected in the aggressive triple-negative breast cancer subtypes (P = 0.039) and associated with decreased 5-year disease-free survival rate. Lastly, MDA-MB-231 cells with reduced ARID1B activity displayed a delay in G1 to S phase cell cycle transition and consequently showed a decrease in cell proliferation compared with controls (P < 0.001). CONCLUSIONS: ARID1B potentially serves as a valuable prognostic and predictive biomarker as well as a therapeutic target in breast cancer.


Asunto(s)
Biomarcadores de Tumor/análisis , Neoplasias de la Mama/patología , Carcinoma Ductal de Mama/patología , Proteínas de Unión al ADN/biosíntesis , Factores de Transcripción/biosíntesis , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/mortalidad , Carcinoma Ductal de Mama/metabolismo , Carcinoma Ductal de Mama/mortalidad , Proteínas de Unión al ADN/análisis , Femenino , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Persona de Mediana Edad , ARN Interferente Pequeño , Reacción en Cadena en Tiempo Real de la Polimerasa , Análisis de Matrices Tisulares , Factores de Transcripción/análisis , Transfección
7.
Mol Biol Cell ; 34(3): ar13, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36598812

RESUMEN

Rho GTPases regulate cell morphogenesis and motility under the tight control of guanine nucleotide exchange factors (GEFs) and GTPase-activating proteins (GAPs). However, the underlying mechanism(s) that coordinate their spatiotemporal activities, whether separately or together, remain unclear. We show that a prometastatic RhoGAP, ARHGAP8/BPGAP1, binds to inactive Rac1 and localizes to lamellipodia. BPGAP1 recruits the RacGEF Vav1 under epidermal growth factor (EGF) stimulation and activates Rac1, leading to polarized cell motility, spreading, invadopodium formation, and cell extravasation and promotes cancer cell migration. Importantly, BPGAP1 down-regulates local RhoA activity, which influences Rac1 binding to BPGAP1 and its subsequent activation by Vav1. Our results highlight the importance of BPGAP1 in recruiting Vav1 and Rac1 to promote Rac1 activation for cell motility. BPGAP1 also serves to control the timing of Rac1 activation with RhoA inactivation via its RhoGAP activity. BPGAP1, therefore, acts as a dual-function scaffold that recruits Vav1 to activate Rac1 while inactivating RhoA to synchronize both Rho and Rac signaling in cell motility. As epidermal growth factor receptor (EGFR), Vav1, RhoA, Rac1, and BPGAP1 are all associated with cancer metastasis, BPGAP1 could provide a crucial checkpoint for the EGFR-BPGAP1-Vav1-Rac1-RhoA signaling axis for cancer intervention.


Asunto(s)
Movimiento Celular , Proteínas Activadoras de GTPasa , Humanos , Secuencia de Aminoácidos , Receptores ErbB/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Proteínas de Unión al GTP rho/metabolismo
8.
J Biol Chem ; 285(38): 29231-8, 2010 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-20630868

RESUMEN

Although mutations in the parkin gene are frequently associated with familial Parkinsonism, emerging evidence suggests that parkin also plays a role in cancers as a putative tumor suppressor. Supporting this, we show here that parkin expression is dramatically reduced in several breast cancer-derived cell lines as well as in primary breast cancer tissues. Importantly, we found that ectopic parkin expression in parkin-deficient breast cancer cells mitigates their proliferation rate both in vitro and in vivo, as well as reduces the capacity of these cells to migrate. Cell cycle analysis revealed the arrestment of a significant percentage of parkin-expressing breast cancer cells at the G1-phase. However, we did not observe significant changes in the levels of the G1-associated cyclin D1 and E. On the other hand, the level of cyclin-dependent kinase 6 (CDK6) is dramatically and selectively elevated in parkin-expressing breast cancer cells, the extent of which correlates well with the expression of parkin. Interestingly, a recent study demonstrated that CDK6 restrains the proliferation of breast cancer cells. Taken together, our results support a negative role for parkin in tumorigenesis and provide a potential mechanism by which parkin exerts its suppressing effects on breast cancer cell proliferation.


Asunto(s)
Neoplasias de la Mama/metabolismo , Quinasa 6 Dependiente de la Ciclina/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Neoplasias de la Mama/genética , Ciclo Celular/genética , Ciclo Celular/fisiología , Línea Celular , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Quinasa 6 Dependiente de la Ciclina/genética , Femenino , Fase G1/genética , Fase G1/fisiología , Humanos , Técnicas In Vitro , Ratones , Ratones Endogámicos NOD , Ratones SCID , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa , Ubiquitina-Proteína Ligasas/genética
9.
Cancer Lett ; 452: 119-131, 2019 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-30905819

RESUMEN

Y-box binding protein-1 (YB-1), an important transcription and translation regulator protein, is known to increase cancer cell invasiveness and spreading. Here, we report its role in breast cancer, particularly in mediating cell invasion in triple-negative breast cancer (TNBC). YB-1 stable knockdown (shYB-1) significantly reduced the invasive potential of MDA-MB-231 TNBC cells in 2D and 3D (spheroid) cultures. Whole proteome mass spectrometry analysis showed an enrichment of cell adhesion and cell to matrix interaction proteins, notably, matrix metalloproteinase-1 (MMP1) and beta-catenin (CTNNB1), which are known to play critical roles in cancer metastasis. shYB-1 cells exhibited substantial downregulation of MMP1 and CTNNB1 mRNA and protein expression, with reduced MMP1 enzyme activity. YB-1 was also observed to bind to the promoter of MMP1 and overexpression of MMP1 plasmid in shYB-1 cells increased cell invasion. Finally, analysis of tumour samples from the Gene Expression-Based Outcome for Breast Cancer Online (GOBO) database revealed that high gene expressions of YBX1, MMP1 and CTNNB1 predict for a significantly lower 10-year distant metastasis free survival. Altogether, this study shows that YB-1 mediates breast cancer invasion and metastasis via regulation of MMP1 and beta-catenin.


Asunto(s)
Metaloproteinasa 1 de la Matriz/biosíntesis , Neoplasias de la Mama Triple Negativas/patología , Proteína 1 de Unión a la Caja Y/genética , beta Catenina/biosíntesis , Movimiento Celular/genética , Proliferación Celular/genética , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Metaloproteinasa 1 de la Matriz/genética , Invasividad Neoplásica/genética , Regiones Promotoras Genéticas/genética , Interferencia de ARN , ARN Mensajero/biosíntesis , ARN Interferente Pequeño/genética , Esferoides Celulares , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/mortalidad , Células Tumorales Cultivadas , beta Catenina/genética
10.
Int J Oncol ; 53(6): 2579-2589, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30221675

RESUMEN

Y-box binding protein-1 (YB-1) facilitates cancer chemoresistance through the upregulation of ATP-binding cassette (ABC) transporters associated with multidrug resistance, which is one of the primary obstacles in cancer treatment. Since aberrant Janus kinase (JAK)/signal transducer and activator of transcription (STAT) signaling is also implicated in chemoresistance in numerous human malignancies, the interaction between YB-1 and JAK/STAT signaling was explored underlying the chemoresistance of NUGC3 gastric cancer cells. It was demonstrated that YB-1 translocated into the nuclei of NUGC3 cells exposed to doxorubicin hydrochloride, suggesting its important role in chemoresistance. Consistently, knockdown of YB-1 significantly decreased the chemoresistance of cells to doxorubicin hydrochloride and epirubicin hydrochloride, as evidenced by a decrease in cell viability. Notably, JAK inhibitor AG490 treatment further decreased the cell viability caused by YB-1 inhibition and doxorubicin hydrochloride. It was also observed that YB-1 transcriptionally regulated the ABCC3 transporter, whereas STAT3 modulated ABCC2 transporter levels. These findings suggest that YB-1 and STAT3 act together to facilitate chemoresistance via modulating the expression of different ABC transporters in NUGC3 cells. Notably, siYB-1 did not exhibit any significant effect on STAT3 expression. Similarly, siSTAT3 failed to alter YB-1 expression, suggesting that the two may not regulate each other in a mutual manner. However, double knockdown of YB-1 and STAT3 led to a synergistic inhibition of cell invasion in NUGC3 cells. Nonetheless, the combined treatment of YB-1 antagonists with STAT3 inhibitors may serve as an effective therapy in gastric cancer.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Resistencia a Antineoplásicos , Factor de Transcripción STAT3/metabolismo , Neoplasias Gástricas/metabolismo , Proteína 1 de Unión a la Caja Y/metabolismo , Línea Celular Tumoral , Núcleo Celular/metabolismo , Supervivencia Celular/efectos de los fármacos , Doxorrubicina/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Proteína 2 Asociada a Resistencia a Múltiples Medicamentos , Transporte de Proteínas/efectos de los fármacos , Factor de Transcripción STAT3/genética , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/genética , Tirfostinos/farmacología , Proteína 1 de Unión a la Caja Y/genética
11.
Exp Biol Med (Maywood) ; 242(2): 140-147, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27633575

RESUMEN

Peroxiredoxin I to VI (PRX I-VI), a family of highly conserved antioxidants, has been implicated in numerous diseases. There have been reports that PRXs are expressed aberrantly in a variety of tumors, implying that they could play an important role in carcinogenesis. Epigenetic mechanisms such as DNA methylation, histone modifications, and microRNAs have been reported to modulate expression of PRXs. In addition, the use of epigenetic regulators, such as histone deacetylases, has been demonstrated to restore PRX to normal levels, indicating that the reversible nature of epigenetics can be exploited for future treatments.


Asunto(s)
Antioxidantes/metabolismo , Epigénesis Genética/genética , Neoplasias/genética , Neoplasias/patología , Peroxirredoxinas/metabolismo , Carcinogénesis/patología , Metilación de ADN/genética , Código de Histonas/fisiología , Humanos , Peróxido de Hidrógeno/metabolismo , MicroARNs/genética , Estrés Oxidativo/fisiología
12.
Oncotarget ; 8(70): 115370-115383, 2017 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-29383166

RESUMEN

Dysregulated JAK/STAT signaling has been implicated in the molecular pathogenesis of gastric cancer. However, downstream effectors of STAT signaling that facilitate gastric carcinogenesis remain to be explored. We previously identified the Drosophila ortholog of human GRAMD1B in our genome-wide RNAi screen to identify novel components of the JAK/STAT signaling pathway in Drosophila. Here, we examined the involvement of GRAMD1B in JAK/STAT-associated gastric carcinogenesis. We found that GRAMD1B expression is positively regulated by JAK/STAT signaling and GRAMD1B inhibition decreases STAT3 levels, suggesting the existence of a positive feedback loop. Consistently, GRAMD1B and JAK/STAT signaling acted synergistically to promote gastric cancer cell survival by upregulating the expression of the anti-apoptotic molecule Bcl-xL. Interestingly, our immunohistochemical analysis for GRAMD1B revealed a gradual loss of cytoplasmic staining but an increase in the nuclear accumulation of GRAMD1B, as gastric tissue becomes malignant. GRAMD1B expression levels were also found to be significantly associated with clinicopathological features of the gastric cancer patients, particularly the tumor grades and lymph node status. Moreover, GRAMD1B and pSTAT3 (Tyr705) showed a positive correlation in gastric tissues, thereby confirming the existence of a close link between these two signaling molecules in vivo. This new knowledge about JAK/STAT-GRAMD1B regulation deepens our understanding of JAK/STAT signaling in gastric carcinogenesis and provides a foundation for the development of novel biomarkers in gastric cancer.

13.
J Clin Pathol ; 69(10): 878-83, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26926102

RESUMEN

AIMS: Altered expression of the Claudin (CLDN) superfamily of tight junction proteins has been reported in breast cancer. The aim of this study was to examine the immunohistochemical expression of CLDN 12 and its prognostic significance in breast cancer tissues. METHODS: Immunohistochemical expression of CLDN 12 was performed on tissue microarrays consisting of 232 cases of breast carcinoma and correlated with clinicopathological features as well as survival of the patients with breast cancer. RESULTS: For the estrogen receptor (ER)-negative subgroup of patients with breast cancer, CLDN 12 expression was shown to be an independent predictor of poor overall survival (HR=2.345; p=0.020) and disease-free survival (HR=2.177; p=0.026) but not for the ER-positive tumours. CONCLUSIONS: The findings suggest that CLDN 12 expression could be clinically useful for predicting the survival of the ER-negative subgroup of patients with breast cancer.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/metabolismo , Claudinas/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias de la Mama/diagnóstico , Supervivencia sin Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunohistoquímica , Persona de Mediana Edad , Pronóstico , Receptores de Estrógenos/metabolismo , Análisis de Matrices Tisulares , Adulto Joven
14.
Int J Oncol ; 47(5): 1617-26, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26398764

RESUMEN

Gastric carcinoma remains one of the most prevalent forms of cancer worldwide, despite the decline in incidence rates, increased awareness of the disease and advancement in treatment strategies. Helicobacter pylori infection, dietary factors, lifestyle influences and various genetic aberrations have been shown to contribute to the development and progression of gastric cancer. Recent studies on the genomic landscape of gastric adenocarcinoma have identified several key signaling molecules, including epidermal growth factor receptor family (ErbB) members, vascular endothelial growth factor receptor family (VEGFR) members and PI3K/Akt/mTOR pathway components, that have been implicated in the molecular pathogenesis of gastric cancers. However, clinical trials with compounds that target these molecules have failed to show a significant improvement in overall survival rates when supplemented with conventional therapies. Therefore, it is essential to identify effective prognostic and/or diagnostic biomarkers and develop molecular targeted therapies. The JAK/STAT cascade is a principal signal transduction pathway in cytokine and growth factor signaling, regulating various cellular processes such as cell proliferation, differentiation, migration and survival. Numerous in vivo and in vitro studies have shown that dysregulated JAK/STAT signaling is a driving force in the pathogenesis of various solid cancers as well as hematopoietic malignancies. Hence, a large number of preclinical and clinical studies of drugs targeting this pathway are currently underway. Notably, aberrant JAK/STAT signaling has also been implicated in gastric cancers. In this review, we focus on the ongoing research on the JAK/STAT cascade in gastric carcinoma and discuss the therapeutic potential of targeting JAK/STAT signaling for the treatment of gastric cancer.


Asunto(s)
Carcinoma/genética , Quinasas Janus/genética , Factores de Transcripción STAT/genética , Neoplasias Gástricas/genética , Carcinoma/patología , Carcinoma/terapia , Proliferación Celular/genética , Receptores ErbB/genética , Humanos , Quinasas Janus/metabolismo , Terapia Molecular Dirigida , Factores de Transcripción STAT/metabolismo , Transducción de Señal , Neoplasias Gástricas/patología , Neoplasias Gástricas/terapia
15.
J Clin Invest ; 125(4): 1401-18, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25798617

RESUMEN

Activating mutations in the RAS oncogene are common in cancer but are difficult to therapeutically target. RAS activation promotes autophagy, a highly regulated catabolic process that metabolically buffers cells in response to diverse stresses. Here we report that casein kinase 1α (CK1α), a ubiquitously expressed serine/threonine kinase, is a key negative regulator of oncogenic RAS-induced autophagy. Depletion or pharmacologic inhibition of CK1α enhanced autophagic flux in oncogenic RAS-driven human fibroblasts and multiple cancer cell lines. FOXO3A, a master longevity mediator that transcriptionally regulates diverse autophagy genes, was a critical target of CK1α, as depletion of CK1α reduced levels of phosphorylated FOXO3A and increased expression of FOXO3A-responsive genes. Oncogenic RAS increased CK1α protein abundance via activation of the PI3K/AKT/mTOR pathway. In turn, elevated levels of CK1α increased phosphorylation of nuclear FOXO3A, thereby inhibiting transactivation of genes critical for RAS-induced autophagy. In both RAS-driven cancer cells and murine xenograft models, pharmacologic CK1α inactivation synergized with lysosomotropic agents to inhibit growth and promote tumor cell death. Together, our results identify a kinase feedback loop that influences RAS-dependent autophagy and suggest that targeting CK1α-regulated autophagy offers a potential therapeutic opportunity to treat oncogenic RAS-driven cancers.


Asunto(s)
Autofagia/fisiología , Quinasa de la Caseína I/fisiología , Proteínas de Neoplasias/fisiología , Neoplasias/enzimología , Proteína Oncogénica p21(ras)/fisiología , Secuencia de Aminoácidos , Animales , Quinasa de la Caseína I/antagonistas & inhibidores , Retroalimentación Fisiológica , Fibroblastos/enzimología , Fibroblastos/fisiología , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Datos de Secuencia Molecular , Trasplante de Neoplasias , Fosfatidilinositol 3-Quinasas/fisiología , Fosforilación , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas c-akt/fisiología , ARN Interferente Pequeño/farmacología , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Serina-Treonina Quinasas TOR/fisiología , Activación Transcripcional
16.
Exp Biol Med (Maywood) ; 239(4): 387-93, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24599885

RESUMEN

Scorpions and their venoms have been used in traditional medicine for thousands of years in China, India and Africa. The scorpion venom is a highly complex mixture of salts, nucleotides, biogenic amines, enzymes, mucoproteins, as well as peptides and proteins (e.g. neurotoxins). One of the recently observed biological properties of animal venoms and toxins is that they possess anticancer potential. An increasing number of studies have shown that scorpion venoms and toxins can decrease cancer growth, induce apoptosis and inhibit cancer progression and metastasis in vitro and in vivo. Several active molecules with anticancer activities, ranging from inhibition of proliferation and cell cycle arrest to induction of apoptosis and decreasing cell migration and invasion, have been isolated from scorpion venoms. These observations have shed light on the application of scorpion venoms and toxins as potential novel cancer therapeutics. This mini-review focuses on the anticancer potential of scorpion venoms and toxins and the possible mechanisms for their antitumor activities.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Venenos de Escorpión/química , Antineoplásicos/aislamiento & purificación , Apoptosis , Descubrimiento de Drogas , Humanos , Neoplasias/patología
17.
Exp Biol Med (Maywood) ; 238(4): 344-9, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23759999

RESUMEN

Gastric carcinoma arises from aberrant growth of normal gastric mucosa. There is increasing evidence that claudins (CLDNs) may play a critical role in the significant steps of gastric tumorigenesis, from metaplasia to metastasis. The CLDN family which consists of at least 27 member proteins is known to mediate selective permeability in cellular tight junctions. It is now established that CLDNs are differentially altered in gastric cancer and CLDN proteins are believed to play different roles in the growth and progression of gastric cancer.


Asunto(s)
Transformación Celular Neoplásica/patología , Claudinas/fisiología , Neoplasias Gástricas/fisiopatología , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Neoplasias Gástricas/patología , Uniones Estrechas/fisiología
18.
Anat Rec (Hoboken) ; 295(9): 1415-20, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22807344

RESUMEN

Serglycin is a multifunctional molecule and one of the first proteoglycans to be cloned. In this manuscript, we examine the physiological roles of serglycin in immunity, hemostasis, cell growth, apoptosis, and reproduction. In addition, we review recent studies on the involvement of serglycin in various pathological conditions, including cancer, inflammatory diseases, and platelet disorders.


Asunto(s)
Proteoglicanos/fisiología , Proteínas de Transporte Vesicular/fisiología , Animales , Apoptosis/fisiología , Aterosclerosis/inmunología , Aterosclerosis/metabolismo , Aterosclerosis/patología , Aumento de la Célula , Hemostasis/fisiología , Humanos , Mediadores de Inflamación/sangre , Mediadores de Inflamación/metabolismo , Mediadores de Inflamación/fisiología , Neoplasias/inmunología , Neoplasias/metabolismo , Neoplasias/patología , Proteoglicanos/sangre , Reproducción/fisiología , Proteínas de Transporte Vesicular/sangre
19.
Int J Oncol ; 36(2): 359-64, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20043069

RESUMEN

Peroxiredoxin III (Prx III), an antioxidant protein found in mitochondria, plays an essential role in mitochondrial homeostasis. Aberrant expression of Prx III has been implicated in the tumorigenesis of various cancers. In this study, we evaluated the expression of Prx III in breast cancer tissues and elucidated its role in cell proliferation, a hallmark of cancer. Breast tissue microarrays comprising 106 breast cancer sections were stained with Prx III antibody using immunohistochemisty and correlated with proliferating cell nuclear antigen (PCNA) immunostaining. To validate the role of Prx III in cell proliferation, expression of Prx III was analyzed at the mRNA and protein levels by real-time RT-PCR, Western blotting and immunofluorescence in vitro. siRNA mediated silencing of Prx III in MDA-MB-231 breast cancer cells was performed and the effect on the cell cycle was examined. Prx III expression in patient tissue microarray samples was found to be positively associated with PCNA immunostaining, a proliferative marker. Prx III was expressed in both MCF-7 and MDA-MB-231 breast cancer cell lines and transient transfection with siPrx III in MDA-MB-231 cells induced inhibition of cell proliferation and cell cycle arrest. The data suggests that Prx III has a significant role in cell cycle regulation and could be a potential proliferation marker in breast cancer.


Asunto(s)
Neoplasias de la Mama/genética , Proliferación Celular , Silenciador del Gen/fisiología , Peroxirredoxinas/genética , Biomarcadores de Tumor/análisis , Biomarcadores de Tumor/genética , Western Blotting , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Ciclo Celular/genética , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Inmunohistoquímica , Peroxiredoxina III , Peroxirredoxinas/metabolismo , ARN Interferente Pequeño , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Matrices Tisulares , Transfección
20.
Exp Biol Med (Maywood) ; 234(9): 1086-94, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19596828

RESUMEN

Depending on the amounts present, reactive oxygen species can exert either beneficial or deleterious effect to cells. In the present study, we observed a decrease in cell viability concomitant with an increase of malondialdehyde concentration in hydrogen peroxide (H(2)O(2))-treated MCF-7 breast cancer cells. There was also a concurrent G1/S phase cell cycle arrest with increased apoptosis in H(2)O(2)-treated cells. Analysis of 84 oxidative stress related genes showed that five genes were significantly and differentially regulated, namely, Cytoglobin (CYGB), Forkhead box M1 (FOXM1), NADPH oxidase (NOX5), Nudix (nucleoside diphosphate linked moiety X)-type motif 1 (NUDT1) and Selenoprotein P1 (SEPP1) genes with H(2)O(2) treatment. It would seem that oxidative stress induces cell cycle arrest in the breast cancer by modulation of these genes. Manipulation of these genes, in particular FOXM1, a proliferation-specific gene associated with human malignancies, could stifle cancer progression and enhance the therapeutic efficacy of drugs which exert their effects by oxidative stress.


Asunto(s)
Ciclo Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Peróxido de Hidrógeno/toxicidad , Estrés Oxidativo , Apoptosis , Línea Celular Tumoral , Femenino , Perfilación de la Expresión Génica , Humanos , Malondialdehído/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA