Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Eur J Surg Oncol ; 49(10): 107040, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37672825

RESUMEN

OBJECTIVE: Internal mammary nodes are important in breast cancer prognosis, but their diagnosis is often missed in clinical practice, leading to inaccurate staging and treatment. We developed a validated nomogram to predict the presence of internal mammary sentinel nodes (IMSN) metastasis. METHODS: A total of 864 sequential IMSN biopsy procedures from a prospective studies database of 1505 cases were used for model development and validation. Multivariable logistic regression was performed on 519 sequential IMSN biopsy procedures from multi-center data between August 2018 and July 2022 to predict the presence of IMSN metastasis. A nomogram was developed based on the logistic regression model and subsequently applied to 345 sequential IMSN biopsy procedures from single-center data between November 2011 and July 2018. The model's discrimination was assessed using the area under the receiver operating characteristic curve. RESULTS: The overall frequency of IMSN metastasis was 17.0% in our study. A predictive model for IMSN metastasis was constructed using tumor size, tumor location, lymphovascular invasion, the number of positive axillary nodes (P < 0.05 for all variables in multivariate analysis), and histological grade (P < 0.05 only in univariate analysis). The nomogram was accurate, with a concordance index of 0.84 in the bootstrapping analysis and an area under the receiver operating characteristic curve of 0.80 in the validation population. CONCLUSION: Our nomogram provides an accurate and validated multivariable predictive model for estimating the individual likelihood of having IMSN metastasis. This may be useful for personalized treatment decisions regarding internal mammary radiotherapy in breast cancer patients.


Asunto(s)
Neoplasias de la Mama , Humanos , Femenino , Neoplasias de la Mama/cirugía , Nomogramas , Metástasis Linfática/patología , Estudios Prospectivos , Ganglios Linfáticos/patología , Biopsia del Ganglio Linfático Centinela
2.
Cancer Lett ; 452: 79-89, 2019 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-30851420

RESUMEN

Cancer-associated fibroblasts (CAFs) remain active even in the absence of cancer cells. However, the molecular mechanism underlying the sustained active status of CAFs is largely unrevealed. We found that in CAFs, DNMT3B was not only a target of miR-200b, miR-200c and miR-221, but was able to induce DNA methylation of miR-200s promoters. DNMT3B eventually reached a stably high level by the counteracting effect of decreasing miR-200b/c and increasing miR-221 in normal fibroblasts (NFs) with long-term exogenous TGF-ß1 treatment, and DNMT3B further led to a low level of miR-200s which established CAF activation. Meanwhile, miR-200s/miR-221/DNMT3B signaling sustained autocrine TGF-ß1 maintaining active CAF status. Destruction of the autocrine TGF-ß1/miR-200s/miR-221/DNMT3B signaling led to demethylation of miR-200s promoters and further restored the NF phenotypes. Moreover, we confirmed that TCF12, the target of miR-141, stimulated c-Myc/Cyclin D1 axis in breast cancer cells to promote cancer growth by enhancing CXCL12 of CAFs. The current study reveals that the TGF-ß1/miR-200s/miR-221/DNMT3B regulatory loop is responsible for the maintenance of CAFs status and is also necessary for CAF function in promoting malignance of breast cancer, which provides a potential target for CAF-driven therapeutic strategy.


Asunto(s)
Neoplasias de la Mama/patología , Fibroblastos Asociados al Cáncer/patología , ADN (Citosina-5-)-Metiltransferasas/genética , MicroARNs/genética , Factor de Crecimiento Transformador beta1/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Neoplasias de la Mama/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Ciclina D1/metabolismo , Metilación de ADN , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Redes Reguladoras de Genes/genética , Humanos , Células MCF-7 , Proteínas Proto-Oncogénicas c-myc/metabolismo , Transducción de Señal , ADN Metiltransferasa 3B
3.
Front Oncol ; 9: 1551, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-32117784

RESUMEN

Background and Objectives: Whether post-mastectomy radiotherapy (PMRT) could improve prognosis for T1-2 breast cancer patients with one to three lymph node metastases remains controversial. The present study aimed to determine the significance of PMRT in patients with T1-2N1M0 breast cancer. Methods: Data of 45,646 patients from the Surveillance, Epidemiology, and End Results (SEER) database were analyzed; 12,585 matched patients were divided into a PMRT group and non-radiotherapy group (no-PMRT), respectively, using the propensity score matching method. Univariate and multivariate analyses were performed to determine the prognostic factors of breast cancer, and subgroup analysis was performed according to the number of lymph node metastases. Results: With the median follow-up of 62 months, 5-year cancer-specific survival was 91.48% in the PMRT group and 91.88% in the no-PMRT group (P = 0.405). PMRT did not improve the breast cancer-specific survival (BCSS) in patients with stage T1-2N1M0 (HR = 0.99, 95% CI = 0.92-1.06, P = 0.715). In subgroup analysis, radiotherapy improved the BCSS in patients with three nodes positive, with the 5-year BCSS at 88.5% in the radiation group and 86.6% in the no-radiation group (HR = 0.78, 95% CI = 0.65-0.90, P < 0.001). In patients with two nodes positive, 5-year BCSS was 90.3% in the PMRT group and 89.5% in the no-PMRT group, with no significant difference between the two groups (HR = 0.96, 95% CI = 0.85-1.09, P = 0.552). In patients with one node positive, 5-year BCSS was higher in the no-PMRT group (92.1%) than that in the PMRT group (90.8%); radiotherapy increased the cancer-related death compared with those who did not receive it (HR = 1.21, 95% CI = 1.08-1.36, P = 0.002). Conclusion: The benefit of PMRT in T1-2N1M0 patients was obviously different, and the recommendation of PMRT for this population should be individualized. PMRT should be considered for patients with three nodes positive, should be suggested cautiously in those with two nodes positive, and could be omitted in those with one node positive.

4.
Breast Cancer Res Treat ; 170(1): 35-43, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29480449

RESUMEN

PURPOSE: To assess the predictive role of pretreatment ki67 and Ki67 changes in breast cancer (BC) patients treated with neoadjuvant chemotherapy (NAC) in various molecular subtypes. METHODS: 1010 BC patients who had undergone anthracycline and taxane-based NAC from January 2012 to July 2017 were retrospectively analyzed. Clinical and pathological parameters of the patients were retrieved and the predictive factors for NAC response were evaluated. RESULTS: 705 patients showed clinical response (cRes), and 131 patients acquired pathologic complete response (pCR). Patients with higher pretreatment Ki67 (≥ 14%), tumor size ≥ 4 cm, and positive clinical nodal had better clinical therapy response, while patients with negative ER and PR, higher pretreatment Ki67 (≥ 14%), and tumor size < 4 cm were more probable to attain pCR. The pretreatment Ki67 could be used as a predictor of NAC only in luminal subtypes, and 25.5% were identified as an ideal cut-off point to differentiate the cRes from non-cRes cases. Although a decrease in Ki67 had been found in almost all molecular subtypes after NAC, no statistically significant differences were found in the decrease of Ki67 were validated between the cRes and non-cRes group in HER2-rich and triple-negative subtypes (P = 0.488 and P = 0.111, respectively). CONCLUSIONS: The best cut-off for pretreatment Ki67 in predicting the connection with the tumor size lessening was 25.5% in luminal subtype. Aggressive adjuvant systemic treatments should be considered for patients with HER2-rich and triple-negative subtype who exhibit tumor shrinkage in NAC but still have high levels of Ki67.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Antígeno Ki-67/genética , Receptor ErbB-2/genética , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Adulto , Anciano , Biomarcadores de Tumor/genética , Neoplasias de la Mama/clasificación , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Receptor alfa de Estrógeno/genética , Femenino , Humanos , Estimación de Kaplan-Meier , Persona de Mediana Edad , Terapia Neoadyuvante/efectos adversos , Receptores de Progesterona/genética , Estudios Retrospectivos , Neoplasias de la Mama Triple Negativas/clasificación , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología
5.
Theranostics ; 7(16): 3972-3988, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29109792

RESUMEN

Tumor microenvironment contributes to tumor angiogenesis. However, the role of the activated cancer associated-fibroblasts (CAFs) in angiogenesis is still unclear. Here we report that miR-205/YAP1 signaling in the activated stromal fibroblasts plays a critical role in VEGF-independent angiogenesis in breast tumor. Methods: miR-205 expression was assessed by quantitative real-time polymerase chain reaction (qRT-PCR); YAP1 expression by qRT-PCR, western blotting and immunohistochemistry; IL11 and IL15 expression by qRT-PCR, western blotting and ELISA. Tube formation and three-dimensioned sprouting assays in vitro, and orthotopic Xenografts in vivo were conducted as angiogenesis experiments. The mechanism of miR-205/YAP1-mediated tumor angiogenesis was analyzed via overexpression and shRNA, siRNA, or antibody neutralization experiments in combination with anti-VEGF antibody or Axitinib. Results: miR-205/YAP1 signaling axis activates breast normal fibroblasts (NFs) into CAFs, promotes tubule formation and sprouting of Human Umbilical Vein Endothelial Cells (HUVECs). Rescue of miR-205 in CAFs blunts angiogenesis processes. YAP1, a target of miR-205, does not regulate VEGF expression but specifically enhances IL11 and IL15 expressions, maintaining tumor angiogenesis even in the presence of Axitinib or after exhaustion of VEGF by neutralizing VEGF antibody. IL11 and IL15 released from CAFs activate STAT3 signaling in HUVECs. Blockage of IL11 and IL15 expression in CAFs results in the inactivation of STAT3-signaling in HUVECs and repression of the CAF-induced angiogenesis. The blunt angiogenesis halts the invasion and metastasis of breast cancer cells in vivo. Conclusions: These results provide a novel insight into breast CAF-induced tumor angiogenesis in a VEGF-independent manner.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Neoplasias de la Mama/metabolismo , MicroARNs/metabolismo , Fosfoproteínas/metabolismo , Factor de Transcripción STAT3/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Neoplasias de la Mama/genética , Femenino , Fibroblastos/metabolismo , Humanos , MicroARNs/genética , Neovascularización Patológica/metabolismo , Fosfoproteínas/genética , Transducción de Señal/genética , Transducción de Señal/fisiología , Factores de Transcripción , Factor A de Crecimiento Endotelial Vascular/metabolismo , Proteínas Señalizadoras YAP
6.
Int J Oncol ; 51(4): 1191-1198, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28902352

RESUMEN

Tamoxifen resistance is a major clinical challenge in breast cancer treatment. Our previous studies find that GPER and its down-stream signaling play a pivotal role in the development of tamoxifen (TAM) resistance. cDNA array analysis indicated a set of genes associated with cell apoptosis are aberrant in GPER activated and TAM-resistant MCF-7R cells compared with TAM-sensitive MCF-7 cells. Among these genes, Bim (also named BCL2-L11), a member of the BH3-only pro-apoptotic protein family is significantly decreased, and TRIM RING finger protein TRIM2 (a ubiquitin ligase) is highly expressed in MCF-7R. To understand the mechanism of TAM-resistance in GPER activated ER+ breast cancer, the function of TRIM2 and Bim inducing cell apoptosis was studied. By using immunohistochemical and western blot analysis, there is an adverse correlation between TRIM2 and Bim in TAM-resistant breast tumor tissues and MCF-7R cells. Knockdown Bim in TAM-sensitive MCF-7 cells or overexpression of Bim in TAM-resistant MCF-7 cells significantly changed its sensibility to TAM through altering the levels of cleaved PARP and caspase-3. Activation of GPER and its downstream signaling MAPK/ERK, not PI3K/AKT, led to enhanced TRIM2 protein levels and affected the binding between TRIM2 and Bim which resulted in a reduced Bim in TAM-resistant breast cancer cells. Thus, the present study provides a novel insight to TAM-resistance in ER-positive breast cancer cells.


Asunto(s)
Proteína 11 Similar a Bcl2/genética , Neoplasias de la Mama/metabolismo , Resistencia a Medicamentos , Proteínas Nucleares/genética , Receptores de Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Apoptosis , Neoplasias de la Mama/genética , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Sistema de Señalización de MAP Quinasas , Células MCF-7 , Proteínas Nucleares/metabolismo , Receptores de Estrógenos/genética , Receptores Acoplados a Proteínas G/genética , Tamoxifeno/farmacología
7.
Cell Death Dis ; 8(3): e2642, 2017 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-28252644

RESUMEN

Drosha is an RNA III-like enzyme that has an aberrant expression in some tumors. Our previous studies showed the aberrant Drosha in gastric tumors. However, the roles of nuclear Drosha, the main regulator of microRNA (miRNA) biogenesis, in gastric cancer (GC) progression remain poorly understood. In this study, we demonstrated that nuclear Drosha is significantly associated with cell invasion of GC and that Drosha silence impedes the tumor invasion. Knockdown of Drosha led to a set of dysregulated miRNAs in GC cells. Multiple targets of these miRNAs were the members in cell migration, invasion and metastasis-associated signaling (e.g. ECM-receptor interaction, focal adhesion, p53 signaling and MAPK signaling pathway) revealed by bioinformatics analysis. LAMC2 (a key element of ECM-receptor signaling) and CD82 (a suppressor of p53 signaling) are the targets of miR-622 and miR-197, respectively. High levels of LAMC2 and low levels of CD82 were significantly related to the worse outcome for GC patients. Furthermore, overexpression of LAMC2 and knockdown of CD82 markedly promoted GC cell invasion and activated EGFR/ERK1/2-MMP7 signaling via upregulation of the expression of phosphorylated (p)-EGFR, p-ERK1/2 and MMP7. Our findings suggest that nuclear Drosha potentially has a role in the development of GC.


Asunto(s)
Receptores ErbB/metabolismo , Proteína Kangai-1/metabolismo , Laminina/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Metaloproteinasa 7 de la Matriz/metabolismo , MicroARNs/metabolismo , Ribonucleasa III/metabolismo , Neoplasias Gástricas/metabolismo , Línea Celular Tumoral , Movimiento Celular/fisiología , Regulación Neoplásica de la Expresión Génica/fisiología , Humanos , Invasividad Neoplásica/patología , Metástasis de la Neoplasia/patología , Fosforilación/fisiología , Transducción de Señal/fisiología , Neoplasias Gástricas/patología , Regulación hacia Arriba/fisiología
8.
Steroids ; 112: 1-11, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27016131

RESUMEN

Cancer associated fibroblasts (CAFs) are crucial contributors to breast cancer development. Estrogen affects mammary stroma in both physiological and pathophysiological conditions. We show here that estrogen (G-protein coupled) receptor (GPER) could be detected by immunohistochemistry in stromal fibroblasts of primary breast cancers. The presence of GPER expression was further confirmed by immunofluorescence and quantitative PCR in CAFs isolated from primary breast cancers. Based on dynamic monitoring by real time cell analyzer (RTCA) system, 17-ß-estradiol (E2) as well as GPER specific agonist G1 were observed to trigger transient cell index increasing within an hour in a dosage-dependent manner in breast CAFs. In addition, E2 and G1 stimulated intracellular calcium modulation and phosphorylation of extracellular signal-regulated kinase (ERK) 1/2 within seconds and minutes in CAFs, respectively. Moreover, E2 and G1 promoted cell proliferation of breast CAFs measured by RTCA monitoring, cell viability assay and cell cycle analysis, and this promotion could be blocked by a GPER-selective antagonist G15. Interestingly, dynamic RTCA monitoring indicated that E2 increased adhesion of resuspended cells, and microscopy confirmed that E2 stimulated cell spreading. Both the adhesion and spreading were proposed to be mediated by GPER, since G1 also stimulated these effects similar to E2, and G15 reduced them. Moreover, GPER was found to mediate migration that was increased by E2 and G1 but reduced by G15 in RTCA cell migration assay and transwell assay. Accordingly, GPER mediates not only rapid actions but also slow effects including adhesion/spreading, proliferation and migration in breast CAFs. Estrogen is likely to affect tumor associated stroma and contributes to mammary carcinoma development through CAFs.


Asunto(s)
Neoplasias de la Mama/metabolismo , Fibroblastos Asociados al Cáncer/metabolismo , Estrógenos/farmacología , Receptores Acoplados a Proteínas G/metabolismo , Benzodioxoles/farmacología , Calcio/metabolismo , Adhesión Celular/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Estradiol/farmacología , Femenino , Humanos , Immunoblotting , Quinolinas/farmacología , Receptores de Estrógenos/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
9.
Stem Cells ; 34(1): 55-66, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26418365

RESUMEN

Cancer stem cells (CSCs) are a subpopulation of neoplastic cells with self-renewal capacity and limitless proliferative potential as well as high invasion and migration capacity. These cells are commonly associated with epithelial-mesenchymal transition (EMT), which is also critical for tumor metastasis. Recent studies illustrate a direct link between EMT and stemness of cancer cells. Long non-coding RNAs (lncRNAs) have emerged as important new players in the regulation of multiple cellular processes in various diseases. To date, the role of lncRNAs in EMT-associated CSC stemness acquisition and maintenance remains unclear. In this study, we discovered that a set of lncRNAs were dysregulated in Twist-positive mammosphere cells using lncRNA microarray analysis. Multiple lncRNAs-associated canonical signaling pathways were identified via bioinformatics analysis. Especially, the Shh-GLI1 pathway associated lncRNA-Hh, transcriptionally regulated by Twist, directly targets GAS1 to stimulate the activation of hedgehog signaling (Hh). The activated Hh increases GLI1 expression, and enhances the expression of SOX2 and OCT4 to play a regulatory role in CSC maintenance. Thus, the mammosphere-formation efficiency (MFE) and the self-renewal capacity in vitro, and oncogenicity in vivo in Twist-positive breast cancer cells are elevated. lncRNA-Hh silence in Twist-positive breast cells attenuates the activated Shh-GLI1 signaling and decreases the CSC-associated SOX and OCT4 levels, thus reduces the MFE and tumorigenesis of transplanted tumor. Our results reveal that lncRNAs function as an important regulator endowing Twist-induced EMT cells to gain the CSC-like stemness properties.


Asunto(s)
Neoplasias de la Mama/metabolismo , Proteínas Hedgehog/metabolismo , Células Madre Neoplásicas/metabolismo , Proteínas Nucleares/metabolismo , ARN Largo no Codificante/metabolismo , Transducción de Señal , Proteína 1 Relacionada con Twist/metabolismo , Animales , Neoplasias de la Mama/patología , Carcinogénesis/patología , Línea Celular Tumoral , Transición Epitelial-Mesenquimal/genética , Femenino , Humanos , Ratones Endogámicos BALB C , Ratones Desnudos , Células Madre Neoplásicas/patología , ARN Largo no Codificante/genética , Esferoides Celulares/patología
10.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 31(9): 1189-93, 1199, 2015 Sep.
Artículo en Chino | MEDLINE | ID: mdl-26359098

RESUMEN

OBJECTIVE: To construct a lentiviral vector (Lenti-GPER-shRNA) targeting G-protein coupled estrogen receptor (GPER) and explore the role of GPER in the effect of tamoxifen on cell proliferation and apoptosis in breast cancer associated fibroblasts (BCAFs). METHODS: The target sequence of GPER gene and negative control were cloned into lentiviral vectors. The recombinant lentivirus and control were extracted after HEK293T cells were transfected with the recombinant vector and helper vectors. After infection of BCAFs with the GPER lentiviral vector under the best interfering condition, GPER expression was detected by real-time quantitative PCR and Western blotting. BCAFs were divided into negative control group, GPER-RNAi group, negative control combined with tamoxifen (10(-8) mmol/L) group and GPER-RNAi combined with tamoxifen (10(-8) mmol/L) group. CCK-8 assay was used to detect the proliferation and annexin V-fluorescein isothiocyanate/propidium iodide (annexin V-FITC/PI) combined with flow cytometry was used to detect the apoptosis of BCAFs after the treatment of tamoxifen. RESULTS: Lenti-GPER-shRNA significantly interfered the expression of GPER in BCAFs. Tamoxifen promoted the growth of BCAFs, which could be attenuated by knockdown of GPER. Moreover, the apoptosis of BCAFs was reduced by tamoxifen, which was also reversed by knockdown of GPER. CONCLUSION: Lenti-GPER-shRNA could effectively silence the GPER expression in BCAFs. The ability of tamoxifen to accelerate cell proliferation and decrease cell apoptosis could be weakened by knockdown of GPER.


Asunto(s)
Apoptosis/efectos de los fármacos , Neoplasias de la Mama/patología , Antagonistas de Estrógenos/farmacología , Receptores de Estrógenos/fisiología , Receptores Acoplados a Proteínas G/fisiología , Tamoxifeno/farmacología , Proliferación Celular/efectos de los fármacos , Femenino , Fibroblastos/fisiología , Células HEK293 , Humanos , Transfección
11.
Breast Cancer Res ; 17: 69, 2015 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-25990368

RESUMEN

INTRODUCTION: Acquired tamoxifen resistance remains the major obstacle to breast cancer endocrine therapy. ß1-integrin was identified as one of the target genes of G protein-coupled estrogen receptor (GPER), a novel estrogen receptor recognized as an initiator of tamoxifen resistance. Here, we investigated the role of ß1-integrin in GPER-mediated tamoxifen resistance in breast cancer. METHODS: The expression of ß1-integrin and biomarkers of epithelial-mesenchymal transition were evaluated immunohistochemically in 53 specimens of metastases and paired primary tumors. The function of ß1-integrin was investigated in tamoxifen-resistant (MCF-7R) subclones, derived from parental MCF-7 cells, and MCF-7R ß1-integrin-silenced subclones in MTT and Transwell assays. Involved signaling pathways were identified using specific inhibitors and Western blotting analysis. RESULTS: GPER, ß1-integrin and mesenchymal biomarkers (vimentin and fibronectin) expression in metastases increased compared to the corresponding primary tumors; a close expression pattern of ß1-integrin and GPER were in metastases. Increased ß1-integrin expression was also confirmed in MCF-7R cells compared with MCF-7 cells. This upregulation of ß1-integrin was induced by agonists of GPER and blocked by both antagonist and knockdown of it in MCF-7R cells. Moreover, the epidermal growth factor receptor/extracellular regulated protein kinase (EGFR/ERK) signaling pathway was involved in this transcriptional regulation since specific inhibitors of these kinases also reduced the GPER-induced upregulation of ß1-integrin. Interestingly, silencing of ß1-integrin partially rescued the sensitivity of MCF-7R cells to tamoxifen and the α5ß1-integrin subunit is probably responsible for this phenomenon. Importantly, the cell migration and epithelial-mesenchymal transition induced by cancer-associated fibroblasts, or the product of cancer-associated fibroblasts, fibronectin, were reduced by knockdown of ß1-integrin in MCF-7R cells. In addition, the downstream kinases of ß1-integrin including focal adhesion kinase, Src and AKT were activated in MCF-7R cells and may be involved in the interaction between cancer cells and cancer-associated fibroblasts. CONCLUSIONS: GPER/EGFR/ERK signaling upregulates ß1-integrin expression and activates downstream kinases, which contributes to cancer-associated fibroblast-induced cell migration and epithelial-mesenchymal transition, in MCF-7R cells. GPER probably contributes to tamoxifen resistance via interaction with the tumor microenvironment in a ß1-integrin-dependent pattern. Thus, ß1-integrin may be a potential target to improve anti-hormone therapy responses in breast cancer patients.


Asunto(s)
Antineoplásicos Hormonales/farmacología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Resistencia a Antineoplásicos , Transición Epitelial-Mesenquimal , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Tamoxifeno/farmacología , Antineoplásicos Hormonales/uso terapéutico , Biomarcadores , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Línea Celular Tumoral , Resistencia a Antineoplásicos/genética , Transición Epitelial-Mesenquimal/genética , Receptores ErbB/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibronectinas/genética , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Humanos , Inmunohistoquímica , Integrina beta1/metabolismo , Células MCF-7 , Receptores de Estrógenos/metabolismo , Moduladores Selectivos de los Receptores de Estrógeno/uso terapéutico , Transducción de Señal/efectos de los fármacos , Tamoxifeno/uso terapéutico , Familia-src Quinasas/metabolismo
12.
Endocr Relat Cancer ; 21(2): 355-69, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24481325

RESUMEN

Cancer-associated fibroblasts (CAFs) are crucial co-mediators of breast cancer progression. Estrogen is the predominant driving force in the cyclic regulation of the mammary extracellular matrix, thus potentially affecting the tumor-associated stroma. Recently, a third estrogen receptor, estrogen (G-protein-coupled) receptor (GPER), has been reported to be expressed in breast CAFs. In this study, GPER was detected by immunohistochemical analysis in stromal fibroblasts of 41.8% (59/141) of the primary breast cancer samples. GPER expression in CAFs isolated from primary breast cancer tissues was confirmed by immunostaining and RT-PCR analyses. Tamoxifen (TAM) in addition to 17ß-estradiol (E2) and the GPER agonist G1 activated GPER, resulting in transient increases in cell index, intracellular calcium, and ERK1/2 phosphorylation. Furthermore, TAM, E2, and G1 promoted CAF proliferation and cell-cycle progression, both of which were blocked by GPER interference, the selective GPER antagonist G15, the epidermal growth factor receptor (EGFR) inhibitor AG1478, and the ERK1/2 inhibitor U0126. Importantly, TAM as well as G1 increased E2 production in breast CAFs via GPER/EGFR/ERK signaling when the substrate of E2, testosterone, was added to the medium. GPER-induced aromatase upregulation was probably responsible for this phenomenon, as TAM- and G1-induced CYP19A1 gene expression was reduced by GPER knockdown and G15, AG1478, and U0126 administration. Accordingly, GPER-mediated CAF-dependent estrogenic effects on the tumor-associated stroma are conceivable, and CAF is likely to contribute to breast cancer progression, especially TAM resistance, via a positive feedback loop involving GPER/EGFR/ERK signaling and E2 production.


Asunto(s)
Neoplasias de la Mama/metabolismo , Proliferación Celular , Fibroblastos/metabolismo , Receptores de Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Ciclopentanos/farmacología , Estradiol/farmacología , Femenino , Fibroblastos/efectos de los fármacos , Humanos , Quinolinas/farmacología , Receptores de Estrógenos/agonistas , Receptores Acoplados a Proteínas G/agonistas , Tamoxifeno/farmacología
13.
Breast Cancer Res ; 15(6): R114, 2013 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-24289103

RESUMEN

INTRODUCTION: Tamoxifen is widely used to treat hormone-dependent breast cancer, but its therapeutic benefit is limited by the development of drug resistance. Here, we investigated the role of estrogen G-protein coupled receptor 30 (GPR30) on Tamoxifen resistance in breast cancer. METHODS: Primary tumors (PTs) of breast cancer and corresponding metastases (MTs) were used to evaluate the expression of GPR30 and epidermal growth factor receptor (EGFR) immunohistochemically. Tamoxifen-resistant (TAM-R) subclones derived from parent MCF-7 cells were used to investigate the role of GPR30 in the development of tamoxifen resistance, using MTT assay, western blot, RT-PCR, immunofluorescence, ELISA and flow cytometry. TAM-R xenografts were established to assess anti-tumor effects of combination therapy with GPR30 antagonist G15 plus 4-hydroxytamoxifen (Tam), using tumor volume measurement and Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL). RESULTS: In 53 human breast cancer specimens, GPR30 expression in MTs increased compared to matched PTs; in MTs, the expression patterns of GPR30 and EGFR were closely related. Compared to parent MCF-7 cells, TAM-R cells had greater growth responses to 17ß-estradiol (E2), GPR30 agonist G1 and Tam, and significantly higher activation of Mitogen-activated protein (MAP) kinases; but this increased activity was abolished by G15 or AG1478. In TAM-R cells, GPR30 cell-surface translocation facilitated crosstalk with EGFR, and reduced cAMP generation, attenuating inhibition of EGFR signaling. Combination therapy both promoted apoptosis in TAM-R cells and decreased drug-resistant tumor progression. CONCLUSIONS: Long-term endocrine treatment facilitates the translocation of GPR30 to cell surfaces, which interferes with the EGFR signaling pathway; GPR30 also attenuates the inhibition of MAP kinases. These factors contribute to tamoxifen resistance development in breast cancer. Combination therapy with GPR30 inhibitors and tamoxifen may provide a new therapeutic option for drug-resistant breast cancer.


Asunto(s)
Antineoplásicos Hormonales/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Resistencia a Antineoplásicos/efectos de los fármacos , Receptores de Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Tamoxifeno/farmacología , Adulto , Anciano , Anciano de 80 o más Años , Animales , Benzodioxoles/farmacología , Neoplasias de la Mama/patología , AMP Cíclico/metabolismo , Receptores ErbB/metabolismo , Estradiol/farmacología , Femenino , Humanos , Células MCF-7 , Ratones Desnudos , Persona de Mediana Edad , Neoplasias Hormono-Dependientes/tratamiento farmacológico , Neoplasias Hormono-Dependientes/metabolismo , Neoplasias Hormono-Dependientes/patología , Quinolinas/farmacología , Receptores de Estrógenos/antagonistas & inhibidores , Receptores de Estrógenos/genética , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/genética , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Mol Oncol ; 7(6): 1116-28, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24011664

RESUMEN

Aberrant expression of c-Ski oncoprotein in some tumor cells has been shown to be associated with cancer development. However, the role of c-Ski in cancer-associated fibroblasts (CAFs) of tumor microenvironment has not been characterized. In the current study, we found that c-Ski is highly expressed in CAFs derived from breast carcinoma microenvironment and this CAF-associated c-Ski expression is associated with invasion and metastasis of human breast tumors. We showed that c-Ski overexpression in immortalized breast normal fibroblasts (NFs) induces conversion to breast CAFs by repressing p53 and thereby upregulating SDF-1 in NFs. SDF-1 treatment or p53 knockdown in NFs had similar effects on the activation of NFs as c-Ski overexpression. The c-Ski-activated CAFs show increased proliferation, migration, invasion and contraction compared with NFs. Furthermore, c-Ski-activated CAFs facilitated the migration and invasion of MDA-MB-231 breast cancer cells. Our data suggest that c-Ski is an important regulator in the activation of CAFs and may serve as a potential therapeutic target to block breast cancer progression.


Asunto(s)
Neoplasias de la Mama/metabolismo , Movimiento Celular , Proteínas de Unión al ADN/metabolismo , Fibroblastos/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Microambiente Tumoral , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Neoplasias de la Mama/terapia , Línea Celular Transformada , Línea Celular Tumoral , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Proteínas de Unión al ADN/genética , Femenino , Fibroblastos/patología , Humanos , Invasividad Neoplásica , Proteínas Proto-Oncogénicas/genética
15.
Nan Fang Yi Ke Da Xue Xue Bao ; 33(4): 618-insidebackcover, 2013 Apr.
Artículo en Chino | MEDLINE | ID: mdl-23644134

RESUMEN

OBJECTIVE: glucagonoma is a rare islet alpha-cell tumor. We report a case of glucagonoma in a 55-year-old male patient with such clinical findings of necrolytic migratory erythema, diabetes mellitus, body weight loss, and anemia. CT examination found a space-occupying lesion in the pancreas, and an elevated serum glucagon level indicate the diagnosis of glucagonoma, which was confirmed postoperatively by pathological examination of the tumor tissue. A definite diagnosis of glucagonoma relies on pathological report, and so far no standard treatment strategy has been available for this tumor. Surgical resection is an effective means for treatment of glucagonoma.


Asunto(s)
Glucagonoma , Neoplasias Pancreáticas , Glucagonoma/diagnóstico , Glucagonoma/patología , Glucagonoma/cirugía , Humanos , Masculino , Persona de Mediana Edad , Pancreatectomía , Neoplasias Pancreáticas/diagnóstico , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/cirugía
16.
Mol Clin Oncol ; 1(4): 745-748, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24649239

RESUMEN

Intra-arterial infusion chemotherapy for locally advanced breast cancer (LABC) has been previously performed. However, the main complications of this type of chemotherapy remain to be clarified. In the present study, catheterization chemotherapy was carried out for 53 LABC cases (stage IIIa-IIIc) between May, 2006 and March, 2007. For IIIB and IIIC patients, the catheters were guided to the opening of the subclavian artery. For stage IIIa patients, the catheters were placed into the thoracic artery through a subcutaneous femoral artery puncture. One to four cycles of chemotherapy (mean, 1.6 cycles) were administered for the patients using taxotere, epidoxorubicin, 5-fluorouracil and/or cyclophosphamide. The interval time between the two cycles was 21 days. Seven cases were identified as complete response (CR, 13.2%), 41 cases were partial response (PR, 77.4%) with a rate of effectiveness of (CR + PR, 90.6%), 5 cases were stable disease (SD, 9.40%) and no case was progressive. Pain of the ipsilateral upper extremity was present in 7 cases. Two cases exhibited ipsilateral upper extremity atrophy following drug administration from the opening of the subclavian artery. One case experienced neck pain and headache, while in one case necrosis of local skin was evident. Hematological toxicity over grade 3 was observed in 6 cases (11.30%). Systemic toxicity was mild and did not affect the quality of life of the patients. Overall survival was identified as 18/51 (35.3%), and free-disease survival as 10/51 (19.6%). In conclusion, intra-arterial infusion chemotherapy is an effective local control treatment for LABC. The main complications are pain of the ipsilateral upper extremity and neck as well as headache. Severe complications are ipsilateral upper extremity atrophy and necrosis of local skin. During the treatment, controlling the pressure of the tourniquet and velocity of drug administration are crucial for reducing local complications.

17.
J Breast Cancer ; 14(3): 185-90, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22031799

RESUMEN

PURPOSE: G-protein coupled estrogen receptor 1 (GPER) probably play important roles in the progression of breast cancer including endocrine therapeutic resistance. We evaluated GPER in primary breast cancers. METHODS: Immunohistochemistry was used to detect GPER in paraffin-embedded tissues of primary breast cancers from 423 patients and GPER expression was correlated with clinicopathological factors. RESULTS: GPER was expressed in 63.8% of specimens, coexpressed with estrogen receptor alpha (ERα) in 36.6% of tumors and was positive in 62.5% of the ERα-negative tumors. The expression of GPER had no relationship with the status of ERα, progesterone receptor and HER2. Although the expression of GPER was significantly inversely related with nodal status (p=0.045), no correlation between GPER expression and other clinicopathological variables (age, menstruation status, tumor size, stage, histologic grade, Nottingham Prognostic Index or pathological type) was found. CONCLUSION: GPER and ERα exhibited independent expression pattern of distribution in primary breast cancers. A long-term follow-up and a more definite molecular phenotype for ER are necessary in confirming studies.

18.
Breast J ; 17(5): 490-7, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21762243

RESUMEN

The stereotactic or ultrasound-guided vacuum-assisted breast biopsy (Mammotome, MMT) system is a minimally invasive surgical technique. Increasingly, it is used to remove benign breast lesions as management. To evaluate the therapeutic value of 8-gauge ultrasound-guided MMT system (UMS) in presumed benign breast lesions, a retrospective analysis was performed on a series of 2,167 consecutive 8-gauge UMS procedures. The parameters used in this analysis included lesion size, location, breast imaging reporting and data system for ultrasound category, histopathologic diagnosis, and others. A total of 1,119 women whose mean ages were 36.6 years (range: 12-71, SD: 9.6) underwent 2,167 consecutive 8-gauge UMS procedures. Among the patients, 298 cases (26.63%) did not have palpable mass, 430 (38.43%) had multiple lesions, and 237 (21.18%) had bilateral ones. The average size of excised lesions was 15.8 mm (range: 5-55, SD: 6.7) in the largest dimension, including 294 lesions < 10 mm (13.57%); 1,359 lesions, 10-19 mm (62.71%); 420 lesions, 20-29 mm (19.38%), and 94 lesions ≥ 30 mm (4.34%). Predominant lesions (81.59%) were solid on ultrasound image and nearly half (48.59%) of them were localized in the upper outer quadrant. Histopathologic diagnosis revealed that the overwhelming majority of specimens (96.61%) were benign, most of which manifested as fibroadenoma and fibrocystic changes, while high-risk lesions were revealed in 31 (2.29%) cases and malignancies in 15 (1.11%). Average time for procedure was 8.6 minutes (range: 3.5-38, SD: 5.4) and mean number of cores removed in the procedure was 9.3 (range: 2-42, SD: 3.7). Complete excision was achieved predominantly (99.82%). Complications (59, 5.27%) in which hematoma (41, 3.66%) was the majority were acceptable. In conclusion, the 8-gauge UMS procedure is a safe and potent therapeutic management with satisfactory cosmetic outcome for benign and high-risk breast lesions, especially for bilateral, multiple, and nonpalpable ones. It is a reliable biopsy tool for suspected lesions as well. Eradication and maximal cosmesis can be achieved with few complications if the targeted lesion is limited to 30 mm.


Asunto(s)
Neoplasias de la Mama/patología , Neoplasias de la Mama/cirugía , Carcinoma Ductal de Mama/patología , Carcinoma Ductal de Mama/cirugía , Fibroadenoma/patología , Fibroadenoma/cirugía , Ultrasonografía Intervencional/métodos , Adolescente , Adulto , Anciano , Análisis de Varianza , Neoplasias de la Mama/diagnóstico por imagen , Carcinoma Ductal de Mama/diagnóstico por imagen , Niño , Equimosis/etiología , Femenino , Fibroadenoma/diagnóstico por imagen , Enfermedad Fibroquística de la Mama/patología , Hematoma/etiología , Humanos , Persona de Mediana Edad , Agujas , Hemorragia Posoperatoria/etiología , Estudios Retrospectivos , Ultrasonografía Intervencional/efectos adversos , Ultrasonografía Intervencional/instrumentación , Vacio , Adulto Joven
19.
J Huazhong Univ Sci Technolog Med Sci ; 29(2): 202-6, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19399405

RESUMEN

In this study, the effects of pirrolidine dithiocarbamate (PDTC) plus leflunomide (Lef) and cyclosporine (CsA) on the NF-kappaB signaling pathway in mouse-to-rat cardiac xeno-transplantation models were investigated. NIH mice and Wistar rats served as donors and recipients respectively. Mouse-to-rat cardiac xenotransplantation was performed. The recipients were divided into 5 groups: group A (the control group), group B (PDTC group), group C (PDTC plus CsA group), group D (PDTC plus Lef group) and group E (PDTC plus Lef and CsA group). The expressions of IKKalpha/beta, NF-kappaB-P65, IkappaBalpha, ICAM-1 and NF-kappaB DNA binding activity in xenograft tissues were determined by immunohistochemistry and Western blot as well as electrophoretic mobility shift assay (EMSA). The median survival time of cardiac xenografts in the control group, PDTC group, PDTC plus CsA group, PDTC plus Lef group and PDTC plus Lef and CsA group was (2.17+/-0.41), (2.33+/-0.52), (4.67+/-1.21), (7.00+/-1.79) and (9.00+/-1.41) days respectively. The survival time of xenografts in the PDTC plus Lef and CsA group was significantly longer than that in other four groups (P<0.05 for all), that in the PDTC plus Lef group longer than that in the control group, PDTC group and PDTC plus CsA group (P<0.05 for all), that in PDTC plus CsA group longer than the control group and PDTC group (P<0.05 for all). The expressions of IKKalpha/beta, NF-kappaB-P65, IkappaBalpha and ICAM-1 and NF-kappaB DNA binding activity were notably increased in mouse-to-rat cardiac xenografts. The expressions were decreased in the control group, PDTC group, PDTC plus CsA group, PDTC plus Lef and PDTC plus Lef and CsA group in turn. It was concluded that PDTC plus Lef and CsA can significantly suppress the expressions of IKKalpha/beta, NF-kappaB-P65, IkappaBalpha, ICAM-1 and NF-kappaB DNA binding activity, thereby prolonging the survival of the cardiac xenografts.


Asunto(s)
Ciclosporina/farmacología , Supervivencia de Injerto/efectos de los fármacos , Trasplante de Corazón , Isoxazoles/farmacología , Pirrolidinas/farmacología , Tiocarbamatos/farmacología , Animales , Ciclosporina/uso terapéutico , Inmunosupresores/farmacología , Inmunosupresores/uso terapéutico , Isoxazoles/uso terapéutico , Leflunamida , Masculino , Ratones , FN-kappa B/metabolismo , Pirrolidinas/uso terapéutico , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Tiocarbamatos/uso terapéutico , Trasplante Heterólogo
20.
Artículo en Chino | WPRIM (Pacífico Occidental) | ID: wpr-301347

RESUMEN

In this study,the effects of pirrolidine dithiocarbamate (PDTC) plus leflunomide (Lef) models were investigated.NIH mice and Wistar rats served as donors and recipients respectively.Mouse-to-rat cardiac xenotransplantation was performed.The recipients were divided into 5 groups:group A (the control group),group B (PDTC group),group C (PDTC plus CsA group),group D (PDTC plus Lef group) and group E (PDTC plus Lef and CsA group).The expressions of IKKα/β,by immunohistochemistry and Western blot as well as electrophoretic mobility shift assay (EMSA).The median survival time of cardiac xenografts in the control group,PDTC group,PDTC plus CsA group,PDTC plus Lef group and PDTC plus Lef and CsA group was (2.17±0.41),(2.33±0.52),(4.67±1.21),(7.00±1.79) and (9.00±1.41) days respectively.The survival time of xenografts in the PDTC plus Lef and CsA group was significantly longer than that in other four groups (P<0.05 for all),that in the PDTC plus Lef group longer than that in the control group,PDTC group and PDTC plus CsA group (P<0.05 for all),that in PDTC plus CsA group longer than the control group and PDTC binding activity were notably increased in mouse-to-rat cardiac xenografts.The expressions were decreased in the control group,PDTC group,PDTC plus CsA group,PDTC plus Lef and PDTC plus Lef and CsA group in turn.It was concluded that PDTC plus Lef and CsA can significantly suppress prolonging the survival of the cardiac xenografts.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...