Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Front Oncol ; 14: 1326385, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38800388

RESUMO

Purpose: This study aimed to investigate the factors associated with pathologic node-negativity (ypN0) in patients who received neoadjuvant chemotherapy (NAC) to develop and validate an accurate prediction nomogram. Methods: The CSBrS-012 study (2010-2020) included female patients with primary breast cancer treated with NAC followed by breast and axillary surgery in 20 hospitals across China. In the present study, 7,711 eligible patients were included, comprising 6,428 patients in the primary cohort from 15 hospitals and 1,283 patients in the external validation cohort from five hospitals. The hospitals were randomly assigned. The primary cohort was randomized at a 3:1 ratio and divided into a training set and an internal validation set. Univariate and multivariate logistic regression analyses were performed on the training set, after which a nomogram was constructed and validated both internally and externally. Results: In total, 3,560 patients (46.2%) achieved ypN0, and 1,558 patients (20.3%) achieved pathologic complete response in the breast (bpCR). A nomogram was constructed based on the clinical nodal stage before NAC (cN), ER, PR, HER2, Ki67, NAC treatment cycle, and bpCR, which were independently associated with ypN0. The area under the receiver operating characteristic curve (AUC) for the training set was 0.80. The internal and external validation demonstrated good discrimination, with AUCs of 0.79 and 0.76, respectively. Conclusion: We present a real-world study based on nationwide large-sample data that can be used to effectively screen for ypN0 to provide better advice for the management of residual axillary disease in breast cancer patients undergoing NAC.

2.
Heliyon ; 10(7): e28502, 2024 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-38586399

RESUMO

Objective: To explore risk factors for defective non-union of bone and develop a nomogram-based prediction model for such an outcome. Methods: This retrospective study analysed the case data of patients with defective bony non-unions who were treated at the authors' hospital between January 2010 and December 2020. Patients were divided into the union and non-union groups according to their Radiographic Union Score for Tibia scores 1 year after surgery. Univariate analysis was performed to assess factors related to demographic characteristics, laboratory investigations, surgery, and trauma in both groups. Subsequently, statistically significant factors were included in the multivariate logistic regression analysis to identify independent risk factors. A nomogram-based prediction model was established using statistically significant variables in the multivariate analysis. The accuracy and stability of the model were evaluated using receiver operating characteristic (ROC) and calibration curves. The clinical applicability of the nomogram model was evaluated using decision curve analysis. Results: In total, 204 patients (171 male, 33 female; mean [±SD] age, 39.75 ± 13.00 years) were included. The mean body mass index was 22.95 ± 3.64 kg/m2. Among the included patients, 29 were smokers, 18 were alcohol drinkers, and 21 had a previous comorbid systemic disease (PCSD). Univariate analysis revealed that age, occupation, PCSD, smoking, drinking, interleukin-6, C-reactive protein (CRP), procalcitonin, alkaline phosphatase, glucose, and uric acid levels; blood calcium ion concentration; and bone defect size (BDS) were correlated with defective bone union (all P < 0.05). Multivariate logistic regression analysis revealed that PCSD, smoking, interleukin-6, CRP, and glucose levels; and BDS were associated with defective bone union (all P < 0.05), and the variables in the multivariate analysis were included in the nomogram-based prediction model. The value of the area under the ROC curve for the predictive model for bone defects was 0.95. Conclusion: PCSD, smoking, interleukin-6, CRP, and glucose levels; and BDS were independent risk factors for defective bony non-union, and the incidence of such non-union was predicted using the nomogram. These findings are important for clinical interventions and decision-making.

3.
Cancer Med ; 12(20): 20287-20298, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37795774

RESUMO

BACKGROUND: The efficacy of breast reconstruction for patients with N2-3M0 stage female breast cancer (FBC) remained unclear due to the lack of randomized clinical trials. This retrospective study aimed to explore the efficacy of breast reconstruction for patients with N2-3M0 stage FBC. METHODS: Two thousand five hundred forty-five subjects with FBC staged by N2-3M0 from 2010 to 2016 were retrieved from the Surveillance, Epidemiology, and End Results database. Generalized boosted model (GBM) and propensity score matching (PSM) analyses and multivariable Cox analyses were employed to assess the clinical prognostic effect of postmastectomy reconstruction for patients with N2-3M0 stage FBC in breast cancer-specific survival (BCSS). RESULTS: Totally, 1784 candidates underwent mastectomy alone (mastectomy group), and 761 candidates underwent postmastectomy reconstruction (PMbR group), with 418 breast-specific deaths after a median follow-up time of 57 months (ranging from 7 to 227 months). BCSS in the mastectomy group showed no statistical difference from that in the PMbR group in the PSM cohort (HR = 0.93, 95% CI: 0.70-1.25, p = 0.400) and GBM cohort (HR = 0.75, 95% CI: 0.56-1.01, p = 0.057). In the multivariate analyses, there was no difference in the effect of PMbR and mastectomy on BCSS in the original cohort (HR = 0.85, 95% CI: 0.66-1.09, p = 0.197), PSM cohort (HR = 0.86, 95% CI: 0.64-1.15, p = 0.310), and GBM cohort (HR = 0.84, 95% CI: 0.61-1.17, p = 0.298). Triple-negative breast cancer (TNBC) was a detrimental factor affecting BCSS for patients in the PMbR group. CONCLUSIONS: Our study demonstrated that PMbR is an oncologically safe surgical treatment and can be widely recommended in clinics for females with non-TNBC staged by T0-3N2-3M0.


Assuntos
Neoplasias da Mama , Mamoplastia , Neoplasias de Mama Triplo Negativas , Feminino , Humanos , Mastectomia/métodos , Estudos Retrospectivos , Pontuação de Propensão , Neoplasias de Mama Triplo Negativas/cirurgia
4.
Cancer Med ; 12(9): 10326-10339, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36880192

RESUMO

BACKGROUND: The use of systematic treatment for tubular carcinoma (TC) of the breast remained controversial. This study aimed to explore the efficacy of chemotherapy on TC to develop individualized treatment strategies. METHODS: Using the Surveillance, Epidemiology, and End Results (SEER) database, 6486 eligible cases with TC and 309,304 with invasive ductal carcinoma (IDC) were collected. Breast cancer-specific survival (BCSS) was assessed through multivariable Cox analyses and Kaplan-Meier analyses. Differences between groups were balanced using propensity score matching (PSM) and inverse probability of treatment weighting (IPTW). RESULTS: Compared with IDC patients, TC patients had a more favorable long-term BCSS after PSM (hazard ratio = 0.62, p = 0.004) and IPTW (hazard ratio = 0.61, p < 0.001). Chemotherapy was an unfavorable predictor of BCSS for TC (hazard ratio = 3.20, p < 0.001). After stratifying by hormone receptor (HR) and lymph node (LN) status, chemotherapy was correlated with worse BCSS in the HR+/LN- subgroup (hazard ratio = 6.95, p = 0.001) but showed no impact on BCSS in the HR+/LN+ (hazard ratio = 0.75, p = 0.780) and HR-/LN- (hazard ratio = 7.87, p = 0.150) subgroups. CONCLUSIONS: Tubular carcinoma is a low-grade malignant tumor with favorable clinicopathological features and excellent long-term survival. Adjuvant chemotherapy was not recommended for TC regardless of HR and LN status, while the therapy regimens should be carefully individualized.


Assuntos
Adenocarcinoma , Neoplasias da Mama , Carcinoma Ductal de Mama , Humanos , Feminino , Carcinoma Ductal de Mama/patologia , Estudos de Coortes , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/epidemiologia , Pontuação de Propensão
5.
Ann Transl Med ; 11(1): 14, 2023 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-36760254

RESUMO

Background: Chemoresistance is problematic and its mechanisms are unclear in breast cancer. More predictive markers are urgently required. Methods: GSE32646, GSE34138, and GSE20271 were downloaded from the Gene Expression Omnibus (GEO) database to identify differentially expressed genes (DEGs) between chemosensitive and chemoresistant tumors. LinkedOmics was used to analyze ADAM-like Decysin-1 (ADAMDEC1) expression among patients with different clinical characteristics and detect co-expression genes for functional analysis. Tumor Immune Estimation Resource (TIMER) and an integrated repository portal for tumor-immune system interactions (TISIDB) were used to investigate the association between the target gene and the immune response. Gene Set Cancer Analysis (GSCA) was utilized to explore the related pathways of ADAMDEC1 and evaluate the correlation between the expression of ADAMDEC1 and drug sensitivity. RNA22, miRWalk, and miRmap were used to predict the upstream micro ribonucleic acids (miRNAs) regulating ADAMDEC1 expression, while DIANA-LncBase v2 was applied to predict the upstream long non-coding ribonucleic acids (lncRNAs). Kaplan-Meier curve analysis was applied to determine the survival time. Results: We identified that ADAMDEC1 was upregulated among chemosensitive triple-negative breast cancer (TNBC) tissues in GSE32646, GSE34138, and GSE20271. Higher expression of ADAMDEC1 indicated longer survival in breast cancer. Next, we found that ADAMDEC1 was significantly related to the immune response in breast cancer through functional enrichment analysis and further meta-data validation. Moreover, we recognized that hsa-miR-4534 was the potential upstream miRNA regulating ADAMDEC1 expression and Taurine Up-Regulated 1 (TUG1) was the most likely upstream lncRNA of ADAMDEC1 and hsa-miR-4534. Finally, the correlation between ADAMDEC1 and chemosensitivity was confirmed through drug database analysis. Conclusions: Elevated ADAMDEC1 expression is associated with increased chemosensitivity and better prognosis in breast cancer patients.

6.
Front Surg ; 9: 1009149, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36338630

RESUMO

Background: Due to the loss of prediction of overall survival (OS) for patients with invasive micropapillary carcinoma (IMPC) after breast-conserving surgery (BCS), this study aimed to construct a nomogram for predicting OS in IMPC patients after BCS. Methods: In total, 481 eligible cases staged 0-III IMPC from 2000 to 2016 were retrieved from the SEER database. A nomogram was built based on the variables selected by LASSO regression to predict the 3-year and 5-year probabilities of OS. Results: A total of 336 patients were randomly assigned to the training cohort and 145 cases in the validation cohort. The LASSO regression revealed that six variables (age at diagnosis, AJCC stage, marital status, ER status, PR status, and chemotherapy) were predictive variables of OS, and then a nomogram model and an easy-to-use online tool were constructed. The C-indices 0.771 in the training cohort and 0.715 in the validation cohort suggested the robustness of the model. The AUC values for 3-year and 5-year OS in the training cohort were 0.782, 0.790, and 0.674, and 0.682 in the validation cohort, respectively. Based on the cutoff values of 147.23 and 222.44 scores calculated by X-tile analysis, participants in the low-risk group (≤147.23 scores) had a more favorable OS in comparison with those in the medium (>147.23, but <222.44 scores)- and high-risk groups (≥222.44 scores). Conclusions: By risk stratification, this model is expected to provide a precise and personalized prediction of the cumulative risk and guide treatment decision-making in improving OS strategies for IMPC patients.

7.
Front Oncol ; 11: 750376, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34660313

RESUMO

PURPOSE: To synthesize the dimer of GX1 and identify whether its affinity and targeting are better than those of GX1. To prepare 68Ga-DOTA-KEK-(GX1)2 and to apply it to PET and Cerenkov imaging of gastric cancer. METHODS: 68Ga-DOTA-KEK-(GX1)2 was prepared, and the labeling yield and stability were determined. Its specificity and affinity were verified using an in vitro cell binding assay and competitive inhibition test, cell immunofluorescence, and cell uptake and efflux study. Its tumor-targeting ability was determined by nano PET/CT and Cerenkov imaging, standardized uptake value (SUV), signal-to-background ratio (SBR) quantification, and a biodistribution study in tumor-bearing nude mice. RESULTS: 68Ga-DOTA-KEK-(GX1)2 was successfully prepared, and the labeling yield was more than 97%. It existed stably for 90 min in serum. The binding of 68Ga-DOTA-KEK-(GX1)2 to cocultured HUVECs (Co-HUVECs) was higher than that to human umbilical vein endothelial cells (HUVECs), BGC823 cells, and GES cells. It was also higher than that of 68Ga-DOTA-GX1, indicating that the dimer did improve the specificity and affinity of GX1. The binding of KEK-(GX1)2 to Co-HUVECs was significantly higher than that of GX1. Additionally, the uptake of 68Ga-DOTA-KEK-(GX1)2 by Co-HUVECs was higher than that of 68Ga-DOTA-GX1 and reached a maximum at 60 min. Nano PET/CT and Cerenkov imaging showed that the tumor imaging of the nude mice injected with 68Ga-DOTA-KEK-(GX1)2 was clear, and the SUV and SBR value of the tumor sites were significantly higher than those of the nude mice injected with 68Ga-DOTA-GX1, indicating that the probe had better targeting in vivo. Finally, the biodistribution showed quantitatively that when organs such as the kidney and liver metabolized rapidly, the radioactivity of the tumor site of the nude mice injected with 68Ga-DOTA-KEK-(GX1)2 decreased relatively slowly. At the same time, the percentage of injected dose per gram (%ID/g) of the tumor site was higher than that of other normal organs except the liver and kidney at 60 min, which indicated that the tumor had good absorption of the probe. CONCLUSION: GX1 was modified successfully, and the in vivo and in vitro properties of the GX1 dimer were significantly better than those of GX1. The imaging probe, 68Ga-DOTA-KEK-(GX1)2, was successfully prepared, which provides a candidate probe for PET and Cerenkov diagnosis of gastric cancer.

8.
Eur J Pharm Biopharm ; 154: 144-152, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32682942

RESUMO

PURPOSE: The GX1 peptide (CGNSNPKSC) can specifically bind to TGM2 and possesses the ability to target the blood vessels of gastric cancer. This study intends to develop an integrated dual-functional probe with higher affinity, specificity and targeting and to characterize it in vivo and in vitro. METHODS: The dimer and tetramer of GX1 were prepared using cross-linked PEG and labeled with 99mTc. The best targeting probe [PEG-(GX1)2] was selected by gamma camera imaging in nude mouse models of gastric cancer. 188Re-PEG-(GX1)2 was prepared and characterized through cell binding analysis and competitive inhibition experiments, gamma camera imaging, MTT analysis and flow cytometry, BLI, immunohistochemistry, HE staining and biochemical analysis. RESULTS: PEG-(GX1)2 bound specifically to Co-HUVEC with higher affinity than GX1. 188Re-PEG-(GX1)2 had better ability to target gastric cancer in tumor-bearing nude mice and higher T/H ratios than 188Re-GX1. 188Re-PEG-(GX1)2 inhibited the growth of Co-HUVEC and induced apoptosis, and its effects were more robust than those of 188Re-GX1. BLI showed that 188Re-PEG-(GX1)2 inhibited tumor proliferation in vivo with a stronger effect than 188Re-GX1. Compared with 188Re-GX1, 188Re-PEG-(GX1)2 suppressed tumor angiogenesis and tumor cell proliferation and induced tumor cell apoptosis in vivo. The 188Re-PEG-(GX1)2 group did not cause visible changes in liver and kidney morphology and function in vivo. CONCLUSION: The dimer of GX1 was synthesized by using cross-linked PEG, and then 188Re-PEG-(GX1)2 was prepared. This radiopharmaceutical played both diagnostic and therapeutic functions, and gamma camera imaging could be utilized to detect the distribution of drugs in vivo during treatment. Through a series of experiments in vitro and in vivo, the feasibility of the drug was confirmed, and these results laid the foundation for the subsequent development and application of GX1.


Assuntos
Inibidores da Angiogênese/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Imagem Molecular/métodos , Fragmentos de Peptídeos/metabolismo , Radioisótopos/metabolismo , Rênio/metabolismo , Neoplasias Gástricas/metabolismo , Transglutaminases/metabolismo , Inibidores da Angiogênese/uso terapêutico , Animais , Linhagem Celular Tumoral , Feminino , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Sondas Moleculares/metabolismo , Proteína 2 Glutamina gama-Glutamiltransferase , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/patologia , Carga Tumoral/efeitos dos fármacos , Carga Tumoral/fisiologia
9.
J Exp Clin Cancer Res ; 37(1): 198, 2018 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-30134988

RESUMO

BACKGROUND: Phosphatase and tensin homolog (PTEN) is an important tumor suppressor gene, and its encoded protein has activities of both a protein phosphatase and a lipid phosphatase. However, the substitution effect of protein phosphatase activity remains unclear. PI3K/Akt is the most common pathway negatively regulated by PTEN. The Hippo and PI3K/Akt pathways have a joint effect in regulating cell proliferation and apoptosis. Therefore, how PTEN lipid phosphatase inactivation contributes to the occurrence and development of gastric cancer and the potential role of the Hippo and PI3K/Akt pathways in PTEN lipid phosphatase inactivation mediated gastric tumorigenesis remain to be explored. METHODS: Immunohistochemical staining was performed to detect the expression of p-PTEN and YAP in a gastric cancer tissue microarray. Stable cell lines expressing a wild-type or dominant-negative mutant PTEN were established. The proliferation and migration of stable cells were detected by MTT, BrdU, and colony-formation, transwell assay and high content analysis in vitro, and tumor growth differences were observed in xenograft nude mice. Changes in the expression of key molecules in the Hippo and Akt signaling pathways were detected by western blot. Nuclear-cytoplasm separation, immunofluorescence and coimmunoprecipitation analyses were conducted to explore the dysregulation of Hippo in the stable cell lines. RESULTS: PTEN lipid phosphatase inactivation strongly promoted the proliferation and migration of gastric cancer cells in vitro and tumor growth in vivo. A immunohistochemical analysis of gastric cancer tissues revealed a significant correlation between phosphorylated PTEN and nuclear YAP expression, and both were determined to be independent prognostic factors for gastric cancer. Mechanistically, PTEN lipid phosphatase inactivation abolished the MOB1-LATS1/2 interaction, decreased YAP phosphorylation and finally promoted YAP nuclear translocation, which enhanced the synergistic effect of YAP-TEAD, thus inducing cell proliferation and migration. Moreover, PTEN lipid phosphatase inactivation promoted the PI3K/Akt pathway, and disruption of YAP-TEAD-driven transcription decreased the activation of Akt in a dose-dependent manner. CONCLUSIONS: Taken together, our findings indicate that PTEN lipid phosphatase inactivation links the Hippo and PI3K/Akt pathways to promote gastric tumorigenesis and cancer development.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Carcinogênese/genética , PTEN Fosfo-Hidrolase/genética , Fosfoproteínas/genética , Neoplasias Gástricas/genética , Animais , Apoptose/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Via de Sinalização Hippo , Humanos , Camundongos , Proteína Oncogênica v-akt/genética , Fosfatidilinositol 3-Quinases/genética , Proteínas Serina-Treonina Quinases/genética , Transdução de Sinais/genética , Neoplasias Gástricas/patologia , Fatores de Transcrição , Ensaios Antitumorais Modelo de Xenoenxerto , Proteínas de Sinalização YAP
10.
Cell Death Dis ; 9(6): 579, 2018 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-29785022

RESUMO

The clinical application of GX1, an optimal gastric cancer (GC) targeting peptide, is greatly limited because its receptor in the GC vasculature is unknown. In this study, we screened the candidate receptor of GX1, transglutaminase-2(TGM2), by co-immunoprecipitation (co-IP) combined with mass spectrometry. We found that TGM2 was up-regulated in GC vascular endothelial cells and that GX1 receptor expression was suppressed correspondingly after TGM2 downregulation. A highly consistent co-localization of GX1 receptor and TGM2 was detected at both the cellular and tissue levels. High TGM2 expression was evident in GC tissues from patients with poor prognosis. After TGM2 downregulation, the GX1-mediated inhibition of proliferation and migration and the induction of the apoptosis of GC vascular endothelial cells were weakened or even reversed. Finally, we observed that GX1 could inhibit the GTP-binding activity of TGM2 by reducing its intracellular distribution and downregulating its downstream molecular targets (nuclear factor-kappa B, NF-κB; hypoxia-inducible factor 1-α, HIF1α) in GC vascular endothelial cells. Our study confirms that peptide GX1 can inhibit angiogenesis by directly binding to TGM2, subsequently reducing the GTP-binding activity of TGM2 and thereby suppressing its downstream pathway(NF-κB/HIF1α). Our conclusions suggest that GX1/TGM2 may provide a new target for the diagnosis and treatment of GC.


Assuntos
Células Endoteliais/enzimologia , Proteínas de Ligação ao GTP/metabolismo , Neovascularização Patológica/tratamento farmacológico , Oligopeptídeos/uso terapêutico , Peptídeos Cíclicos/uso terapêutico , Neoplasias Gástricas/irrigação sanguínea , Transglutaminases/metabolismo , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células , Regulação para Baixo/efeitos dos fármacos , Feminino , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células Endoteliais da Veia Umbilical Humana/ultraestrutura , Humanos , Masculino , Pessoa de Meia-Idade , Oligopeptídeos/farmacologia , Peptídeos Cíclicos/farmacologia , Prognóstico , Proteína 2 Glutamina gama-Glutamiltransferase , Neoplasias Gástricas/patologia , Regulação para Cima
11.
Biochem Biophys Res Commun ; 500(4): 924-929, 2018 06 12.
Artigo em Inglês | MEDLINE | ID: mdl-29705704

RESUMO

The transcription factor Forkhead box protein M1 (FOXM1) plays critical roles in cancer development and progression, including human hepatocellular carcinoma (HCC). However, the regulatory role and underlying mechanisms of FOXM1 is still limited. Here, we found that the high level expression of FOXM1 and CCNB1 is closely associated with poor prognosis in HCC patients. And FOXM1 and CCNB1 were overexpressed concomitantly in liver tumor tissues. Knockdown of FOXM1 significantly inhibited the expression levels of CCNB1 in HCC cell lines at both the mRNA and protein levels. Mechanistic studies revealed that FOXM1 binds directly to the promoter region of CCNB1 and regulates the expression levels of the CCNB1 gene in the transcriptional level. Furthermore, the loss of functional and rescue experiments showed that CCNB1 is essential for FOXM1-driven proliferation in HCC cells. In the present study, our results partially explained the dysregulated expression of FOXM1 play an important role in proliferation of human hepatocellular carcinoma cells via transcriptional activation of CCNB1 expression. And it also highlights a FOXM1/CCNB1 axis could be a potential target for the treatment of HCCs.


Assuntos
Carcinoma Hepatocelular/genética , Ciclina B1/genética , Proteína Forkhead Box M1/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Hepáticas/genética , RNA Mensageiro/genética , Sítios de Ligação , Carcinoma Hepatocelular/diagnóstico , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/mortalidade , Linhagem Celular Tumoral , Ciclina B1/antagonistas & inibidores , Ciclina B1/metabolismo , Proteína Forkhead Box M1/metabolismo , Células Hep G2 , Humanos , Neoplasias Hepáticas/diagnóstico , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/mortalidade , Prognóstico , Regiões Promotoras Genéticas , Ligação Proteica , RNA Mensageiro/antagonistas & inibidores , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Análise de Sobrevida , Ativação Transcricional
12.
J Cell Physiol ; 233(2): 1414-1423, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28543431

RESUMO

Existing studies on the mechanism of cell volume regulation are mainly relevant to ion channels and osmosis in extracellular fluid. Recently, accumulating evidence has shown that cellular mechanical microenvironment also influences the cell volume. Herein, we investigated the regulation of substrate stiffness on the cell volume homeostasis of MCF-7 cells and their following migration behaviors. We found that cell volume increases with increasing substrate stiffness, which could be affected by blocking the cell membrane anion permeability and dopamine receptor. In addition, the cell migration is significantly inhibited by decreasing the cell volume using tamoxifen and such inhibition effect on migration is enhanced by increasing substrate stiffness. The cell membrane anion permeability might be the linker between cellular mechanical microenvironment and cellular volume homeostasis regulation. This work revealed the regulation of substrate stiffness on cell volume homeostasis for the first time, which would provide a new perspective into the understanding of cancer metastasis and a promising anti-cancer therapy through regulation of cell volume homeostasis.


Assuntos
Resinas Acrílicas/química , Neoplasias da Mama/metabolismo , Membrana Celular/metabolismo , Tamanho Celular , Osmorregulação , Ácido 4,4'-Di-Isotiocianoestilbeno-2,2'-Dissulfônico/farmacologia , Proteínas de Transporte de Ânions/antagonistas & inibidores , Proteínas de Transporte de Ânions/metabolismo , Antineoplásicos Hormonais/farmacologia , Benzazepinas/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Adesão Celular , Membrana Celular/efeitos dos fármacos , Membrana Celular/patologia , Permeabilidade da Membrana Celular , Movimento Celular , Tamanho Celular/efeitos dos fármacos , Colágeno/metabolismo , Feminino , Humanos , Hidrogéis , Soluções Hipotônicas/farmacologia , Células MCF-7 , Osmorregulação/efeitos dos fármacos , Porosidade , Receptores de Dopamina D1/antagonistas & inibidores , Receptores de Dopamina D1/metabolismo , Tamoxifeno/farmacologia , Microambiente Tumoral
13.
Oncotarget ; 7(18): 25315-27, 2016 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-27015365

RESUMO

Multi-drug resistance (MDR) remains a great obstacle to effective chemotherapy for gastric cancer. A number of secreted glycoproteins have been reported to be involved in the development of MDR in gastric cancer. However, whether glycosylation of secreted glycoproteins changes during MDR of gastric cancer is unclear. Our present work manifested that N-glycosites and site-specific glycoforms of secreted proteins in drug-resistant cell lines were distinctly different from those in the parental cell line for the first time. Further characterization highlighted the significance of some aberrantly glycosylated secretory proteins in MDR, suggesting that manipulating the glycosylation of specific glycoproteins could be a potential target for overcoming multi-drug resistance in gastric cancer.


Assuntos
Resistência a Múltiplos Medicamentos/fisiologia , Resistencia a Medicamentos Antineoplásicos/fisiologia , Glicosilação , Neoplasias Gástricas/metabolismo , Linhagem Celular Tumoral , Glicoproteínas/metabolismo , Humanos , Proteômica
14.
Arch Pathol Lab Med ; 138(7): 910-9, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24978917

RESUMO

CONTEXT: The expression of membrane-bound complement regulatory proteins (mCRPs) that inhibit the complement system in normal tissues is essential for self-protection against an autologous immune reaction. However, the expression patterns of mCRPs, including CD46, CD55, and CD59, are inconsistent in different types of cancer cells. OBJECTIVES: To determine whether CD46, CD55, and CD59 are differentially expressed in neoplastic and adjacent normal colon tissues and to assess their clinical significance. DESIGN: Immunohistochemistry was performed on tissue microarrays of cancerous and adjacent normal colon tissues. RESULTS: The expression levels of CD46, CD55, and CD59 were significantly higher in colon cancer tissues compared with the normal adjacent colon tissues. We found that the expression levels of CD55 and CD59 correlated with the grade of differentiation in colon cancers. In addition, the expression of CD55 and CD59 was greater in stage III and stage IV colon cancers than in stage I and stage II cancers according to staging by the TNM classification. CONCLUSIONS: CD46, CD55, and CD59 are up-regulated in colon cancer. Specifically, CD55 and CD59 are of clinical relevance to differentiation and TNM staging of colon cancer. These data suggest that CD46, CD55, and CD59 have the potential to be used for molecular staging diagnoses and for colon cancer therapies.


Assuntos
Antígenos CD55/metabolismo , Antígenos CD59/metabolismo , Neoplasias do Colo/imunologia , Proteína Cofatora de Membrana/metabolismo , Adenocarcinoma/imunologia , Adenocarcinoma/patologia , Adenocarcinoma Mucinoso/imunologia , Adenocarcinoma Mucinoso/patologia , Adulto , Idoso , Biomarcadores Tumorais/imunologia , Biomarcadores Tumorais/metabolismo , Carcinoma de Células em Anel de Sinete/imunologia , Carcinoma de Células em Anel de Sinete/patologia , Colo/imunologia , Neoplasias do Colo/patologia , Feminino , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Valores de Referência , Análise Serial de Tecidos , Regulação para Cima
15.
BMC Cancer ; 14: 378, 2014 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-24886657

RESUMO

BACKGROUND: FOXO4, a member of the FOXO family of transcription factors, is currently the focus of intense study. Its role and function in gastric cancer have not been fully elucidated. The present study was aimed to investigate the expression profile of FOXO4 in gastric cancer and the effect of FOXO4 on cancer cell growth and metastasis. METHODS: Immunohistochemistry, Western blotting and qRT-PCR were performed to detect the FOXO4 expression in gastric cancer cells and tissues. Cell biological assays, subcutaneous tumorigenicity and tail vein metastatic assay in combination with lentivirus construction were performed to detect the impact of FOXO4 to gastric cancer in proliferation and metastasis in vitro and in vivo. Confocal and qRT-PCR were performed to explore the mechanisms. RESULTS: We found that the expression of FOXO4 was decreased significantly in most gastric cancer tissues and in various human gastric cancer cell lines. Up-regulating FOXO4 inhibited the growth and metastasis of gastric cancer cell lines in vitro and led to dramatic attenuation of tumor growth, and liver and lung metastasis in vivo, whereas down-regulating FOXO4 with specific siRNAs promoted the growth and metastasis of gastric cancer cell lines. Furthermore, we found that up-regulating FOXO4 could induce significant G1 arrest and S phase reduction and down-regulation of the expression of vimentin. CONCLUSION: Our data suggest that loss of FOXO4 expression contributes to gastric cancer growth and metastasis, and it may serve as a potential therapeutic target for gastric cancer.


Assuntos
Proliferação de Células , Pontos de Checagem da Fase G1 do Ciclo Celular/genética , Neoplasias Gástricas/genética , Fatores de Transcrição/biossíntese , Animais , Apoptose/genética , Proteínas de Ciclo Celular , Fatores de Transcrição Forkhead , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Metástase Neoplásica , Neoplasias Gástricas/patologia , Fatores de Transcrição/genética , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Fitoterapia ; 83(6): 1014-22, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22668974

RESUMO

Berberine, a compound isolated from Chinese Goldthread Rhizome, has been widely used as a non-prescription drug to treat diarrhoea in China. Previous studies have demonstrated multiple pharmacological activities for berberine, including its significant role in antimicrobial activity. However, its effect on ion exchange and water transfer remains unclear. The present study aims to explore the effect of berberine on the expression of Na(+)/H(+) exchanger3 (NHE3) and aquaporin4 (AQP4) in both diarrhoea mouse model induced by sennosideA and human intestinal epithelium cell line (HIEC). Semi-quantitative RT-PCR, immunohistochemistry and western blotting were adopted to detect the mRNA and protein expression levels of NHE3 and AQP4. Furthermore, the absorption of berberine and the PKC activity were detected by HPLC and PepTag® Assay to elucidate the underlying mechanisms. It was shown that the expression levels of NHE3 and AQP4 were significantly increased in the diarrhoea mice treated with berberine compared with the untreated diarrhoea mice. Similarly, the expression levels of NHE3 and AQP4 were strikingly enhanced in HIEC co-treated with sennosideA and berberine compared with samples treated with sennosideA only. We also found the maximal absorption of berberine to be approximately 0.01%. In addition, no significant change of PKC activity was observed in the different HIEC treated groups. These results showed that berberine was able to increase the expression of NHE3 and AQP4, suggesting that berberine might exhibit its anti-diarrhoeal effect partially by enhancing the absorption of Na(+) and water.


Assuntos
Aquaporina 4/metabolismo , Berberina/uso terapêutico , Coptis/química , Diarreia/tratamento farmacológico , Medicamentos de Ervas Chinesas/uso terapêutico , Fitoterapia , Trocadores de Sódio-Hidrogênio/metabolismo , Animais , Antidiarreicos/farmacologia , Antidiarreicos/uso terapêutico , Aquaporina 4/genética , Berberina/farmacocinética , Berberina/farmacologia , Linhagem Celular , Diarreia/induzido quimicamente , Diarreia/metabolismo , Medicamentos de Ervas Chinesas/farmacologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Feminino , Humanos , Absorção Intestinal/efeitos dos fármacos , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Proteína Quinase C/metabolismo , RNA Mensageiro/metabolismo , Rizoma , Extrato de Senna , Senosídeos , Sódio/metabolismo , Trocador 3 de Sódio-Hidrogênio , Trocadores de Sódio-Hidrogênio/genética , Água/metabolismo
17.
Carcinogenesis ; 32(12): 1798-805, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21934092

RESUMO

MicroRNAs (miRNAs) regulate tumor progression and invasion via direct interaction with target messenger RNAs (mRNAs). We defined miRNAs involved in cancer metastasis (metastamirs) using an established in vitro colorectal cancer (CRC) model of minimally metastatic cells (SW480 line) from a colon adenocarcinoma primary lesion and highly metastatic cells (SW620 line) from a metastatic lymph node from the same patient 1 year later. We used microarray analysis to identify miRNAs differentially expressed in SW480 and SW620 cells, focusing on miR-499-5p as a novel candidate prometastatic miRNA whose functions in cancer had not been studied. We confirmed increased miR-499-5p levels in highly invasive CRC cell lines and lymph node-positive CRC specimens. Furthermore, enhancing the expression of miR-499-5p promoted CRC cell migration and invasion in vitro and lung and liver metastasis in vivo, while silencing its expression resulted in reduced migration and invasion. Additionally, we identified FOXO4 and PDCD4 as direct and functional targets of miR-499-5p. Collectively, these findings suggested that miR-499-5p promoted metastasis of CRC cells and may be useful as a new potential therapeutic target for CRC.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Neoplasias Colorretais/patologia , MicroRNAs/fisiologia , Invasividade Neoplásica , Metástase Neoplásica , Proteínas de Ligação a RNA/metabolismo , Fatores de Transcrição/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Sequência de Bases , Proteínas de Ciclo Celular , Linhagem Celular Tumoral , Neoplasias Colorretais/metabolismo , Primers do DNA , Feminino , Fatores de Transcrição Forkhead , Humanos , Masculino , MicroRNAs/metabolismo , Pessoa de Meia-Idade , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase/métodos , Ligação Proteica
18.
Cancer Sci ; 101(7): 1653-60, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20491781

RESUMO

It has been reported that the 67-kDa laminin receptor (67LR) is implicated in cancer metastasis. We recently showed that 37LRP, the 67LR precursor, is a hypoxia-inducible factor 1 (HIF-1) target gene exposed to hypoxia in gastric cancer. Here, we investigated the role of 67LR in hypoxic metastasis and invasion in gastric cancer. Immunohistochemical analysis, western blotting, and RT-PCR assays revealed that 67LR was highly expressed in metastatic gastric cancers in vivo. Knockdown of the 67LR protein by RNA interference significantly decreased the adhesive, invasive, and in vivo metastatic abilities of the gastric cancer cell lines SGC7901 and MKN-45. Western blot analysis showed that 67LR increased the expression of urokinase-type plasminogen activator (uPA) and matrix metalloproteinase (MMP)-9, and decreased tissue inhibitor of matrix metalloproteinase (TIMP)-1 protein. We further showed that hypoxia induced 67LR expression in a time-dependent manner and this induction was inhibited by HIF-1 small-interfering (si) RNA. Both ERK and JNK inhibitors significantly inhibited hypoxia-induced expression of 67LR and the subsequent expression of uPA and MMP 9. SiRNA against 67LR or antibody against MMP9 and uPA significantly inhibited hypoxia-induced in vitro invasive ability. Taken together, these results reveal that 67LR promotes the invasive and metastatic ability of the gastric cancer cells through increasing uPA and MMP 9 expression, with involvement of the ERK and JNK signal pathway in hypoxia-induced 67 LR expressions and subsequent uPA and MMP9 expression.


Assuntos
Regulação Neoplásica da Expressão Gênica , Receptores de Laminina/genética , Neoplasias Gástricas/genética , Actinas/química , Actinas/genética , Sequência de Bases , Adesão Celular , Hipóxia Celular , Humanos , Imuno-Histoquímica , Linfonodos/patologia , Metaloproteinase 9 da Matriz/genética , Dados de Sequência Molecular , Invasividade Neoplásica , Metástase Neoplásica , RNA Interferente Pequeno/genética , Receptores de Laminina/química , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Neoplasias Gástricas/patologia , Neoplasias Gástricas/fisiopatologia , Ativador de Plasminogênio Tipo Uroquinase/genética
19.
Mol Carcinog ; 48(1): 56-65, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18506760

RESUMO

Hepatitis B virus (HBV) X protein (HBx) and cyclooxygenase-2 (COX-2) are all playing roles in hepatocellular carcinoma (HCC), but the reversing effects of COX-2 inhibitors on the neoplastic features caused by HBx protein is still unclear. To further evaluate the therapeutic potential of celecoxib on HBx mediated transformation, HCC cells transfected with HBx gene were treated with COX-2 selective inhibitor, celecoxib. The amount the main metabolite of COX-2, prostaglandin E2 (PGE2), was determined by using high sensitivity ELISA. Electron microscope and flow cytometry was used to analyze cell apoptosis and cell cycle distribution. RT-PCR and Western blot were used to identify the molecules involved in celecoxib induced cell apoptosis. The results showed that celecoxib inhibited cell growth more significantly and also induced more cell apoptosis in HBx over-expression cells than in control cells. Celecoxib could selectively inhibited COX-2 expression and PGE2 production. Celecoxib also inhibited p(473Ser)Akt, raf and p53 expression, and induced apoptosis by release of cytochrome c and activation of caspase 9, 3, and 6, which were more remarkably in HBx positive cells than in control cells. These results suggest that celecoxib had potent cell growth inhibitory effects on HBx positive HCC cells mainly through inducing more cell apoptosis, and these findings provide a new insight into the anticancer effects of celecoxib against HBx related HCC.


Assuntos
Apoptose/efeitos dos fármacos , Carcinoma Hepatocelular/patologia , Inibidores de Ciclo-Oxigenase 2/farmacologia , Neoplasias Hepáticas/patologia , Pirazóis/farmacologia , Sulfonamidas/farmacologia , Transativadores/fisiologia , Proteínas Virais Reguladoras e Acessórias/fisiologia , Western Blotting , Carcinoma Hepatocelular/enzimologia , Carcinoma Hepatocelular/virologia , Inibidores de Caspase , Caspases/metabolismo , Celecoxib , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Ciclo-Oxigenase 2/química , Ciclo-Oxigenase 2/metabolismo , Dinoprostona/metabolismo , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Imunofluorescência , Humanos , Immunoblotting , Técnicas Imunoenzimáticas , Imunoprecipitação , Neoplasias Hepáticas/enzimologia , Neoplasias Hepáticas/virologia , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-raf/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/metabolismo
20.
Cancer Lett ; 276(1): 88-94, 2009 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-19081179

RESUMO

Our previous studies indicated a direct correlation with loss of CIAPIN1 and carcinogenesis of tumor in human gastric cancer. Here we presented that the expression of CIAPIN1 was absent or significantly decreased in 102 cases of clear cell renal cell carcinoma (CCRCC) tissues (P<0.05). Up-regulating CIAPIN1 by adenoviral vectors exhibited significant inhibition of CCRCC-derived cell growth in vitro and in vivo with G1 cell cycle arrest. Simultaneously, CIAPIN1-induced growth suppression was found partially to regulate various proteins, including inhibition of cyclinD1, cyclinE, cdk2, cdk4, p-Rb and VEGF, but up-regulation of p27Kip1 and Rb.


Assuntos
Carcinoma de Células Renais/metabolismo , Proliferação de Células , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Neoplasias Renais/metabolismo , Biomarcadores Tumorais/análise , Western Blotting , Carcinoma de Células Renais/genética , Expressão Gênica , Humanos , Imuno-Histoquímica , Peptídeos e Proteínas de Sinalização Intracelular/genética , Neoplasias Renais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA