Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Breast Cancer Res ; 20(1): 63, 2018 07 02.
Article in English | MEDLINE | ID: mdl-29966525

ABSTRACT

After the publication of this work [1] an error in Fig. 1c was brought to our attention: the Western blots for PRDX6 and ß-actin were similar to those shown in lanes 5-6 of Fig. 4g. To verify these findings, we have repeated this experiment and the results are shown in a new Fig. 1c below. The repeated experimental results are consistent with the previously reported findings in the original study [1] and the functional role for PRDX6 in malignant progression of human cancer including breast cancer has been widely documented and recognized in numerous other studies [2]. We apologize for the error. However, this correction does not affect the conclusions of the article.

2.
Int J Cancer ; 142(5): 959-967, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29047106

ABSTRACT

To identify the role and to explore the mechanism of extracellular 5'-nucleotidase (CD73) in human breast cancer growth, CD73 expression was measured firstly in breast cancer tissues and cell lines, and then interfered with or over-expressed by recombinant lentivirus in cell lines. Impacts of CD73 on breast cancer cell proliferation and cell cycle were investigated with colony formation assay, CCK-8 and flow cytometry. The relationship between CD73 and AKT/GSK-3ß/ß-catenin pathway was assessed with adenosine, adenosine 2A receptor antagonist (SCH-58261), adenosine 2A receptor agonist (NECA), CD73 enzyme inhibitor (APCP) and Akt inhibitor (MK-2206). Moreover, the effect of CD73 on breast cancer growth in vivo was examined with human breast cancer transplanting model of nude mice. The results showed that the expression of CD73 was high in breast cancer tissues and increased with advanced tumor grades and lympho-node status. CD73 expression was higher in more malignant cells, and CD73 overexpression promoted breast cancer cell proliferation in both in vivo and in vitro. It activated AKT/GSK-3ß/ß-catenin/cyclinD1 signaling pathway through CD73 enzyme activity and other mechanism.


Subject(s)
5'-Nucleotidase/metabolism , Breast Neoplasms/pathology , Cell Proliferation , Cyclin D1/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Proto-Oncogene Proteins c-akt/metabolism , beta Catenin/metabolism , 5'-Nucleotidase/genetics , Animals , Apoptosis , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Cycle , Cell Movement , Cyclin D1/genetics , Female , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Glycogen Synthase Kinase 3 beta/genetics , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Prognosis , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , beta Catenin/genetics
3.
Cancer Lett ; 412: 69-80, 2018 01 01.
Article in English | MEDLINE | ID: mdl-28964785

ABSTRACT

Metastasis and chemoresistance are two major causes of breast cancer death. We show here that the chemokine receptor CXCR2 was overexpressed in breast cancer cell lines and tissues. CXCR2 promoted anti-apoptosis, anti-senescence, and epithelial-to-mesenchymal transition (EMT) of breast cancer cells, leading to the enhanced metastasis and chemoresistance. Further study suggested that AKT1 and cyclooxygenase-2 (COX2; PTGS2) might mediate the CXCR2 signaling to inversely control the breast cancer metastasis and chemoresistance through the regulation of EMT, apoptosis, and senescence. Analyses of clinical data indicate that the high expression of CXCR2 was correlated with the high expression of COX2 and the low expression of AKT1, P85α, E-cadherin, and ß-catenin in cancer tissues. Poor outcomes were associated with the high expression of CXCR2 or COX2 while favorable survivals were associated with the high expression of P85α, AKT1, or E-cadherin in all cancer patients. Cox multivariate analysis demonstrated that CXCR2, COX2, and AKT1 could be independent predictors for disease free survivals. All these data suggest that CXCR2 promotes breast cancer metastasis and chemoresistance via suppressing AKT1 and activating COX2. Thus, antagonists of the CXCR2 signaling molecules may be used to treat breast cancer patients particularly with high metastasis and chemoresistance.


Subject(s)
Breast Neoplasms/pathology , Cyclooxygenase 2/physiology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Receptors, Interleukin-8B/physiology , Apoptosis , Breast Neoplasms/drug therapy , Breast Neoplasms/mortality , Cell Line, Tumor , Cell Movement , Cell Proliferation , Drug Resistance, Neoplasm , Epithelial-Mesenchymal Transition , Female , Humans , Proto-Oncogene Proteins c-akt/physiology , Receptors, Interleukin-8B/antagonists & inhibitors
5.
Sci Rep ; 6: 22831, 2016 Mar 09.
Article in English | MEDLINE | ID: mdl-26956544

ABSTRACT

Mitochondria are the powerhouses of eukaryotic cells and the main source of reactive oxygen species (ROS) in hypoxic cells, participating in regulating redox homeostasis. The mechanism of tumor hypoxia tolerance, especially the role of mitochondria in tumor hypoxia resistance remains largely unknown. This study aimed to explore the role of mitochondria in tumor hypoxia resistance. We observed that glycolysis in hypoxic cancer cells was up-regulated more rapidly, with far lesser attenuation in aerobic oxidation, thus contributing to a more stable ATP/ADP ratio. In hypoxia, cancer cells rapidly convert hypoxia-induced O(2˙)(-) into H2O2. H2O2 is further decomposed by a relatively stronger antioxidant system, causing ROS levels to increase lesser compared to normal cells. The moderate ROS leads to an appropriate degree of autophagy, eliminating the damaged mitochondria and offering nutrients to promote mitochondria fusion, thus protects mitochondria and improves hypoxia tolerance in cancer. The functional mitochondria could enable tumor cells to flexibly switch between glycolysis and oxidative phosphorylation to meet the different physiological requirements during the hypoxia/re-oxygenation cycling of tumor growth.


Subject(s)
Homeostasis , Mitochondria/drug effects , Mitochondria/physiology , Reactive Oxygen Species/metabolism , Tumor Hypoxia , Aerobiosis , Cell Line, Tumor , Glycolysis , Humans , Oxidation-Reduction , Oxidative Phosphorylation
6.
Med Oncol ; 32(11): 254, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26487662

ABSTRACT

The human chemokine receptor CCRL2 is a member of the atypical chemokine receptor family. CCRL2 is unable to couple with G-proteins and fails to induce classical chemokine signaling for the highly conserved DRYLAIV motif essential for signaling has been changed to QRYLVFL. We investigated whether CCRL2 is involved in the chemotaxis, invasion, and proliferation of human breast cancer cells. Firstly, expression of CCRL2 was determined in six breast cancer cell lines by real-time RT-PCR and Western blot. Then, we established stable cell lines overexpressing CCRL2 to explore the function of CCRL2 in chemotaxis and invasion by transwell assays, and the signaling downstream was further investigated. The effect of CCRL2 on proliferation was detected by colony formation assays and tumor xenograft study. We found that stable overexpression of CCRL2 in MDA-MB-231 and BT-549 cells attenuated the chemotaxis and invasion stimulated by its ligand CCL2. CCRL2 inhibits p38 MAPK (p38) phosphorylation and up-regulates the expression of E-cadherin. This effect was eliminated by the inhibitor of p38 MAPK. CCRL2 inhibited the growth of breast cancer cells in vitro and in vivo. Our results suggest that CCRL2 functions as a tumor suppressor in human breast cancer cells.


Subject(s)
Breast Neoplasms/metabolism , Chemokine CCL2/metabolism , Chemotaxis/physiology , Receptors, CCR/biosynthesis , p38 Mitogen-Activated Protein Kinases/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Chemokine CCL2/antagonists & inhibitors , Female , Humans , MCF-7 Cells , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/prevention & control , Phosphorylation/physiology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
7.
Med Oncol ; 31(9): 145, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25097078

ABSTRACT

The aim of this study was to determine the frequency of axillary lymph node (ALN) metastasis of early breast cancers by evaluating the status of DARC, D6 and CCX-CKR and the levels of VEGF and MMP-9. The status of DARC, D6 and CCX-CKR and the levels VEGF and MMP-9 were evaluated in ALN- (n = 130) and ALN + (n = 88) patients with T1 breast cancer by immunohistochemical staining. For ALN, likelihood ratio χ (2)-tests were used for univariate analysis and logistic regression for multivariate analysis. Univariate analysis identified the nuclear grade, VEGF and MMP-9 expression and absence of DARC, D6 and CCX-CKR as predictors of ALN involvement. When combining the three receptors (DARC, D6 and CCX-CKR) together, tumors with multiple absence (multi-absence, any two or three loss) had a higher likelihood of being ALN positive than non-multi-absence (coexpression of any two or three) tumors (56.2 vs. 27.9 %, P < 0.001). The final multivariate logistic regression revealed nuclear grade, VEGF, MMP-9 and non-multi-absence versus multi-absence to be independent predictors of ALN involvement; the odds ratio (OR) and 95 % CI for non-multi-absence tumors versus multi-absence were 0.469 (0.233-0.943). Multi-absence was also associated with the involvement of four or more lymph nodes among ALN + tumors. Moreover, tumors with multi-absence had higher VEGF (78.1 vs. 50.0 %, P < 0.001) and MMP-9 (81.3 vs. 36.1 %, P < 0.001) expression than non-multi-absence tumors. Our data highlight that the absence of DARC, D6 and CCX-CKR in combination, which is associated with higher VEGF and MMP-9 expression, predicts the presence and extent of ALN metastasis in breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Lymphatic Metastasis , Matrix Metalloproteinase 9/metabolism , Vascular Endothelial Growth Factor A/metabolism , Adult , Aged , Aged, 80 and over , Breast Neoplasms/epidemiology , Chi-Square Distribution , Duffy Blood-Group System/analysis , Duffy Blood-Group System/metabolism , Female , Humans , Immunohistochemistry , Matrix Metalloproteinase 9/analysis , Middle Aged , Odds Ratio , Receptors, CCR/analysis , Receptors, CCR/metabolism , Receptors, Cell Surface/analysis , Receptors, Cell Surface/metabolism , Vascular Endothelial Growth Factor A/analysis
8.
Cancer Cell Int ; 14: 107, 2014.
Article in English | MEDLINE | ID: mdl-25729327

ABSTRACT

BACKGROUND: Suberoyl bis-hydroxamic acid (SBHA) is a histone deacetylase (HDAC) inhibitor and exerts anti-growth effects in several malignancies including breast cancer. Proteasome inhibitors such as Bortezomib and MG-132 constitute novel anticancer agents. In this study, we investigated the synergistic antitumour activity of SBHA in combination with proteasome inhibitors. METHODS: MCF-7 and MDA-MB-231 breast cancer cells were treated with SBHA, Bortezomib, and MG-132 alone or in combination for 72 h. Cell proliferation, colony formation, apoptosis and gene expression changes were examined. RESULTS: SBHA, Bortezomib, and MG-132 alone significantly inhibited the proliferation and colony formation and induced apoptosis in MCF-7 and MDA-MB-231 cells. Combined treatment showed a good synergistic antitumour effect against breast cancer cells. The p53 protein level was significantly elevated by combined treatment with SBHA and proteasome inhibitors. Moreover, combined treatment increased the expression of Bax, Bcl-xS, and Bak and decreased the expression of Bcl-2. Combination of SBHA with proteasome inhibitors causes synergistic anticancer effects on breast cancer cells. The potential molecular mechanism may involve induction of p53 and modulation of the Bcl-2 family proteins. CONCLUSION: These findings warrant further investigation of the therapeutic benefits of combination of SBHA with proteasome inhibitors in breast cancer.

9.
PLoS One ; 8(11): e78901, 2013.
Article in English | MEDLINE | ID: mdl-24260134

ABSTRACT

The inhibitory effect of two chemokine decoy receptors (CDRs), DARC and D6, on breast cancer metastasis is mainly due to their ability to sequester pro-malignant chemokines. We hypothesized that genetic variants in the DARC and CCBP2 (encoding D6) genes may be associated with breast cancer progression. In the present study, we evaluated the genetic contributions of DARC and CCBP2 to metastatic potential, indicated by lymph node metastasis (LNM). Ten single-nucleotide polymorphisms (SNPs) (potentially functional SNPs and block-based tagging SNPs) in DARC and CCBP2 were genotyped in 785 breast cancer patients who had negative lymph nodes and 678 patients with positive lymph nodes. Two non-synonymous SNPs, rs12075 (G42D) in DARC and rs2228468 (S373Y) in CCBP2, were observed to be associated with LNM in univariate analysis and remained significant after adjustment for conventional clinical risk factors, with odds ratios (ORs) of 0.54 (95% confidence interval [CI], 0.37 to 0.79) and 0.78 (95% CI, 0.62 to 0.98), respectively. Additional functional experiments revealed that both of these significant SNPs could affect metastasis of breast cancer in xenograft models by differentially altering the chemokine sequestration ability of their corresponding proteins. Furthermore, heterozygous GD genotype of G42D on human erythrocytes had a significantly stronger chemokine sequestration ability than homozygous GG of G42D ex vivo. Our data suggest that the genetic variants in the CDR genes are probably associated with the varied metastatic potential of breast cancer. The underlying mechanism, though it needs to be further investigated, may be that CDR variants could affect the chemokine sequestration ability of CDR proteins.


Subject(s)
Breast Neoplasms/genetics , Duffy Blood-Group System/genetics , Polymorphism, Single Nucleotide , Receptors, Cell Surface/genetics , Receptors, Chemokine/genetics , Adult , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Duffy Blood-Group System/metabolism , Female , Heterografts , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Neoplasm Metastasis , Neoplasm Transplantation , Receptors, Cell Surface/metabolism , Receptors, Chemokine/metabolism
10.
ACS Appl Mater Interfaces ; 5(21): 10721-30, 2013 Nov 13.
Article in English | MEDLINE | ID: mdl-24083448

ABSTRACT

In the treatment of cancer, multidrug resistance (MDR) has been the major obstacle to the success of chemotherapy. The underlying mechanism relies on the overexpression of drug-efflux transporters that prevent the intracellular transport of the drug. In this study, reduction-cleavable vesicles were designed and developed with efficient glutathione-mediated drug-release behavior for reversing drug resistance. Polymeric vesicles were self-assembled from triblock copolymers with disulfide-bond-linked poly(ethylene glycol) (PEG) and poly(ε-benzyloxycarbonyl-L-lysine) (PzLL). Observations from transmission electron microscopy (TEM) and confocal laser scanning microscopy (CLSM) outline an obvious hollow structure surrounded by a thin outer layer, indicating the successful formation of the vesicles. Using fluorescently detectable doxorubicin hydrochloride (DOX·HCl) as the model drug, a significant acceleration of drug release regulated by glutathione (GSH) was found (>3-fold difference). Upon incubation of the DOX·HCl-loaded polymeric vesicles with the HeLa cervical cancer cell line exposed to glutathione, an enhanced nuclear accumulation of DOX·HCl was observed, elicited by the preferred disassembly of the vesicle structure under reducing conditions. Importantly, by using the gemcitabine hydrochloride (GC·HCl)-resistant breast cancer cell line MDA-MB-231, it was found that cell viability was significantly reduced after treatment with GC·HCl-loaded polymeric vesicles, indicating that these vesicles can help to reverse the drug resistance.


Subject(s)
Drug Delivery Systems , Drug Resistance, Neoplasm/drug effects , Glutathione/chemistry , Neoplasms/drug therapy , Doxorubicin/administration & dosage , Glutathione/administration & dosage , HeLa Cells , Humans , Microscopy, Electron, Transmission , Polymers/chemistry
11.
Tumour Biol ; 34(5): 2817-26, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23771849

ABSTRACT

Radioresistance is one of the major barriers to improve the survival rate of breast cancer patients. Cyclooxygenase 2 (COX-2) is usually overexpressed in highly invasive and metastatic breast cancer, which may indicate an association with breast cancer radioresistance. The function role of COX-2 was investigated by using a radioresistant breast cancer cell line MDA-MB-231/RR10 and its parental cell line MDA-MB-231 cells before or after COX-2 was silenced by a specific small hairpin RNA (shRNA). The cell proliferation, migration, invasion, colony formation, and apoptosis were measured by CCK-8, scratch-wound, transwell, clone formation assay, and flow cytometry. Protein and mRNA expression were analyzed by Western blot and quantitative reverse transcriptase-polymerase chain reaction. COX-2 is upregulated in MDA-MB-231/RR10 cells compared with in MDA-MB-231 cells, and silencing of COX-2 expression by shRNA in MDA-MB-231/RR10 cells decreases the expression of Bcl-2 and Bcl-XL, but increases the proapoptotic protein BAK, leading to the increased apoptosis following treatment with γ-irradiation in comparison with those in control cells. Silencing of COX-2 also increases the expression of ß-catenin and E-cadherin, two anti-invasion proteins, resulting in reduced cell migration and invasion tested by transwell chambers and wound-healing assays. Further study demonstrated that COX-2-induced radioresistance is negatively regulated through the phosphorylation of p38 at Tyr182, and that the phosphorylation of p38 induced by TNF-alpha reduces the expression of Bcl-2, BCL-XL, but increases ß-catenin and E-cadherin, leading to the decreased invasiveness of cells. Our data suggest that COX-2, p38, Bcl-2, Bcl-XL, ß-catenin, and E-cadherin may be considered as potential therapeutic targets against radioresistant breast cancer.


Subject(s)
Apoptosis/radiation effects , Breast Neoplasms/enzymology , Cyclooxygenase 2/physiology , p38 Mitogen-Activated Protein Kinases/metabolism , Apoptosis Regulatory Proteins/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/radiotherapy , Cell Line, Tumor/radiation effects , Cell Movement , Cell Proliferation , Cell Survival/radiation effects , Female , Humans , MAP Kinase Signaling System , Neoplasm Invasiveness , Phosphorylation , Protein Processing, Post-Translational , Radiation Tolerance , Tumor Necrosis Factor-alpha/physiology
12.
Clin Exp Metastasis ; 30(5): 671-80, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23508889

ABSTRACT

Angiogenesis is essential for tumor growth, progression and metastasis. Studies indicate that expression and activity of ecto-5'-nucleotidase (CD73) are elevated in metastatic carcinomas. Our previous studies found that angiogenesis of tumor xenografts was decreased when the activity of CD73 in cancer cells was inhibited, implying that this enzyme is involved in tumor angiogenesis. To elucidate the mechanism, we investigated CD73 influence on tumor angiogenesis in both in vitro assays and in tumor bearing mice. We found that capillary-like structures were formed more in CD73(+/+) pulmonary microvascular endothelial cells (PMECs) than CD73(-/-) PMECs, and this was more pronounced when the cells were cultured in cancer-conditioned medium. Meanwhile, CD73 decreased endothelial cells adhesion to collagen IV and promoted migration. Additionally, the extent of tumor angiogenesis and the size of tumors were greater in CD73(+/+) mice than in CD73(-/-) mice. Thus, we concluded that CD73 can promote endothelial cells forming new vessels in cancer condition, facilitating tumor growth and hematogenous metastasis.


Subject(s)
5'-Nucleotidase/physiology , Neoplasms/blood supply , Neovascularization, Pathologic/therapy , 5'-Nucleotidase/immunology , Animals , Cells, Cultured , Culture Media, Conditioned , GPI-Linked Proteins/immunology , Humans , Mice , Xenograft Model Antitumor Assays
13.
PLoS One ; 8(1): e49312, 2013.
Article in English | MEDLINE | ID: mdl-23382798

ABSTRACT

Experimental data suggest a protective effect of vitamin D on breast cancer; however, epidemiologic results remain inclusive. With a Chinese population-based case-control study and meta-analysis of the observational studies, we here systematically evaluated the association of blood 25(OH)D level and breast cancer risk. With 593 breast cancer cases and 580 cancer-free controls from Shanghai, China, we found that 80% of the normal women had severe vitamin D deficiency (less than 20 ng/mL) and 15.2% had mild deficiency (20 to 30 ng/mL) and only 4.8% of women had sufficient vitamin D level (>30 ng/mL) while the proportion was 96.1%, 3.2% and 0.7% respectively for the breast cancer patients. Compared to those with the lowest quartile of plasma 25(OH)D level, women with highest quartile 25(OH)D level showed a significant decreased breast cancer risk (Q4 vs.Q1: OR = 0.10, 95% CI = 0.06-0.15) and every 1 ng/ml increment of plasma 25(OH)D level led to a 16% lower odds of breast cancer (OR = 0.84, 95% CI = 0.81-0.87; P<0.001). From the meta-analysis of the observational studies, we found that women with highest quantile of blood 25(OH)D level was associated with a significantly reduced breast cancer risk compared to those with lowest quantile of blood 25(OH)D level for the 11 nested case-control and retrospective studies (pooled OR = 0.86, 95% CI = 0.75-1.00) and 10 case-control studies (7 population based, OR = 0.35, 95% CI = 0.24-0.52; 3 hospital based, OR = 0.08, 95% CI = 0.02-0.33). These results suggest that vitamin D may have a chemo-preventive effect against breast cancer.


Subject(s)
Breast Neoplasms/genetics , Genetic Association Studies , Vitamin D/genetics , Adult , Aged , Aged, 80 and over , Breast Neoplasms/blood , Breast Neoplasms/pathology , Case-Control Studies , Female , Humans , Middle Aged , Retrospective Studies , Risk Factors , Vitamin D/analogs & derivatives , Vitamin D/blood , Vitamin D/metabolism
14.
J Radiat Res ; 54(2): 230-4, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23090953

ABSTRACT

Intensity-modulated radiation therapy, when used in the clinic, prolongs fraction delivery time. Here we investigated both the in vivoand in vitroradiobiological effects on the A549 cell line, including the effect of different delivery times with the same dose on A549 tumor growth in nude mice. The in vitroeffects were studied with clonogenic assays, using linear-quadratic and incomplete repair models to fit the dose-survival curves. Fractionated irradiation of different doses was given at one fraction per day, simulating a clinical dose-time-fractionation pattern. The longer the interval between the exposures, the more cells survived. To investigate the in vivoeffect, we used sixty-four nude mice implanted with A549 cells in the back legs, randomly assigned into eight groups. A 15 Gy radiation dose was divided into different subfractions. The maximum and minimum tumor diameters were recorded to determine tumor growth. Tumor growth was delayed for groups with prolonged delivery time (40 min) compared to the group receiving a single dose of 15 Gy (P< 0.05), and tumors with a 20 min delivery time had delayed growth compared to those with a 40 min delivery time [20' (7.5 Gy × 2 F) vs 40' (7.5 Gy × 2 F), P= 0.035; 20' (3 Gy × 5 F) vs 40' (3 Gy × 5 F); P= 0.054; 20' (1.67 Gy × 9 F) vs 40' (1.67 Gy × 9 F), P= 0.028]. A prolonged delivery time decreased the radiobiological effects, so we strongly recommend keeping the delivery time as short as possible.


Subject(s)
Dose Fractionation, Radiation , Lung Neoplasms/physiopathology , Lung Neoplasms/radiotherapy , Radiotherapy, Conformal/methods , Animals , Cell Line, Tumor , Cell Survival/radiation effects , Dose-Response Relationship, Radiation , Lung , Lung Neoplasms/pathology , Mice , Mice, Nude , Radiotherapy Dosage , Treatment Outcome
15.
BMC Cancer ; 12: 374, 2012 Aug 28.
Article in English | MEDLINE | ID: mdl-22928984

ABSTRACT

BACKGROUND: Different ethnicities have different distribution of Duffy blood group (DBG) phenotypes and different breast cancer morbidity. A study in our lab demonstrated that Duffy antigen/receptor for chemokines (DARC, also known as DBGP, the Duffy protein phenotype), led to the inhibition of tumorigenesis. Therefore, we tested the hypothesis that DBGP is correlated with breast cancer occurrence. METHODS: DBGP proteins were examined by indirect antiglobulin testing with anti-FYa and anti-FYb antibodies. The phenotypes were classified into four groups according to the agglutination reactions: FYa + FYb+, FYa + FYb-, FYa-FYb + and FYa-FYb-. The phenotypes and pathological diagnosis of consecutively hospitalized female patients (n = 5,022) suffering from breast cancer at the Shanghai Cancer Hospital and Henan Province Cancer Hospital were investigated. The relationships between DBGP expression with breast cancer occurrence, axillary lymph status, histological subtype, tumor size pathological grade and overall survival were analyzed. RESULTS: The incidence of breast cancer was significantly different between FYa + FYb + (29.8%), FYa + FYb- (33.2%), FYa-FYb + (45.6%) and FYa-FYb- (59.1%; P = 0.001). Significant different numbers of breast cancer patients had metastases to the axillary lymph nodes in the FYa + FYb + group (25.1%), FYa + FYb- (36.9%), FYa-FYb + (41.0%) and FYa-FYb- (50.0%, (P = 0.005). There was a statistical significance (p = 0.022) of the overall survival difference between patients with difference phenotypes. No significant difference was observed in cancer size (t-test, p > 0.05), histological cancer type (Fisher's exact test, p > 0.05) or histological grade (Fisher's exact test, p > 0.05) between every each DBGP group. CONCLUSIONS: DBGP is correlated with breast cancer incidence and axillary lymph node metastasis and overall survival. Further investigations are required to determine the underlying mechanism of Duffy blood group phenotype on breast cancer risk.


Subject(s)
Breast Neoplasms/blood , Duffy Blood-Group System , Adolescent , Adult , Aged , Aged, 80 and over , Breast Neoplasms/epidemiology , Breast Neoplasms/pathology , Breast Neoplasms/surgery , China/epidemiology , Coombs Test , Female , Humans , Incidence , Middle Aged , Phenotype , Survival Analysis
16.
Breast Cancer Res Treat ; 135(3): 725-35, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22910931

ABSTRACT

CXCL14, also known as breast and kidney-expressed chemokine, was initially identified as a chemokine highly expressed in the kidney and breast. The exact function of CXCL14 in human breast cancer is still unclear, although it has been testified to play an anti-tumor role in other tumors, including head and neck squamous cell carcinoma, lung cancer, prostate cancer, and so on. In this study, we tried to demonstrate the relationship between CXCL14 and breast cancer. CXCL14 expressions were detected by reverse transcription-PCR and western blot in 2 normal breast epithelial cell lines and 6 breast cancer cell lines. The effects of CXCL14 on the proliferation and invasion in vitro were tested using the CXCL14-overexpressing cells (MDA-MB-231HM-CXCL14) which were established by stable transfection. We established an orthotropic xenograft tumor model in SCID mice using the MDA-MB-231HM-CXCL14 cells and explored the influence of CXCL14 overexpression on tumor growth and metastasis in vivo. Furthermore, we detected the protein level of CXCL14 in 208 breast cancer patients by immunohistochemistry and discussed the correlation between CXCL14 and the prognosis of breast cancer. CXCL14 mRNA expression is lower in breast cancer cell lines, and MDA-MB-231HM express the lowest levels of CXCL14 mRNA. Overexpression of CXCL14 inhibited cell proliferation and invasion in vitro and attenuated xenograft tumor growth and lung metastasis in vivo. CXCL14 protein level is positively correlated to the overall survival of all patients as well as the patients with lymph node metastasis, and it has a negative correlation with the lymph node metastasis. Our study showed for the first time that CXCL14 is a negative regulator of growth and metastasis in breast cancer. The re-expression or up-regulation of this gene may provide a novel strategy in breast cancer therapy in the future.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Chemokines, CXC/genetics , Chemokines, CXC/metabolism , Adult , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Cell Line, Tumor , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/secondary , Lymphatic Metastasis/genetics , Lymphatic Metastasis/pathology , Mice , Mice, SCID , Middle Aged , Xenograft Model Antitumor Assays
17.
Breast Cancer Res Treat ; 131(3): 837-48, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21479551

ABSTRACT

Increasing evidence has shown that chemokines and chemokine receptors are associated with tumor growth and metastasis. CCR4, an important chemokine receptor for regulating immune homeostasis, is thought to be involved in hematologic malignancies and has also recently implicated in some solid tumors, such as gastric cancer. The possible role of CCR4 in breast cancer has not been well elucidated. In this study, we show that CCR4 is differentially expressed in human breast cancer cell lines. Specifically, we find that CCR4 is overexpressed in breast cancer cell lines with high metastatic potential. More importantly, we used a combination of overexpression and RNA interference to demonstrate that CCR4 promotes breast tumor growth and lung metastasis in mice. Furthermore, we find that microvessel density is significantly increased in tumors formed by CCR4-overexpressing cells and decreased in those formed by CCR4-knockdown cells. We find that overexpression of CCR4 can enhance the chemotactic response of breast cancer cells to CCL17. However, the expression of CCR4 does not affect the proliferation of breast cancer cells in vitro. Furthermore, we show that CCR4 expression is positively correlated with HER2 expression, tumor recurrence and lymph node, lung and bone metastasis (P < 0.05). Multivariate analysis showed that CCR4 expression is a significant independent prognostic factor for overall survival (P = 0.036) but not for disease-free survival in patients with breast cancer (P = 0.071). Survival analysis indicated a strong association between CCR4 expression and lower overall survival (P = 0.0001) and disease-free survival (P = 0.016) in breast cancer.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Lung Neoplasms/genetics , Lung Neoplasms/secondary , Receptors, CCR4/genetics , Animals , Breast Neoplasms/mortality , Cell Line, Tumor , Cell Proliferation , Chemokine CCL17/metabolism , Chemokine CCL22/metabolism , Disease Progression , Female , Gene Expression , Genetic Vectors/genetics , Humans , Lentivirus/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Metastasis , Prognosis , RNA Interference , Survival Analysis , Transduction, Genetic , Xenograft Model Antitumor Assays
18.
Breast Cancer Res Treat ; 131(1): 65-73, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21465172

ABSTRACT

Polycomb group (PcG) proteins have recently been shown related to cancer development. The PcG protein EZH2 is involved in progression of prostate and breast cancers, and has been identified as a molecular marker in breast cancer. Nevertheless, the molecular mechanism by which PcG proteins regulate cancer progression and malignant metastasis is still unclear. PcG proteins methylate H3K27 in undifferentiated epithelial cells, resulting in the repression of differentiation genes such as HOX. FOXC1 is a member of the Forkhead box transcription factor family, which plays an important role in differentiation, and is involved in eye development. We discovered in this study that the expression of FOXC1 gene was negatively correlated to that of PcG genes, i.e., Bmi1, EZH2, and SUZ12, in MCF-7 and MDA-MB-231 cells. To investigate the regulatory effects of PcG proteins on FOXC1 gene, the two cell lines were transfected with either expression plasmids or siRNA plasmids of Bmi1, EZH2, and SUZ12, and we found that PcGs, especially EZH2, could repress the transcription of FOXC1 gene. Chromatin immunoprecipitation (ChIP) assay showed that histone methylation and acetylation modifications played critical roles in this regulatory process. When FOXC1 was stably transfected into MDA-MB-231 cells, the migration and invasion of the cells were repressed. Moreover, the tumorigenicity and the spontaneous metastatic capability regulated by FOXC1 were determined by using an orthotropic xenograft tumor model of athymic mice with the FOXC1-MDA-MB-231HM and the GFP-MDA-MB-231HM cells, and the results showed that FOXC1 in MDA-MB-231HM cells inhibited migration and invasion in vitro and reduced the pulmonary metastasis in vivo. Data presented in this report contribute to the understanding of the mechanisms by which EZH2 participates in tumor development.


Subject(s)
Breast Neoplasms/pathology , Forkhead Transcription Factors/metabolism , Repressor Proteins/metabolism , Acetylation , Animals , Breast Neoplasms/genetics , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line, Tumor , Cell Movement , Chromatin Immunoprecipitation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Enhancer of Zeste Homolog 2 Protein , Female , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Neoplastic , Histones/metabolism , Humans , Methylation , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Metastasis , Neoplasm Proteins , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Polycomb Repressive Complex 1 , Polycomb Repressive Complex 2 , Polycomb-Group Proteins , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA Interference , RNA, Small Interfering , Repressor Proteins/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Transfection/methods
19.
Breast Cancer Res Treat ; 125(3): 715-27, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20369284

ABSTRACT

Some evidence suggests that atypical chemokine binders (ACBs) including DARC, D6, and CCX-CKR play an important role in inhibiting invasion and metastasis of cancer cells; however, their expression in breast cancer has not been well characterized. The purpose of this study was to determine the predictive value of ACBs for relapse-free survival and overall survival in breast cancer. The expressions of the three molecules were analyzed immunohistochemically in a total of 558 consecutive breast specimens comprising 12 normal breast tissues, 29 noninvasive (carcinoma in situ), and 517 invasive breast carcinoma and their relationships to clinicopathological features and survival were investigated in invasive breast cancer. Coexpression of ACBs in invasive breast carcinoma (55.9%) was much lower that of noninvasive breast carcinoma (93.1%) and normal breast tissue (100.0%), P = 0.0004, 0.0096, respectively. Their separate stainings in invasive cancer were significantly conversely correlated with lymph node status and tumor stage. In univariate analysis, the three proteins and their coexpression were significantly associated with higher relapse-free survival and overall survival. In multivariate analysis, each of these molecules was favorable for relapse-free survival, but not overall survival. Surprisingly, their coexpression was not only independently prognostic factor for relapse-free survival (RR = 0.182, 95% CI: 0.101-0.327, P < 0.001), but also for overall survival (RR = 0.271, 95% CI: 0.081-0.910, P = 0.035). These findings highlight that the multiple loss of ACBs may occur during the development of tumorigenesis and their coexpression in breast cancer is predictive of favorable outcomes.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/therapy , Chemokines/metabolism , Gene Expression Regulation, Neoplastic , Adult , Aged , Aged, 80 and over , Duffy Blood-Group System/biosynthesis , Female , Humans , Immunohistochemistry/methods , Menopause , Middle Aged , Receptors, CCR/biosynthesis , Receptors, CCR10/biosynthesis , Receptors, Cell Surface/biosynthesis , Treatment Outcome , Chemokine Receptor D6
20.
Clin Cancer Res ; 15(9): 2962-70, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19383822

ABSTRACT

PURPOSE: The biological axes of chemokines and chemokine receptors, such as CXCR4/CXCL12, CCR7/CCL19 (CCL21), CCR9/CCL25, and CXCR5/CXCL13, are involved in cancer growth and metastasis. This study is aimed at the potential regulatory role of atypical chemokine binder CCX-CKR, as a scavenger of CCL19, CCL21, CCL25, and CXCL13, in human breast cancer. EXPERIMENTAL DESIGN: The role of CCX-CKR in human breast cancer was investigated in cell lines, animal models, and clinical samples. RESULTS: Overexpression of CCX-CKR inhibited cancer cell proliferation and invasion in vitro and attenuated xenograft tumor growth and lung metastasis in vivo. CCX-CKR can be regulated by cytokines such as interleukin-1beta, tumor necrosis factor-alpha, and IFN-gamma. Lack or low expression of CCX-CKR correlated with a poor survival rate in the breast cancer patients. A significant correlation between CCX-CKR and lymph node metastasis was observed in human breast cancer tissues. CCX-CKR status was an independent prognostic factor for disease-free survival in breast cancer patients. CONCLUSION: We showed for the first time that CCX-CKR is a negative regulator of growth and metastasis in breast cancer mainly by sequestration of homeostatic chemokines and subsequent inhibition of intratumoral neovascularity. This finding may lead to a new therapeutic strategy against breast cancer.


Subject(s)
Breast Neoplasms/mortality , Breast Neoplasms/pathology , Gene Expression Regulation, Neoplastic/physiology , Receptors, CCR/genetics , Animals , Blotting, Western , Breast Neoplasms/genetics , Carcinoma, Ductal, Breast/genetics , Carcinoma, Ductal, Breast/mortality , Carcinoma, Ductal, Breast/secondary , Cell Proliferation , Enzyme-Linked Immunosorbent Assay , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Interferon-gamma/pharmacology , Interleukin-1beta/pharmacology , Lung Neoplasms/genetics , Lung Neoplasms/mortality , Lung Neoplasms/secondary , Lymphatic Metastasis , Mice , Middle Aged , Neoplasm Invasiveness , Prognosis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...