Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 184
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Intern Med ; 295(5): 634-650, 2024 May.
Article in English | MEDLINE | ID: mdl-38439117

ABSTRACT

BACKGROUND: The immune reconstitution after allogeneic hematopoietic stem cell transplantation (allo-HSCT) is crucial for preventing infections and relapse and enhancing graft-versus-tumor effects. B cells play an important role in humoral immunity and immune regulation, but their reconstitution after allo-HSCT has not been well studied. METHODS: In this study, we analyzed the dynamics of B cells in 252 patients who underwent allo-HSCT for 2 years and assessed the impact of factors on B-cell reconstitution and their correlations with survival outcomes, as well as the development stages of B cells in the bone marrow and the subsets in the peripheral blood. RESULTS: We found that the B-cell reconstitution in the bone marrow was consistent with the peripheral blood (p = 0.232). B-cell reconstitution was delayed by the male gender, age >50, older donor age, the occurrence of chronic and acute graft-versus-host disease, and the infections of fungi and cytomegalovirus. The survival analysis revealed that patients with lower B cells had higher risks of death and relapse. More importantly, we used propensity score matching to obtain the conclusion that post-1-year B-cell reconstitution is better in females. Meanwhile, using mediation analysis, we proposed the age-B cells-survival axis and found that B-cell reconstitution at month 12 posttransplant mediated the effect of age on patient survival (p = 0.013). We also found that younger patients showed more immature B cells in the bone marrow after transplantation (p = 0.037). CONCLUSION: Our findings provide valuable insights for optimizing the management of B-cell reconstitution and improving the efficacy and safety of allo-HSCT.


Subject(s)
Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Female , Humans , Male , Transplantation, Homologous , Hematopoietic Stem Cell Transplantation/adverse effects , Graft vs Host Disease/etiology , Graft vs Host Disease/prevention & control , Graft vs Host Disease/epidemiology , B-Lymphocytes , Recurrence
2.
Cancer Invest ; 41(2): 119-132, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36346393

ABSTRACT

A disintegrin-like and metalloprotease with therombospondin type1 motif 8 (ADAMTS8) plays an important role in many malignancies. However, the clinical and biological significance of ADAMTS8 in breast cancer remain unknown. In this study, the clinical data from 1066 breast cancer patients were analyzed by The Cancer Genome Atlas (TCGA) database, and were analyzed using the correlation between ADAMTS8 expression and the clinicopathological features and prognoses. The CCK-8 assay, clone formation assay, flow cytometry and Transwell assay were used to characterize the effects of ADAMTS8 on proliferation, migration and invasion of breast cancer cells. Gene set enrichment analysis (GSEA) and western blotting were used to identify the potential molecular mechanism on how ADAMTS8 exert its biological function. ADAMTS8 overexpression correlated longer overall survival (OS) and progression-free survival (PFS). ADAMTS8 was considered as an independent prognostic factor for OS. ADAMTS8 overexpression inhibited breast cancer cell proliferation, migration and invasion in vitro, and induced G2/M cell cycle arrest. ADAMTS8 was also involved in cell cycle regulation and was associated with the EGFR/Akt signaling pathway. ADAMTS8 knockdown showed the reverse effect. Together, the results showed that ADAMTS8 functioned as a tumor suppressor gene (TGS) and could be a prognostic biomarker for breast cancer.


Subject(s)
Breast Neoplasms , Humans , Female , Breast Neoplasms/pathology , Prognosis , Cell Line, Tumor , Genes, Tumor Suppressor , Signal Transduction/genetics , ADAMTS Proteins/genetics , ADAMTS Proteins/metabolism
3.
J Transl Med ; 20(1): 615, 2022 12 23.
Article in English | MEDLINE | ID: mdl-36564797

ABSTRACT

BACKGROUND: Breast cancer is a complex disease with a highly immunosuppressive tumor microenvironment, and has limited clinical response to immune checkpoint blockade (ICB) therapy. T-helper 2 (Th2) cells, an important component of the tumor microenvironment (TME), play an essential role in regulation of tumor immunity. However, the deep relationship between Th2-mediated immunity and immune evasion in breast cancer remains enigmatic. METHODS: Here, we first used bioinformatics analysis to explore the correlation between Th2 infiltration and immune landscape in breast cancer. Suplatast tosilate (IPD-1151 T, IPD), an inhibitor of Th2 function, was then employed to investigate the biological effects of Th2 blockade on tumor growth and immune microenvironment in immunocompetent murine breast cancer models. The tumor microenvironment was analyzed by flow cytometry, mass cytometry, and immunofluorescence staining. Furthermore, we examined the efficacy of IPD combination with ICB treatment by evaluating TME, tumor growth and mice survival. RESULTS: Our bioinformatics analysis suggested that higher infiltration of Th2 cells indicates a tumor immunosuppressive microenvironment in breast cancer. In three murine breast cancer models (EO771, 4T1 and EMT6), IPD significantly inhibited the IL-4 secretion by Th2 cells, promoted Th2 to Th1 switching, remodeled the immune landscape and inhibited tumor growth. Remarkably, CD8+ T cell infiltration and the cytotoxic activity of cytotoxic T lymphocyte (CTL) in tumor tissues were evidently enhanced after IPD treatment. Furthermore, increased effector CD4+ T cells and decreased myeloid-derived suppressor cells and M2-like macrophages were also demonstrated in IPD-treated tumors. Importantly, we found IPD reinforced the therapeutic response of ICB without increasing potential adverse effects. CONCLUSIONS: Our findings demonstrate that pharmaceutical inhibition of Th2 cell function improves ICB response via remodeling immune landscape of TME, which illustrates a promising combinatorial immunotherapy.


Subject(s)
Immunotherapy , Neoplasms , Animals , Mice , T-Lymphocytes, Cytotoxic , CD8-Positive T-Lymphocytes , Pharmaceutical Preparations , Tumor Microenvironment , Cell Line, Tumor
4.
Med Sci Monit ; 28: e938619, 2022 Nov 30.
Article in English | MEDLINE | ID: mdl-36447408

ABSTRACT

BACKGROUND Reproductive period for women, begins at menarche and ends at menopause, which represented the total time period of exposure to cycling reproductive hormones. The potential associations between clinicopathological features and the exposure of cycling reproductive hormones has not been extensively studied. The retrospective study enrolled 14,731 patients diagnosed with invasive breast cancer was designed to evaluate factors associated with the reproductive period on breast cancer type and patient outcomes. MATERIAL AND METHODS 14, 731 female breast cancer (BC) patients from Western China Clinical Cooperation Group (WCCCG) between January 1, 2008 to December 31, 2017 were identified. Unconditional logistic regression was performed to assess the associations between clinicopathological features and menarche age, menopause age, and reproductive years. The differences in risk factors between lower and higher number of reproductive years (<35 and ³35 yrs) were examined with the chi-square test. RESULTS First, patients with late menarche age were more likely to present with tumors of higher histological grade and larger sizes. Second, the findings suggested a higher likelihood of smaller tumor sizes in postmenopausal patients with a greater length of reproductive years. Conversely, higher histological grade was associated with this group of patients, compared to their counterparts with shorter reproductive years. Third, in luminal breast cancer, patients with a greater length of reproductive years were more probably present larger tumor. CONCLUSIONS Our findings indicated that several clinicopathologic factors including tumor size and histological grade were associated with the length of reproductive years in patients diagnosed with breast cancer.


Subject(s)
Breast Neoplasms , Humans , Female , Retrospective Studies , Menopause , Risk Factors , Hormones
5.
Biochem Biophys Res Commun ; 553: 17-24, 2021 05 14.
Article in English | MEDLINE | ID: mdl-33756341

ABSTRACT

Breast cancer (BC) threatens the life and health of women worldwide because of its high morbidity and mortality. The present study aimed to explore the biological functions and potential mechanism of BTNL9 in BC. RNA sequence and clinical data extracted from the Kaplan-Meier plotter database and The Cancer Genome Atlas (TCGA) were utilized to analyze the relationship between the expression level of BTNL9 in BC tissues and clinicopathological features and the effects of BTNL9 expression on the prognosis of BC. The diagnostic efficacy of BTNL9 expression was estimated by receiver operating characteristic (ROC) curve analysis. The mRNA and protein expression levels of BTNL9 in BC cell lines and in BC tissue were determined by quantitative real-time PCR (qPCR) and western blotting, respectively. The functions of BTNL9 were measured by colony formation, CCK-8, Transwell, flow cytometry and EdU assays. Western blotting analysis was also performed to explore the latent mechanism of BTNL9. The results showed that the expression of BTNL9 declined in BC tissues and cell lines. Low expression of BTNL9 was significantly associated with early progression of T stage, relapse-free survival (RFS), and poor overall survival (OS). Ectopic expression of BTNL9 inhibited cell proliferation, colony formation and metastasis and induced apoptosis in BC, while knockdown of BTNL9 had the opposite result. Furthermore, BTNL9 blocked BC cells in the G2/M phase via the P53/CDC25C and P53/GADD45 pathways. Our results suggest that BTNL9 may play a tumor-suppressive role in BC and has the potency to become a new biomarker for early BC diagnosis.


Subject(s)
Breast Neoplasms/metabolism , Butyrophilins/metabolism , Cell Cycle Proteins/metabolism , Cell Proliferation , Down-Regulation , Neoplasm Metastasis , Tumor Suppressor Protein p53/metabolism , cdc25 Phosphatases/metabolism , Apoptosis , Breast Neoplasms/pathology , Butyrophilins/deficiency , Butyrophilins/genetics , Cell Line, Tumor , Cell Movement , G2 Phase Cell Cycle Checkpoints , Humans , M Phase Cell Cycle Checkpoints , Neoplasm Invasiveness/pathology , Prognosis , T-Lymphocytes/immunology , Tumor Suppressor Proteins/metabolism
6.
Int J Med Sci ; 18(14): 3082-3089, 2021.
Article in English | MEDLINE | ID: mdl-34400878

ABSTRACT

Objectives: To determine the relationship between the endometrial thickness (ET) and metabolic associated fatty liver disease (MAFLD) in the postmenopausal women who have a comprehensive health examination. Methods: This was a population-based, retrospective observational study of the prevalence of MAFLD in 8594 postmenopausal women with different ET in the Quality Control Center of Health Examination in Chongqing, China. Binary and multivariable logistic regression analyses were used to obtain odds ratios and 95% confidence intervals for patients of different ET with MAFLD after adjusting for age. Results: The incidences of MAFLD were 28.6% (1352), 30.3% (1058), 34.9% (133) in postmenopausal women with ET of < 3 mm, 3 mm ≤ & < 5 mm, and ≥ 5 mm, respectively. Compared with a baseline ET of less than 5.0 mm, the risk of MAFLD in patients with ET of ≥5.0 mm is higher (OR=1.291, 95% CI: 1.041-1.603, P<0.05). After adjustment for age, a statistically significant positive correlation was still observed. The increased prevalence of MAFLD in patients with ET of 3 mm ≤ &<5 mm (OR=1.110, 95% CI: 1.008-1.223) and ≥5 mm (OR=1.383, 95% CI: 1.109-1.724) achieved statistical significance, respectively. In addition, multiple logistic analyses controlling for age also confirmed the finding of positive correlation among body mass index (BMI) and ET. Conclusion: Our results suggest that there is a positive correlation between MAFLD and ET in postmenopausal women. In addition, increased BMI is also associated with an increased risk of thickened endometrium.


Subject(s)
Endometrium/diagnostic imaging , Non-alcoholic Fatty Liver Disease/epidemiology , Postmenopause , Aged , Asymptomatic Diseases/epidemiology , Body Mass Index , China/epidemiology , Cross-Sectional Studies , Endometrium/anatomy & histology , Female , Humans , Incidence , Middle Aged , Non-alcoholic Fatty Liver Disease/diagnosis , Non-alcoholic Fatty Liver Disease/metabolism , Prevalence , Retrospective Studies , Ultrasonography
7.
Oncologist ; 25(10): e1473-e1480, 2020 10.
Article in English | MEDLINE | ID: mdl-32333626

ABSTRACT

BACKGROUND: There are limited nationwide data regarding breast cancer surgery in China. The Chinese Anti-Cancer Association's Committee of Breast Cancer Society and the Chinese Society of Breast Surgeons conducted a nationwide survey to examine the use of and barriers associated with surgical options among patients with breast cancer. METHODS: Surveys were sent via e-mail to the directors of 110 centers that performed at least 200 breast cancer operations in 2017. The electronic questionnaire contained 183 questions and covered six aspects, including demographic information about the hospitals and surgeons, surgical practice, and application of breast reconstruction. RESULTS: The selected hospitals were from 31 provinces or municipalities. The overall proportion of breast-conserving surgery (BCS) was 22%. Local gross domestic product was significantly related to the rate of BCS (p = .046). Sentinel lymph node biopsy was performed routinely in 76% of hospitals. Only 14.5% (16/110) of hospitals used the dual-tracer method, including radioisotopes. For patients with cN0 disease receiving BCS with one or two positive sentinel lymph nodes, 20% (22/110) of hospitals accepted omitting axillary lymph node dissection (ALND). For patients who underwent mastectomy, only 4% (4/110) of hospitals accepted omitting ALND. There was an obvious polarization trend in the proportion of oncoplastic breast-conserving surgery (OPS); 35/110 (32%) performed OPS in fewer than 10% of cases, whereas 36/110 (33%) performed OPS in more than 50% of cases. OPS was more likely to be performed in academic hospitals. Volume displacement was more commonly used than volume replacement (p < .001). Breast reconstruction was routinely performed in 96/110 (87%) of hospitals, 62% of which involved cooperation with the plastic surgery department. Factors influencing breast reconstruction after mastectomy included the establishment of a plastic surgery department, regional economy, and cooperation between the plastic and general surgery departments. Overall, the proportion of breast reconstruction procedures after mastectomy was 10.7%, with 70% being implant-based reconstruction, 17% autologous tissue reconstruction, and 13% a combination. Overall, 22% of the hospitals predominantly performed immediate breast reconstruction. For delayed reconstruction, two-stage implant-based breast reconstruction was the first choice for 46% of centers, whereas 20% of centers chose autologous reconstruction. Among the 96 centers that performed autologous-based reconstruction, 96% performed latissimus dorsi flap reconstruction, 65% performed transverse rectus abdominis musculocutaneous flap reconstruction, and 45% used deep inferior epigastric artery perforator flaps. CONCLUSION: The results are of great value for promoting the implementation of a consensus on diagnostic and treatment standards, development of guidelines for breast cancer, and training of breast specialists. IMPLICATIONS FOR PRACTICE: This study aimed to establish comprehensive baseline data on the status of current breast cancer treatment in China by presenting the statistics on clinical treatments and surgeries, the distribution of clinical stages, and the demographic characteristics of patients. This report is based on a survey conducted by the Chinese Anti-Cancer Association's Committee of Breast Cancer Society and the Chinese Society of Breast Surgeons, which examined the use of breast cancer surgical options in hospitals all over the country and the factors hindering the adoption of procedures and techniques. This study makes a significant contribution to the literature because there are limited nationwide data regarding breast cancer surgery in China.


Subject(s)
Breast Neoplasms , Mastectomy , Breast Neoplasms/surgery , China , Cross-Sectional Studies , Female , Hospitals , Humans
8.
Biochem Biophys Res Commun ; 526(2): 306-313, 2020 05 28.
Article in English | MEDLINE | ID: mdl-32209259

ABSTRACT

CircRNA is a kind of covalent head-to-tail looped RNA and plays an important role in tumor development. However, the identification of new potential targetable circRNAs to inhibit cancer development is still a huge challenge. In this study, we found that circEHMT1 inhibited migration and invasion of breast cancer cells. Mechanistically, we identified miR-1233-3p as a target of circEHMT1, and the circEHMT1/miR-1233-3p axis regulated matrix metalloprotease 2 (MMP2) by modulating the transcription factor Krϋppel-like factor 4 (KLF4). In summary, we showed that circEHMT1 has potential as a prognostic factor in breast cancer and played a tumor suppressor role via the circEHMT1/miR-1233-3p/KLF4/MMP2 axis.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Kruppel-Like Transcription Factors/genetics , Matrix Metalloproteinase 2/genetics , MicroRNAs/genetics , RNA, Circular/genetics , Animals , Breast Neoplasms/pathology , Cell Movement , Female , Humans , Kruppel-Like Factor 4 , Mice, Inbred BALB C , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology
9.
J Cell Physiol ; 234(8): 13906-13916, 2019 08.
Article in English | MEDLINE | ID: mdl-30618183

ABSTRACT

In this study, we found that the phospholipase C delta1 (PLCD1) protein expression is reduced in colorectal tumor tissues compared with paired surgical margin tissues. PLCD1-promoted CpG methylation was detected in 29/64 (45%) primary colorectal tumors, but not in nontumor tissues. The PLCD1 RNA expression was also reduced in three out of six cell lines, due to PLCD1 methylation. The ectopic expression of PLCD1 resulted in inhibited proliferation and attenuated migration of colorectal tumor cells, yet promoted colorectal tumor cell apoptosis in vitro. We also observed that PLCD1 suppressed proliferation and promoted apoptosis in vivo. In addition, PLCD1 induced G1/S phase cell cycle arrest. Furthermore, we found that PLCD1 led to the downregulation of several factors downstream of ß-catenin, including c-Myc and cyclin D1, which are generally known to be promoters of tumorigenesis. This downregulation was caused by an upregulation of E-cadherin in colorectal tumor cells. Our findings provide insights into the role of PLCD1 as a tumor suppressor gene in colorectal cancer (CRC), and demonstrate that it plays significant roles in proliferation, migration, invasion, cell cycle progression, and epithelial-mesenchymal transition. On the basis of these results, tumor-specific methylation of PLCD1 could be used as a novel biomarker for early detection and prognostic prediction in CRC.


Subject(s)
Colorectal Neoplasms/enzymology , Colorectal Neoplasms/pathology , Epithelial-Mesenchymal Transition , Gene Silencing , Phospholipase C delta/metabolism , Aged , Animals , Apoptosis/genetics , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Cell Movement/genetics , Clone Cells , Colorectal Neoplasms/genetics , DNA Methylation/genetics , Demethylation , Down-Regulation/genetics , Epithelial-Mesenchymal Transition/genetics , Female , G1 Phase/genetics , Gene Expression Regulation, Neoplastic , Humans , Mice, Inbred BALB C , Mice, Nude , Phospholipase C delta/genetics , Promoter Regions, Genetic , S Phase/genetics , Signal Transduction , beta Catenin/metabolism
10.
J Cell Biochem ; 120(1): 917-927, 2019 01.
Article in English | MEDLINE | ID: mdl-30187949

ABSTRACT

There are increasing reports of aberrant expression of GATA4, correlated with oncogenesis and malignant progression in some solid tumors, but whether GATA4 functions as an oncogenic driver or a tumor suppressor in carcinogenesis remains controversial. Because the role and mechanism of GATA4 in breast cancer (BrCa) remain poorly understood, we focused on the expression of GATA4 in BrCa cell lines and tissues and its mechanism in breast oncogenesis. Semiquantitative real-time polymerase chain reaction (RT-PCR), quantitative RT-PCR, Western blot analysis, and immunohistochemistry were used to detect expression of GATA4 in BrCa cell lines and adjacent breast tissues. Methylation statuses of the GATA4 promoter were studied using methylation-specific PCR in BrCa cell lines.The effects of GATA4 on proliferation, invasion, and cell cycle were also analyzed. Compared with adjacent breast tissue, GATA4 expression in BrCa tissue and cell lines was obviously lower and low expression levels of GATA4 predicted poor outcome. Methylation of GATA4 occurred in almost all of BrCa cell lines . GATA4 overexpression decreased viability, invasion, migration, and epithelial-to-mesenchymal transition of MB-231 and BT549 cells, and markedly induced cell cycle arrest and apoptosis. Exogenous expression GATA4 accompanied a significant alteration of MMP2, MMP3, E-cadherin, and N-cadherin expression and induction of the caspase-8 pathway. Moreover, GATA4 could directly repress RelA (p65) transcription, reduce the nuclear phosphorylation-p65 and upregulate inhibitor kappa B expression. Altogether, GATA4 plays a tumor-suppressive role via repression of NF-κB signaling in BrCa cells. Our findings suggest that GATA4 is a potential prognostic biomarker and gene therapeutic target for human BrCa.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , GATA4 Transcription Factor/genetics , NF-kappa B p50 Subunit/metabolism , Transcription Factor RelA/genetics , Antigens, CD/metabolism , Apoptosis/genetics , Breast Neoplasms/pathology , Cadherins/metabolism , Caspase 8/metabolism , Cell Cycle/genetics , Cell Movement/genetics , Cell Survival/genetics , Disease Progression , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 3/metabolism , Methylation , Neoplasm Invasiveness , Phosphorylation , Promoter Regions, Genetic , Transfection
11.
FASEB J ; : fj201800092R, 2018 Jun 06.
Article in English | MEDLINE | ID: mdl-29874127

ABSTRACT

Poly(ADP-ribose) polymerase (PARP)-1 may act in an error-prone pathway called alternative end joining (Alt-EJ) for DNA double-strand break (DSB) repair when nonhomologous end joining is defective. We examined the recruitment of PARP-1 to chromatin in response to radiomimetic agents and the effects of PARP-1 inhibition on DSB repair and recruitment of the meiotic recombination (MRE)-11-double-strand break repair (RAD50) protein-Nijmegen breakage syndrome (NSB)-1 (MRN) complex to the chromatin in Ku70-deficient breast cancer cells. The chromatin-binding affinity of PARP-1 was enhanced in response to neocarzinostatin (NCS) or calicheamicin treatment in the absence of Ku70. PARP-1 inhibition impaired the repair of both NCS-induced DSBs and intron-encoded endonuclease from Physarum polycephalum-induced site-specific DSB. Both fractionation and chromatin immunoprecipitation assays demonstrated that chromatin recruitment of MRN was PARP-1 dependent. These data suggest that PARP-1 is vital for DSB repair in breast cancer cells when Alt-EJ is activated.-Huang, Y., Shao, Q., Luo, X., Yang, D., Zeng, B., Xiang, T., Ren, G., Cheng, Q. Poly(ADP-ribose) polymerase-1 promotes recruitment of meiotic recombination-11 to chromatin and DNA double-strand break repair in Ku70-deficient breast cancer cells.

12.
J Cell Mol Med ; 22(2): 1257-1271, 2018 02.
Article in English | MEDLINE | ID: mdl-29193730

ABSTRACT

Chromosome region 3p12-14 is an important tumour suppressor gene (TSG) locus for multiple cancers. ADAMTS9, a member of the metalloprotease large family, has been identified as a candidate 3p14.2 TSG inactivated by aberrant promoter CpG methylation in several carcinomas, but little known about its expression and function in breast cancer. In this report, ADAMTS9 expression and methylation was analysed in breast cancer cell lines and tissue samples. ADAMTS9 RNA was significantly down-regulated in breast cancer cell lines (6/8). After treating the cells with demethylation agent Aza and TSA, ADAMTS9 expression was dramatically increased. Bisulphite genomic sequencing and methylation-specific PCR detected promoter methylation, which was associated with decreased ADAMTS9 expression. Hypermethylation was also detected in 130/219 (59.4%) of primary tumours but only in 4.5% (2/44) of paired surgical margin tissues. Ectopic expression of ADAMTS9 in tumor cells induced significant growth suppression, cell cycle arrest at the G0/G1 phase, enhanced apoptosis and reduced cell migration and invasion. Conditioned culture medium from ADAMTS9-transfected BT549 cells markedly disrupted tube formation ability of human umbilical vein endothelial cell (HUVEC) in Matrigel. Furthermore, ADAMTS9 inhibited AKT signaling and its downstream targets (MDM2, p53, p21, p27, E-cadherin, VIM, SNAIL, VEGFA, NFκB-p65 and MMP2). In addition, we demonstrated, for the first time, that ADAMTS9 inhibits AKT signaling, through suppressing its upstream activators EGFR and TGFß1/TßR(I/II) in breast cancer cells. Our results suggest that ADAMTS9 is a TSG epigenetically inactivated in breast cancer, which functions through blocking EGFR- and TGFß1/TßR(I/II)-activated AKT signaling.


Subject(s)
ADAMTS9 Protein/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Chromosomes, Human, Pair 3/metabolism , CpG Islands/genetics , DNA Methylation/genetics , Promoter Regions, Genetic , ADAMTS9 Protein/genetics , Adult , Apoptosis/genetics , Breast Neoplasms/blood supply , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Epithelial-Mesenchymal Transition/genetics , ErbB Receptors/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Invasiveness , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Transforming Growth Factor beta/metabolism , Tumor Stem Cell Assay
13.
Cell Physiol Biochem ; 51(2): 961-978, 2018.
Article in English | MEDLINE | ID: mdl-30466106

ABSTRACT

BACKGROUND/AIMS: Interferon consensus sequence-binding protein 8 (IRF8) belongs to a family of interferon (IFN) regulatory factors that modulates various important physiological processes including carcinogenesis. As reported by others and our group, IRF8 expression is silenced by DNA methylation in both human solid tumors and hematological malignancies. However, the role of IRF8 in lung carcinoma remains elusive. In this study, we determined IRF8 epigenetic regulation, biological functions, and the signaling pathway involved in non-small cell lung cancer (NSCLC). METHODS: IRF8 expression were determined by Q- PCR. MSP and A+T determined promotor methylation. MTS, clonogenic, Transwell assay, Flow cytometry, three-dimensional culture and AO/EB stain verified cell function. In vivo tumorigenesis examed the in vivo effects. By Chip-QPCR, RT-PCR, Western blot and Immunofluorescence staining, the mechanisms were studied. RESULTS: IRF8 was significantly downregulated in lung tumor tissues compared with adjacent non-cancerous tissues. Furthermore, methylation-specific PCR analyses revealed that IRF8 methylation in NSCLC was a common event, and demethylation reagent treatment proved that downregulation of IRF8 was due to its promoter CpG hypermethylation. Clinical data showed that the IRF8 methylation was associated with tumor stage, lymph node metastasis status, patient outcome, and tumor histology. Exogenous expression of IRF8 in the silenced or downregulated lung cancer cell lines A549 and H1299 at least partially restored the sensitivity of lung cancer cells to apoptosis, and arrested cells at the G0/G1 phase. Cell viability, clonogenicity, and cell migration and invasive abilities were strongly inhibited by restored expression of IRF8. A three-dimensional culture system demonstrated that IRF8 changed the cells to a more spherical phenotype. Moreover, ectopic expression of IRF8 enhanced NSCLC chemosensitivity to cisplatin. Furthermore, as verified by Chip-qPCR, immunofluorescence staining, and western blotting, IRF8 bound to the T-cell factor/lymphoid enhancer factor (TCF /LEF) promoter, thus repressing ß-catenin nuclear translocation and its activation. IRF8 significantly disrupted the effects of Wnt agonist, bml284, further suggesting its involvement in the Wnt/ß-catenin pathway. CONCLUSION: IRF8 acted as a tumor suppressor gene through the transcriptional repression of ß-catenin-TCF/LEF in NSCLC. IRF8 methylation may serve as a potential biomarker in NSCLC prognosis.


Subject(s)
Interferon Regulatory Factors/metabolism , Wnt Signaling Pathway , Aged , Animals , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Cell Movement/drug effects , Cisplatin/pharmacology , DNA Methylation , Female , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Interferon Regulatory Factors/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lymphoid Enhancer-Binding Factor 1/chemistry , Lymphoid Enhancer-Binding Factor 1/genetics , Lymphoid Enhancer-Binding Factor 1/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Neoplasm Staging , Promoter Regions, Genetic , Wnt Proteins/agonists , Wnt Proteins/metabolism , Wnt Signaling Pathway/drug effects , beta Catenin/metabolism
14.
Biochem Biophys Res Commun ; 505(1): 302-308, 2018 10 20.
Article in English | MEDLINE | ID: mdl-30249392

ABSTRACT

The abnormal expression of microRNAs is a key hallmark of breast cancer. Nevertheless, the biological roles of miR-1247-5p in breast cancer remain unknown. In this study, we revealed that miR-1247-5p expression was markedly decreased in breast cancer. It was a valuable diagnostic biomarker for breast cancer with the area under the curve of more than 0.80. Reduced miR-1247-5p expression was significantly correlated with patient age, tumor size, and poor prognosis in The Cancer Genome Atlas cohort including 839 breast cancer patients. Multivariate Cox regression analysis demonstrated that miR-1247-5p was an independent prognostic indicator for overall survival (hazard radio [HR] = 1.683, 95% confidence interval [CI] = 1.087-2.606, p = 0.020) and recurrence-free survival (HR = 2.496, 95% CI = 1.576-3.951, p < 0.001). Moreover, functional studies showed that overexpression of miR-1247-5p inhibited proliferation and induced apoptosis in breast cancer cells. Bioinformatics analysis and mechanistic investigations revealed that Dishevelled 1 (DVL1) was a direct target of miR-1247-5p. Inhibition of DVL1 by miR-1247-5p resulted in the suppression of Wnt/ß-catenin signaling, whereas overexpression of DVL1 abrogated the miR-1247-5p-mediated effect. These data reveal that miR-1247-5p, as an oncosuppressor in breast cancer, may be a promising prognostic biomarker and therapeutic target.


Subject(s)
Breast Neoplasms/genetics , Cell Proliferation/genetics , Dishevelled Proteins/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Wnt Signaling Pathway/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Dishevelled Proteins/metabolism , Down-Regulation , Female , HEK293 Cells , Humans , Kaplan-Meier Estimate , MCF-7 Cells , Middle Aged , Prognosis
15.
Biochem Biophys Res Commun ; 506(3): 685-691, 2018 11 30.
Article in English | MEDLINE | ID: mdl-30376993

ABSTRACT

BACKGROUND: Aberrant expression of interferon regulatory factor 4 (IRF4) has been reported in several hematologic malignancies. However, the prognostic significance of IRF4 expression in esophageal squamous cell carcinoma (ESCC) remains unknown. METHODS: IRF4 protein expression in ESCC tumor specimens was determined immunohistochemically. The correlation of IRF4 expression with clinico-pathological features was assessed from a cohort of 100 patients with primary ESCC. Kaplan-Meier and Cox proportional regression analyses were used to evaluate the association between IRF4 expression and patient survival. RESULTS: A Kaplan-Meier analysis indicated that patients with high IRF4 expression had a significantly longer overall survival rate than those with low IRF4 expression (p = 0.0006). Furthermore, multi-variate analyses revealed that IRF4 protein expression is an independent prognostic indicator for ESCC patients. CONCLUSION: Our results suggest that increased IRF4 protein expression correlates with improved outcome in ESCC. IRF4 may therefore represent a promising prognostic biomarker and potential immuno-therapeutic target for patients with ESCC.


Subject(s)
Esophageal Squamous Cell Carcinoma/metabolism , Interferon Regulatory Factors/metabolism , Adult , Aged , Cell Line, Tumor , Esophageal Squamous Cell Carcinoma/pathology , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Multivariate Analysis , Prognosis
16.
Cell Commun Signal ; 16(1): 32, 2018 06 18.
Article in English | MEDLINE | ID: mdl-29914512

ABSTRACT

BACKGROUND: To determine whether adipocyte-derived lipids could be transferred into breast cancer cells and investigate the underlying mechanisms of subsequent lipolysis and fatty acid trafficking in breast cancer cells. METHODS: A Transwell co-culture system was used in which human breast cancer cells were cultured in the absence or presence of differentiated murine 3 T3-L1 adipocytes. Migration/invasion and proliferation abilities were compared between breast cancer cells that were cultivated alone and those co-cultivated with mature adipocytes. The ability of lipolysis in breast cancer cells were measured, as well as the expression of the rate-limiting lipase ATGL and fatty acid transporter FABP5. ATGL and FABP5 were then ablated to investigate their impact on the aggressiveness of breast cancer cells that were surrounded by adipocytes. Further, immunohistochemistry was performed to detect differential expression of ATGL and FABP5 in breast cancer tissue sections. RESULTS: The migration and invasion abilities of cancer cells were significantly enhanced after co-culture with adipocytes, accompanied by elevated lipolysis and expression of ATGL and FABP5. Abrogation of ATGL and FABP5 sharply attenuated the malignancy of co-cultivated breast cancer cells. However, this phenomenon was not observed if a lipid emulsion was added to the culture medium to substitute for adipocytes. Furthermore, epithelial-mesenchymal transaction was induced in co-cultivated breast cancer cells. That may partially due to the stimulation of PPARß/δ and MAPK, which was resulted from upregulation of FABP5. As evidenced by immunohistochemistry, ATGL and FABP5 also had higher expression levels at the invasive front of the breast tumor, in where the adipocytes abound, compared to the central area in tissue specimens. CONCLUSIONS: Lipid originating from tumor-surrounding adipocytes could be transferred into breast cancer cells. Adipocyte-cancer cell crosstalk rather than lipids alone induced upregulation of lipases and fatty acid transport protein in cancer cells to utilize stored lipids for tumor progression. The increased expression of the key lipase ATGL and intracellular fatty acid trafficking protein FABP5 played crucial roles in this process via fueling or signaling.


Subject(s)
Adipocytes/pathology , Breast Neoplasms/pathology , Disease Progression , Fatty Acids/metabolism , Intracellular Space/metabolism , 3T3-L1 Cells , Adipocytes/metabolism , Animals , Biological Transport , Cell Communication , Fatty Acid-Binding Proteins/metabolism , Humans , Lipase/metabolism , Lipolysis , MCF-7 Cells , Mice
18.
Med Sci Monit ; 24: 574-581, 2018 Jan 29.
Article in English | MEDLINE | ID: mdl-29376515

ABSTRACT

BACKGROUND Breast cancer and its treatment provoke a series of emotional changes in patients during their breast cancer journeys. Social support is critical in helping women cope with their negative emotional responses. However, few studies have described the experiences of women with breast cancer within the Chinese context. This qualitative study explored the experiences of Chinese women with breast cancer regarding social support. MATERIAL AND METHODS This qualitative study was based on constructivism epistemology. Chinese women with breast cancer were invited to interview between June and August 2016. Purposive sampling was used, and the women were recruited until data saturation was reached (n=25). RESULTS We found that all participants expressed the importance of social support during their breast cancer journeys. The thematic framework of social support was identified, which included the following 4 interrelated themes: providers, types of support, outcomes of support, and challenges for care. CONCLUSIONS Social support is important in the health care of women living with breast cancer. Healthcare professionals should provide more individualized support and care to women during this vulnerable period.


Subject(s)
Asian People , Breast Neoplasms/psychology , Qualitative Research , Social Support , Adult , Cognition , Demography , Family , Female , Health Personnel , Humans , Middle Aged
19.
Cell Physiol Biochem ; 41(5): 1709-1724, 2017.
Article in English | MEDLINE | ID: mdl-28365691

ABSTRACT

BACKGROUND/AIMS: Aberrant activation of the Wnt/ß-catenin signaling pathway plays a key role in the pathogenesis of multiple tumors including digestive cancers. Recent studies have reported that Dickkopf-related protein 2 (DKK2) is epigenetically inactivated in numerous types of cancers and that its gene products exhibit tumor-suppressive properties. However, the biological functions and underlying molecular mechanisms of DKK2 in colon carcinoma remains obscure. METHODS: We examined the expression of DKK2 in colon tumor cell lines by RT-PCR and its promoter methylation status in colon tumor cell lines and primary tumors by methylation-specific PCR (MSP). Ectopic expression of DKK2 was measured by RT-PCR prior to the other experiments. To investigate the function of DKK2, we assayed colony formation and cell proliferation, utilized flow cytometric analyses of the cell cycle and acridine orange/ethidium bromide (AO/EB) fluorescence staining for apoptosis, and examined wound healing, transwell migration and tumor growth in vivo. Western blots were used to explore the mechanisms of DKK2 in epithelial- mesenchymal transition and canonical Wnt/ß-catenin signaling. RESULTS: We show here that downregulation or silencing of DKK2 was closely associated with the hypermethylation status of its promoter and that DKK2 expression could be restored by demethylation treatment. Methylation of the DKK2 promoter was detected in nearly all tumors and tumor-adjacent tissues, but not in normal colon tissues. Ectopic expression of DKK2 in colon cell lines HCT116 and HT-29 inhibited colony formation and cell viability by inducing cell cycle G0/G1 arrest and apoptosis, and growth of stable DKK2-infected HCT116 cells in nude mice was decreased compared to controls. Furthermore, DKK2 restrained cell migration through partial reversal of epithelial-to- mesenchymal transition and also by downregulating several stem cell markers. Our data further showed that restoration of DKK2 expression resulted in downregulation of active ß-catenin and its downstream target genes. CONCLUSION: DKK2 appears to be a functional tumor suppressor regulating tumorigenesis of colorectal cancer by antagonizing Wnt/ß-catenin signaling.


Subject(s)
Colorectal Neoplasms/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Intercellular Signaling Peptides and Proteins/biosynthesis , Neoplasm Proteins/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism , Animals , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Female , Humans , Intercellular Signaling Peptides and Proteins/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Metastasis , Neoplasm Proteins/genetics , beta Catenin/genetics
20.
Cell Physiol Biochem ; 44(4): 1370-1380, 2017.
Article in English | MEDLINE | ID: mdl-29186710

ABSTRACT

BACKGROUND/AIMS: ADAMTS (disintegrin-like and metalloproteinase with thrombospondin motifs) proteins are extracellular zinc metalloproteinases that play an important role in extracellular matrix assembly and degradation, connective tissue structuring, angiogenesis, and cell migration. Multiple studies suggest that ADAMTS proteins (e.g. ADAMTS9) can act as tumor suppressors. In gastric, esophageal, and nasopharyngeal carcinomas ADAMTS9 is frequently down-regulated by promoter methylation. Whether ADAMTS9 can function as a tumor suppressor gene (TSG) in colorectal cancer is still unclear. METHODS: We performed immunohistochemistry, RT-PCR, and qRT-PCR, to examine the expression of ADAMTS9 in colorectal cancer cell lines and primary colorectal cancer tissues. Methylation-specific PCR was also carried out to investigate the promoter methylation status of ADAMTS9. We also explored the functions of ADAMTS9 in colorectal cancer cell lines through in vitro experiments. RESULTS: ADAMTS9 expression was down-requlated or silenced in 83.3% (5/6) of colorectal cancer cell lines, and frequently repressed in 65.6% (21/32) of colorectal cancer tissues. Down-regulation of ADAMTS9 was partially due to promoter methylation. Exogenous expression of ADAMTS9 in colorectal cancer cell lines inhibited cell proliferation and migration through the regulation of cell cycle and apoptosis. In addition, ADAMTS9 prevented the activation of Akt, and its downstream targets in colorectal cancer cell lines. CONCLUSION: Our findings suggest ADAMTS9 is a TSG in colorectal cancer.


Subject(s)
ADAMTS9 Protein/metabolism , Colorectal Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , Tumor Suppressor Protein p53/metabolism , ADAMTS9 Protein/antagonists & inhibitors , ADAMTS9 Protein/genetics , Apoptosis , Cell Line, Tumor , Cell Movement , Cell Proliferation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , CpG Islands , DNA Methylation , Down-Regulation , Epithelial-Mesenchymal Transition , G1 Phase Cell Cycle Checkpoints , Genetic Vectors/genetics , Genetic Vectors/metabolism , HCT116 Cells , Humans , Immunohistochemistry , Promoter Regions, Genetic , RNA Interference , RNA, Small Interfering/metabolism , Real-Time Polymerase Chain Reaction , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL