Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters











Publication year range
1.
Ren Fail ; 45(2): 2253930, 2023.
Article in English | MEDLINE | ID: mdl-37724535

ABSTRACT

BACKGROUND: Cyclophosphamide (CTX) and calcineurin inhibitors (CNIs) based regimens are recommended as immunosuppressive therapies for patients with idiopathic membranous nephropathy (IMN). Focal and segmental glomerular sclerosis (FSGS) lesions, which are common in membranous nephropathy (MN), are poor predictors of outcome. This study compared the differences of prognosis between two regimens in patients with IMN combined with FSGS lesions. METHODS: This retrospective study enrolled 108 patients with biopsy-proven IMN, accompanied with FSGS lesions, nephrotic syndrome and an estimated glomerular filtration rate (eGFR)≥60 mL/min/1.73 m2 who were treated with CTX or CNIs. We used propensity score matching (PSM) for balancing the confounding variables. RESULTS: During follow-up, 10 patients (10/55 [18.2%]; nine males) in the CNIs group showed a 50% decline in eGFR; eight had a not otherwise specified variant. Patients initially treated with CNIs had a significantly higher risk of progression to the primary outcome and a lower probability of complete or total remission. The relapse rate was higher in patients who initially received CNIs- than in those who received CTX-based treatment. Before PSM, age and 24-h urine protein level differed significantly between the groups. The PSM model included data from 72 patients. Worse outcomes were also noted among patients who initially received CNIs than those who received CTX-based treatments after matching. CONCLUSIONS: Patients with MN combined with FSGS lesions have a higher risk of renal functional decline and a higher rate of relapse after CNIs than after CTX therapy.


Subject(s)
Glomerulonephritis, Membranous , Glomerulosclerosis, Focal Segmental , Male , Humans , Young Adult , Adult , Glomerulonephritis, Membranous/complications , Glomerulonephritis, Membranous/drug therapy , Glomerulosclerosis, Focal Segmental/complications , Glomerulosclerosis, Focal Segmental/drug therapy , Retrospective Studies , Calcineurin Inhibitors/therapeutic use , Cyclophosphamide/therapeutic use , China
2.
Biometals ; 36(3): 549-562, 2023 06.
Article in English | MEDLINE | ID: mdl-35650365

ABSTRACT

Lactoferrin (Lf), a multiple functional natural immune protein, is widely distributed in mammalian milk and glandular secretions (bile, saliva, tears and nasal mucosal secretions, etc.). In the previous study, we found that Lf plays an anti-inflammatory and anti-tumorigenesis role in AOM/DSS (azoxymethane/dextran sulfate sodium) induced mouse colitis-associated colon cancer model. Although we found that Lf has anti-inflammatory effects in chronic inflammation, its specific role and mechanisms in acute inflammation have not been clarified. Here, we reported that the expression levels of Lf were significantly increased when the organism was infected by Gram-negative bacteria. We then explored the role and potential mechanism of Lf in lipopolysaccharide (LPS)-induced acute inflammation. In the LPS-induced acute abdominal inflammation model, Lf deficiency aggravated inflammatory response and promoted macrophage chemotaxis to the inflammation site. Lf inhibited macrophage chemotaxis by suppressing the expression of macrophage-associated chemokines Ccl2 and Ccl5. Highly activated NF-κB signaling in Lf-/- mice was responsible for the high expression of Ccl2 and Ccl5. Our results suggested that the anti-inflammatory effect of Lf offers a new potential treatment for acute inflammatory diseases.


Subject(s)
Inflammation , Lactoferrin , Animals , Mice , Inflammation/immunology , Inflammation/metabolism , Lactoferrin/deficiency , Lactoferrin/genetics , Lipopolysaccharides/pharmacology , Macrophages/metabolism , NF-kappa B/metabolism , NF-kappa B/pharmacology
3.
J Hematol Oncol ; 14(1): 58, 2021 04 07.
Article in English | MEDLINE | ID: mdl-33827645

ABSTRACT

Lactoferrin (Lf) is widely distributed in mammalian milk, various tissues, and their exocrine fluids and has many physiological functions, such as bacteriostasis, antivirus, and immunoregulation. Here, we provide evidence that lactoferrin is required for early stages of B cell development in mice. Lactoferrin-deficient (Lf-/-) C57BL/6 mice showed systematic reduction in total B cells, which was attributed to the arrest of early B cell development from pre-pro-B to pro-B stage. Although the Lf-/- B cell "seeds" generated greater pro-B cells comparing to wild type (WT) littermates, the Lf-/- mice bone marrow had less stromal cells, and lower CXCL12 expression, produced a less favorable "microenvironment" for early B cell development. The underlying mechanism was mediated through ERK and AKT signalings and an abnormality in the transcription factors related to early differentiation of B cells. The Lf-/- mice also displayed abnormal antibody production in T cell-dependent and T cell-independent immunization experiments. In a pristane-induced lupus model, Lf-/- mice had more serious symptoms than WT mice, whereas lactoferrin treatment alleviated these symptoms. This study demonstrates a novel role of lactoferrin in early B cell development, suggesting a potential benefit for using lactoferrin in B cell-related diseases.


Subject(s)
Anti-Infective Agents/therapeutic use , B-Lymphocytes/metabolism , Lactoferrin/therapeutic use , Animals , Humans , Mice , Mice, Inbred C57BL
4.
Transl Oncol ; 14(5): 101049, 2021 May.
Article in English | MEDLINE | ID: mdl-33676177

ABSTRACT

BACKGROUND: The low prevalence of the BRAF V600E mutation in colorectal cancers (CRCs) in Chinese populations has stimulated concern about the efficacy of BRAF mutation analysis for Lynch syndrome (LS) screening. METHODS: In total, 169 of 4104 consecutive CRC patients with absent MLH1 staining were analyzed to compare the utility of the BRAF V600E mutation testing with MLH1 promoter methylation analysis in the Chinese population. Germline genetic testing was performed in patients with wild-type BRAF/methylated MLH1. RESULTS: Compared with BRAF genotyping, the use of MLH1 methylation testing alone to evaluate patients with MLH1 deficiency reduced referral rates for germline testing by 1.8-fold (82.8% vs. 47.1%). However, 6 patients harboring MLH1 promoter methylation were verified to have LS through germline genetic testing. It is notable that all 6 patients had a family history of CRC in at least 1 first-degree relative (FDR) or second-degree relative (SDR). The combination of MLH1 promoter methylation analysis and a family history of CRC could preclude significantly more patients from germline genetic testing than from BRAF mutation testing alone (45.5% vs. 17.2%, p<0.001) and decrease the number of misdiagnosed LS patients with MLH1 promoter methylation. CONCLUSION: The combination of a family history of CRC with MLH1 promoter methylation analysis showed better performance than BRAF mutation testing in the selection of patients in the Chinese population for germline genetic testing.

5.
J Clin Pathol ; 74(4): 223-227, 2021 Apr.
Article in English | MEDLINE | ID: mdl-32220941

ABSTRACT

AIMS: The aim of this study is to investigate the expression profiles of cell cycle related proteins in nasal extranodal NK/T cell lymphoma, nasal type (ENKTCL). METHODS: The expression profiles of cell cycle related proteins were assessed with a cell cycle antibody array and validated by immunohistochemistry. Correlations between the expression levels of proteins and clinical outcomes of patients with nasal ENKTCL were evaluated. RESULTS: The expression of full length ataxia telangiectasia mutated (ATM) in nasal ENKTCL significantly decreased compared with that in nasal benign lymphoid proliferative disease (NBLPD), but the expression levels of p-ATM, CHK2 and RAD51 significantly increased in nasal ENKTCL compared with that in NBLPD. Kaplan-Meier analysis showed that the expression levels of p-ATM and CHK2 in nasal ENKTCL were inversely related to overall survival (p=0.011 and p=0.025, respectively). CONCLUSION: Abnormalities in the ATM pathway may play a crucial role in the oncogenesis and chemoradiotherapy resistance of nasal ENKTCL.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/analysis , Biomarkers, Tumor/analysis , Checkpoint Kinase 2/analysis , Lymphoma, Extranodal NK-T-Cell/enzymology , Cell Proliferation , Drug Resistance, Neoplasm , Humans , Immunohistochemistry , Lymphoma, Extranodal NK-T-Cell/mortality , Lymphoma, Extranodal NK-T-Cell/pathology , Lymphoma, Extranodal NK-T-Cell/therapy , Neoplasm Grading , Phosphorylation , Rad51 Recombinase/analysis , Radiation Tolerance , Risk Factors , Time Factors , Treatment Outcome , Up-Regulation
6.
J Int Med Res ; 48(7): 300060520936879, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32721244

ABSTRACT

OBJECTIVE: Hepatocellular carcinoma (HCC) is one of the most common causes of cancer-related deaths worldwide. This study investigated the relationship between cyclin-dependent kinase inhibitor (CDKN)3 and prognosis and pathological characteristics in HCC patients to determine whether it could be used as a prognostic factor and/or therapeutic target for HCC drug development. METHODS: We previously showed that CDKN3 is deregulated in HCC tumor samples. Here, bioinformatics analysis was used to assess the relationship between CDKN3 gene expression and the characteristics of HCC patients from Gene Expression Omnibus and The Cancer Genome Atlas databases. Additionally, CDKN3 expression was silenced by small interfering RNA to determine its effect on HCC cell proliferation and on HCC cell sensitivity to adriamycin chemotherapy. RESULTS: Bioinformatics analysis showed a negative correlation between CDKN3 expression and both disease-free survival and overall survival. CDKN3 silencing did not significantly suppress the proliferation of HCC cells, but did decrease their sensitivity to adriamycin. CONCLUSIONS: CDKN3 may have a dual role during the development of HCC, and could be used as an independent prognostic factor and therapeutic target for HCC treatment.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Cyclin-Dependent Kinase Inhibitor Proteins , Doxorubicin/pharmacology , Dual-Specificity Phosphatases , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Prognosis , RNA, Small Interfering/genetics
7.
Int J Cancer ; 146(11): 3077-3086, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32030746

ABSTRACT

Lynch syndrome (LS) is the most common hereditary colorectal cancer (CRC) predisposition syndrome. We performed a large-scale study to assess a screening strategy for identifying LS in Chinese CRC patients in routine clinical testing. A total of 4,195 eligible CRCs were universally screened. Then, 8.7% of CRCs were detected with dMMR. The incidence of LS was 2.7% (115 of 4,195) in this cohort; among patients over 70 years of age, only 0.3% (2 of 678) were diagnosed as LS. Then, 17.4% of LS cases showed large genomic deletions/duplications. LS probands developed CRCs predominantly at proximal colon location. The frequency of BRAF V600E mutation among Chinese CRCs was significantly lower than that among Western populations, and MLH1 promoter methylation significantly improved the efficiency of genetic screening for LS among MLH1-deficient patients. A comprehensive molecular testing strategy that includes detection of large genomic rearrangements is imperative for the diagnosis of LS. Among CRC patients aged 70 years or younger, a selective strategy for LS screening might be considered for routine clinical testing.


Subject(s)
Colorectal Neoplasms, Hereditary Nonpolyposis/epidemiology , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , MutL Protein Homolog 1/genetics , Proto-Oncogene Proteins B-raf/genetics , Adult , China/epidemiology , Colorectal Neoplasms, Hereditary Nonpolyposis/diagnosis , DNA Methylation/genetics , Evidence-Based Medicine/methods , Female , Genetic Testing/methods , Humans , Male , Mass Screening/methods , Middle Aged , Retrospective Studies
8.
J Cutan Pathol ; 47(4): 387-389, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31658378

ABSTRACT

Primary cutaneous EBV (Epstein-Barr virus)-positive diffuse large B-cell lymphoma, not otherwise specified (DLBCL, NOS), is an extremely rare disease. The clinical and pathological features of DLBCL, NOS have not been clearly illustrated. We report a case of primary cutaneous EBV-positive DLBCL, NOS in a 35-year-old Chinese male with multiple ulcerated and nodular lesions on his trunk and arms for 6 months. No other organs, except the skin, were involved. The lesions were localized in the dermis with focal necrosis. The tumor cells were immunoblastic- or centroblastic-like cells and diffusely distributed. There were numerous inflammatory cells in the background. The tumor cells were positive for CD20, PAX-5, MUM1, LMP1, CD30, and Epstein-Barr virus encodedsmall RNA, but negative for EBNA2. Polymerase chain reaction showed a monoclonal IG gene rearrangement. The patient received 6 cycles of CHOP (cyclophosphamide, hydroxydaunorubicin, Oncovin®, prednisone) chemotherapy and went into complete remission. There was no evidence of relapse after 35 months of follow-up.


Subject(s)
Epstein-Barr Virus Infections , Herpesvirus 4, Human/metabolism , Lymphoma, Large B-Cell, Diffuse , Skin Neoplasms , Transcription Factor CHOP/administration & dosage , Adult , Epstein-Barr Virus Infections/drug therapy , Epstein-Barr Virus Infections/metabolism , Epstein-Barr Virus Infections/pathology , Epstein-Barr Virus Nuclear Antigens/metabolism , Humans , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Large B-Cell, Diffuse/pathology , Lymphoma, Large B-Cell, Diffuse/virology , Male , Neoplasm Proteins/metabolism , Skin Neoplasms/drug therapy , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Skin Neoplasms/virology , Viral Proteins/metabolism
9.
Oncogene ; 39(1): 122-135, 2020 01.
Article in English | MEDLINE | ID: mdl-31462711

ABSTRACT

Lactoferrin, an innate immunity molecule, is involved in anti-inflammatory, anti-microbial, and anti-tumor activities. We previously reported that lactoferrin is downregulated in specimens of nasopharyngeal carcinoma and negatively associated with tumor progression and metastasis of patients with nasopharyngeal carcinoma. However, the relationship between lactoferrin and the pro-metastatic microenvironment has not been reported yet. Here, by using the lactoferrin knockout mouse, we found that lactoferrin deficiency facilitated melanoma cells metastasizing to lungs, through recruiting myeloid-derived suppressor cells (MDSCs) in the lungs. Mechanistic studies showed that in the lung microenvironment of the lactoferrin knockout mice, the TLR9 signaling was the most repressed signaling. Lactoferrin can induce MDSCs differentiation and apoptosis, as well as upregulate TLR9 expression. TLR9 agonist or lactoferrin treatment can rescue this phenotype in the tumor metastasis mouse model. Our results suggest a protective role of lactoferrin in cancer metastasis, along with a deficiency in certain components of the innate immune system, may lead to a pro-metastatic tumor microenvironment.


Subject(s)
Lactoferrin/genetics , Melanoma, Experimental/genetics , Myeloid-Derived Suppressor Cells/metabolism , Toll-Like Receptor 9/genetics , Animals , Apoptosis/genetics , Cell Differentiation/genetics , Cell Line, Tumor , Disease Models, Animal , Gene Expression Regulation, Neoplastic/genetics , Heterografts , Humans , Immunity, Innate/genetics , Lactoferrin/deficiency , Lactoferrin/pharmacology , Lung/metabolism , Lung/pathology , Melanoma, Experimental/pathology , Mice , Mice, Knockout , Myeloid-Derived Suppressor Cells/pathology , Neoplasm Metastasis , Signal Transduction/genetics , Toll-Like Receptor 9/agonists , Tumor Microenvironment/genetics
10.
BMC Nephrol ; 20(1): 328, 2019 08 22.
Article in English | MEDLINE | ID: mdl-31438882

ABSTRACT

BACKGROUND: Focal segmental lesions (FSLs) are not uncommon in idiopathic membranous nephropathy (IMN). The reported percentage of IMN patients with focal segmental glomerulosclerosis (FSGS) lesions varies widely between studies. The objective of this study was to differentiate atypical FSL (aFSL) from typical FSGS in IMN and to analyse the clinicopathological predictors of primary outcome of IMN patients. METHODS: A total of 716 patients with biopsy-proven IMN between January 1, 2007 and December 31, 2017 were enrolled in the study. An atypical focal segmental lesion was defined as pure synechia, segmental hyperplasia of podocytes or thickening of the GBM accompanied by proliferation of the mesangial matrix, and absence of typical FSGS. The patients were divided into three groups: patients without FSL (FSL-), patients with typical FSGS (FSGS+), and patients with aFSL (aFSL+).The primary outcome was a 50% decline in the initial estimated glomerular filtration rate or end-stage renal disease (ESRD) incidence. Secondary outcomes included all-cause death and ESRD. RESULTS: FSGS was present in 174 patients, while aFSL was noted in 161 patients. Systolic blood pressure was higher in both aFSL+ group and FSGS+ groups compared with the FSL- group. IMN patients without FSL and with aFSL had lower serum creatinine levels than IMN patients with FSGS. Both the FSGS+ and aFSL+ groups had higher levels of proteinuria and lower albumin levels than the FSL- group. Renal tissue lesions, including tubulointerstitial fibrosis, glomerular obsolescence, and vascular sclerosis were significantly more severe in the FSGS+ group. Cox multivariate analysis showed that older age ≥ 60 years, eGFR< 60 ml/(min·1.73m2), tubulointerstitial fibrosis area ≥ 15% and FSGS at biopsy were independent risk factors for the primary outcome. CONCLUSIONS: No significant difference in outcome was found between the FSL- and aFSL+ groups, although the patients with aFSL had lower levels of serum albumin and eGFR, higher level of urinary protein, more severe renal lesions with proliferation of the mesangial area,tubulointerstitial fibrosis and vascular sclerosis. FSGS, excluding atypical lesions, was an independent predictor of the primary outcome.


Subject(s)
Glomerulonephritis, Membranous/pathology , Glomerulosclerosis, Focal Segmental/pathology , Biopsy , Cause of Death , Diagnosis, Differential , Female , Follow-Up Studies , Glomerular Filtration Rate , Glomerulonephritis, Membranous/physiopathology , Glomerulosclerosis, Focal Segmental/physiopathology , Humans , Kidney/pathology , Kidney Failure, Chronic/diagnosis , Kidney Failure, Chronic/etiology , Kidney Failure, Chronic/therapy , Male , Middle Aged , Proportional Hazards Models , Retrospective Studies
11.
J Innate Immun ; 9(6): 574-586, 2017.
Article in English | MEDLINE | ID: mdl-28877527

ABSTRACT

Recognition of viral pathogen-associated molecular patterns by pattern recognition receptors (PRRs) is the first step in the initiation of a host innate immune response. As a PRR, RIG-I detects either viral RNA or replication transcripts. Avoiding RIG-I recognition is a strategy employed by viruses for immune evasion. Epstein-Barr virus (EBV) infects the majority of the human population worldwide. During the latent infection period there are only a few EBV proteins expressed, whereas EBV-encoded microRNAs, such as BART microRNAs, are highly expressed. BART microRNAs regulate both EBV and the host's gene expression, modulating virus proliferation and the immune response. Here, through gene expression profiling, we found that EBV miR-BART6-3ps inhibited genes of RIG-I-like receptor signaling and the type I interferon (IFN) response. We demonstrated that miR-BART6-3p rather than other BARTs specifically suppressed RIG-I-like receptor signaling-mediated IFN-ß production. RNA-seq was used to analyze the global transcriptome change upon EBV infection and miR-BART6-3p mimics transfection, which revealed that EBV infection-triggered immune response signaling can be repressed by miR-BART6-3p overexpression. Furthermore, miR-BART6-3p inhibited the EBV-triggered IFN-ß response and facilitated EBV infection through targeting the 3'UTR of RIG-I mRNA. These findings provide new insights into the mechanism underlying the strategies employed by EBV to evade immune surveillance.


Subject(s)
Epstein-Barr Virus Infections/immunology , Herpesvirus 4, Human/genetics , Kidney Tubules, Proximal/virology , MicroRNAs/genetics , RNA, Viral/genetics , Cell Line , Host-Pathogen Interactions , Humans , Immune Evasion , Immunity, Innate , Kidney Tubules, Proximal/immunology , Pathogen-Associated Molecular Pattern Molecules/immunology , Receptors, Pattern Recognition/metabolism , Receptors, Retinoic Acid/metabolism , Signal Transduction
12.
Front Immunol ; 8: 1774, 2017.
Article in English | MEDLINE | ID: mdl-29326691

ABSTRACT

The association between chronic inflammation and cancer has long been recognized. The inflammatory bowel disease ulcerative colitis frequently progresses to colon cancer; however, the underlying mechanism is still unclear. S100a9 has been emerged as an important pro-inflammatory mediator in acute and chronic inflammation, and the aberrant expression of S100a9 also contributes to tumorigenic processes such as cell proliferation, angiogenesis, metastasis, and immune evasion. We previously revealed that S100a8 and S100a9 are highly activated and play an important role in the process of colitis-associated carcinogenesis, which suggests an attractive therapeutic target for ulcerative colitis and related colon cancer. Here, we report that administration of a neutralizing anti-S100a9 antibody significantly ameliorated dextran sulfate sodium (DSS)-induced colitis and accompanied by diminished cellular infiltrate of innate immunity cells (macrophages, neutrophils, and dendritic cells) and production of pro-inflammatory cytokines (Tnfα, Il1ß, Ifnγ, Il6, Il17a, Il23a, Il4, and Il12a). The protective effect of anti-S100a9 antibody treatment was also observed in azoxymethane (AOM)/DSS-induced colitis-associated cancer (CAC) mouse model. The inflammatory response, tumor cell proliferation, and immune cells infiltration in the colon tissues were suppressed by anti-S100a9 antibody. Gene expression profiling showed that key pathways known to be involved in CAC development, such as Wnt signaling pathway, PI3K-Akt signaling pathway, cytokine-cytokine receptor interaction, and ECM-receptor interaction pathway, were suppressed after treatment with anti-S100a9 antibody in CAC mice. In view of the protective effect of neutralizing anti-S100a9 antibody against DSS-induced colitis and AOM/DSS-induced CAC in mouse model, this study suggests that anti-S100a9 antibody may provide a novel therapeutic approach to treat ulcerative colitis and may decrease the risk for developing CAC.

13.
Oncotarget ; 7(22): 32421-32, 2016 May 31.
Article in English | MEDLINE | ID: mdl-27107415

ABSTRACT

We previously reported that miR-1207-5p can inhibit epithelial-mesenchymal transition (EMT) induced by growth factors such as EGF and TGF-ß, but the exact mechanism is unclear. Here we identified that Colony stimulating factor 1 (CSF1) is a target gene of miR-1207-5p. CSF1 controls the production, differentiation and function of macrophage and promotes the release of proinflammatory chemokines. We showed that miR-1207-5p inhibited lung cancer cell A549 proliferation, migration and invasion in vitro, and suppressed the STAT3 and AKT signalings. miR-1207-5p overexpression can increase HUVEC angiogenesis, and can modulate the M2 phenotype of macrophage. miR-1207-5p also significantly inhibited A549 cells metastasis in a nude mouse xenograft model. miR-1207-5p and CSF1 expression levels and their relationship with lung cancer survival and metastasis status were assayed by means of a lung cancer tissue microarray. Macrophage is an essential part of the tumor microenvironment, thus the miR-1207-5p-CSF1 axis maybe a new regulator of lung cancer development through modulating the tumor microenvironment.


Subject(s)
Lung Neoplasms/genetics , Lung Neoplasms/therapy , Macrophage Colony-Stimulating Factor/genetics , MicroRNAs/genetics , A549 Cells , Animals , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Female , HEK293 Cells , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Macrophage Colony-Stimulating Factor/biosynthesis , Male , Mice , Mice, Nude , MicroRNAs/biosynthesis , Middle Aged , Neoplasm Metastasis , Tissue Array Analysis , Transfection , Tumor Microenvironment , Xenograft Model Antitumor Assays
14.
J Cell Physiol ; 231(1): 120-9, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26032086

ABSTRACT

As regulators in gene expression, microRNAs take part in most biological processes including cell differentiation, apoptosis, cell cycle, and epithelial-to-mesenchymal transition (EMT). In order to evaluate their roles in EMT process, microRNA expression profile changes induced by EGF or TGF-ß treatment on nasopharyngeal carcinoma cell HK-1 were analyzed, and miR-21, miR-148a, miR-505, and miR-1207-5p were found to be upregulated in growth factors-induced EMT process. miR-21 is already known as an oncogenic miRNA to promote metastasis, however, the exact functions of other three miRNAs in EMT are unclear. To our surprise, we found that miR-148a, miR-505, and miR-1207-5p can suppress EMT and metastasis phenotypes in HK-1 cells both in vitro and in vivo, which may relate to their inhibition on EMT and Wnt signaling molecules. MiRNAs confer robustness to biological processes by posttranscriptional repression of key transcriptional programs that are related to previous developmental stages or to alternative cell fates. Our findings indicate that miRNA feedback circuit is tuned to respond to growth factors-induced EMT, and we suggested a new negative feedback loop which may be an important element of the EMT process and confer biological robustness.


Subject(s)
Cell Differentiation/genetics , Epidermal Growth Factor/metabolism , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition/genetics , MicroRNAs/genetics , Transforming Growth Factor beta/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/genetics , Humans , Signal Transduction/genetics
15.
Int J Cancer ; 137(12): 2803-14, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26135667

ABSTRACT

The aberrant expression of S100A8 and S100A9 is linked to nonresolving inflammation and ultimately to carcinogenesis, whereas the underlying mechanism that allows inflammation to progress to specific cancer types remains unknown. Here, we report that S100A8 was induced by inflammation and then promoted colorectal tumorigenesis downstream by activating Id3 (inhibitor of differentiation 3). Using gene expression profiling and immunohistochemistry, we found that both S100A8 and S100A9 were upregulated in the chemically-induced colitis-associated cancer mouse model and in human colorectal cancer specimens. Furthermore, we showed that S100A8 and S100A9 acted as chemoattractant proteins by recruiting macrophages, promoting the proliferation and invasion of colon cancer cell, as well as spurring the cycle that culminates in the acceleration of cancer metastasis in a nude mouse model. S100A8 regulated colon cancer cell cycle and proliferation by inducing Id3 expression while inhibiting p21. Id3 expression was regulated by Smad5, which was directly phosphorylated by Akt1. Our study revealed a novel mechanism in which inflammation-induced S100A8 promoted colorectal tumorigenesis by acting upstream to activate the Akt1-Smad5-Id3 axis.


Subject(s)
Adenocarcinoma/metabolism , Calgranulin A/physiology , Colitis/metabolism , Colorectal Neoplasms/metabolism , Inhibitor of Differentiation Proteins/metabolism , Lung Neoplasms/metabolism , Adenocarcinoma/immunology , Adenocarcinoma/secondary , Animals , Calgranulin B/metabolism , Carcinogenesis/immunology , Carcinogenesis/metabolism , Cell Cycle , Cell Line, Tumor , Cell Movement , Cell Proliferation , Colitis/immunology , Colitis/pathology , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/immunology , Lung Neoplasms/secondary , Male , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Neoplasm Transplantation , Transcriptome
16.
Oncotarget ; 6(7): 5412-25, 2015 Mar 10.
Article in English | MEDLINE | ID: mdl-25742789

ABSTRACT

Emerging evidence has implicated microRNAs in regulating the production of multiple inflammatory mediators including cytokines and chemokines. We previously elucidated the dynamic activation of key signals that link colitis to colorectal cancer. In this study, we observed a sharp increase in the levels of matrix metalloproteinases (Mmps) that provided a basis for the inflammation-cancer link, and we questioned whether this was a consequence of the dysregulation of Mmp-specific microRNAs, at least partly. We assayed a panel of murine microRNAs that were predicted to target Mmps and found they were downregulated in the inflammation-cancer link. Furthermore, we demonstrated that three murine microRNAs, namely miR-128, -134, and -330, can target the three Mmps Mmp3, Mmp10, and Mmp13, respectively. We also found that the level of the microRNA-processing enzyme Dicer1 was decreased in the inflammation-cancer link. These microRNAs functioned as tumor suppressors in colon cancer cells, attenuating the proliferation, migration, and invasion potential of murine colon cancer cells as well as angiogenesis and the growth of tumors derived from these cells. Our results suggest that microRNAs modulate the production of key inflammatory mediators and that microRNA dysfunction may contribute to the non-resolving inflammation associated with cancer.


Subject(s)
Colitis/chemically induced , Colonic Neoplasms/enzymology , Colonic Neoplasms/etiology , Lipopolysaccharides/toxicity , Matrix Metalloproteinases/metabolism , MicroRNAs/genetics , Neovascularization, Pathologic , Animals , Apoptosis , Blotting, Western , Cell Movement , Cell Proliferation , Cells, Cultured , Colitis/enzymology , Colitis/pathology , Colonic Neoplasms/pathology , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Humans , Immunoenzyme Techniques , Male , Matrix Metalloproteinases/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation , Xenograft Model Antitumor Assays
17.
Lab Invest ; 94(11): 1188-99, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25068657

ABSTRACT

Epstein-Barr virus (EBV) infection contributes to tumorigenesis of various human malignancies including nasopharyngeal carcinoma (NPC). EBV triggers innate immune and inflammatory responses partly through Toll-like receptor (TLR) signaling. Lactoferrin (LF), with its anti-inflammatory properties, is an important component of the innate immune system. We previously reported that LF protects human B lymphocytes from EBV infection by its ability to bind to the EBV receptor CD21, but whether LF can suppress EBV-induced inflammation is unclear. Here, we report that LF reduced synthesis of IL-8 and monocyte chemoattractant protein-1 (MCP-1) induced by EBV in macrophages via its suppression of NF-κB activity. LF interacted with TLR2 and interfered with EBV-triggered TLR2-NF-κB activation. LF inhibited the ability of TLR9 to recognize dsDNA by binding to its co-receptor CD14, which blocked the interaction between CD14 and TLR9. EBV-induced inflammation was thus aggravated in the presence of CD14. In addition, LF expression levels were significantly downregulated in NPC specimens, and correlated inversely with IL-8 and MCP-1 expression. These findings suggest that LF may suppress the EBV-induced inflammatory response through interfering with the activation of TLR2 and TLR9.


Subject(s)
Herpesvirus 4, Human/immunology , Lactoferrin/physiology , Macrophages/physiology , Toll-Like Receptor 2/immunology , Toll-Like Receptor 9/immunology , Chemokine CCL2/metabolism , Epstein-Barr Virus Infections/immunology , Female , HEK293 Cells , Host-Pathogen Interactions , Humans , Interleukin-8/metabolism , Lipopolysaccharide Receptors/metabolism , Male , Middle Aged , NF-kappa B/metabolism , Up-Regulation
18.
PLoS One ; 9(7): e103298, 2014.
Article in English | MEDLINE | ID: mdl-25057912

ABSTRACT

Nonresolving inflammatory processes affect all stages of carcinogenesis. Lactoferrin, a member of the transferrin family, is involved in the innate immune response and anti-inflammatory, anti-microbial, and anti-tumor activities. We previously found that lactoferrin is significantly down-regulated in specimens of nasopharyngeal carcinoma (NPC) and negatively associated with tumor progression, metastasis, and prognosis of patients with NPC. Additionally, lactoferrin expression levels are decreased in colorectal cancer as compared with normal tissue. Lactoferrin levels are also increased in the various phases of inflammation and dysplasia in an azoxymethane-dextran sulfate sodium (AOM-DSS) model of colitis-associated colon cancer (CAC). We thus hypothesized that the anti-inflammatory function of lactoferrin may contribute to its anti-tumor activity. Here we generated a new Lactoferrin knockout mouse model in which the mice are fertile, develop normally, and display no gross morphological abnormalities. We then challenged these mice with chemically induced intestinal inflammation to investigate the role of lactoferrin in inflammation and cancer development. Lactoferrin knockout mice demonstrated a great susceptibility to inflammation-induced colorectal dysplasia, and this characteristic may be related to inhibition of NF-κB and AKT/mTOR signaling as well as regulation of cell apoptosis and proliferation. Our results suggest that the protective roles of lactoferrin in colorectal mucosal immunity and inflammation-related malignant transformation, along with a deficiency in certain components of the innate immune system, may lead to serious consequences under conditions of inflammatory insult.


Subject(s)
Colitis/chemically induced , Colonic Neoplasms/etiology , Inflammation/pathology , Lactoferrin/metabolism , Animals , Apoptosis , Azoxymethane/toxicity , Cell Proliferation , Colitis/metabolism , Colitis/pathology , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Dextran Sulfate/toxicity , Disease Models, Animal , Down-Regulation , Gene Knockout Techniques , Humans , Inflammation/metabolism , Lactoferrin/genetics , Male , Mice , NF-kappa B/metabolism , Signal Transduction
19.
Tumour Biol ; 34(3): 1793-800, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23479198

ABSTRACT

LTF (lactotransferrin, or lactoferrin) plays important role in innate immunity, and its anti-tumor function has also been reported in multiple cancers. We previously reported that LTF is significantly down-regulated in nasopharyngeal carcinoma (NPC) and acts as a tumor suppressor by suppressing AKT signaling. However, the exact mechanism of the down-regulation of LTF in NPC has not been revealed. In the current study, we screened and identified LTF is a bona fide target of miR-214 in NPC cells. miR-214 mimics significantly suppressed LTF mRNA and protein expression levels in NPC cells. miR-214 not only can promote NPC cell proliferation and invasion abilities in vitro, but also can accelerate tumor formation and lung metastasis in a mouse xenograft model. The pro-tumor function of miR-214 was depended on LTF suppression since LTF re-expression can reverse it. miR-214 can also activate AKT signaling by suppressing LTF expression. Furthermore, miR-214 expression level was up-regulated in NPC especially in metastasis-prone NPC tumor tissues compared with normal nasopharyngeal epithelial tissues, while the LTF expression level was negatively correlated with miR-214, suggesting that miR-214 targeting is partly responsible for LTF down-regulation in NPC specimens.


Subject(s)
Cell Transformation, Neoplastic/pathology , Gene Expression Regulation, Neoplastic , Lactoferrin/metabolism , MicroRNAs/genetics , Nasopharyngeal Neoplasms/pathology , Nasopharynx/metabolism , Animals , Apoptosis , Blotting, Western , Case-Control Studies , Cell Movement , Cell Proliferation , Cells, Cultured , Down-Regulation , Female , Humans , Immunoenzyme Techniques , Lactoferrin/antagonists & inhibitors , Lactoferrin/genetics , Luciferases/metabolism , Lymphatic Metastasis , Male , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/metabolism , Middle Aged , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/metabolism , Nasopharynx/pathology , Neoplasm Staging , Prognosis , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
20.
J Innate Immun ; 4(4): 387-98, 2012.
Article in English | MEDLINE | ID: mdl-22433582

ABSTRACT

Lactoferrin (LF) is a multifunctional glycoprotein that plays an important role in native immune defense against infections, including human herpetic viruses, such as cytomegalovirus and herpes simplex virus types 1 and 2. However, its anti-Epstein-Barr virus (EBV, a γ-herpesvirus) function has not been reported in the literature. EBV is widespread in all human populations and is believed to be linked to tumorigenesis, such as lymphomas and nasopharyngeal carcinoma (NPC). We previously reported that LF expressed a significantly lower level in NPC tissues and was a likely tumor suppressor. Since EBV infection is a major carcinogen of NPC development, we investigated the effect of LF on EBV infection and found that LF could protect human primary B lymphocytes and nasopharyngeal epithelial cells from EBV infection, but had no effect on EBV genome DNA replication. LF prevented EBV infection of primary B cells mediated by its direct binding to the EBV receptor (CD21) on the B-cell surface. Tissue array immunohistochemistry revealed that LF expression was significantly downregulated in NPC specimens, in which high EBV viral capsid antigen-IgA levels were observed. These data suggest that LF may inhibit EBV infection and that its downregulation could contribute to NPC development.


Subject(s)
Epstein-Barr Virus Infections/immunology , Herpesvirus 4, Human/drug effects , Lactoferrin/pharmacology , Antigens, Viral/immunology , B-Lymphocytes/virology , Capsid Proteins/immunology , Carcinoma , Cell Line, Tumor , Cells, Cultured , DNA Replication , DNA, Viral/drug effects , DNA, Viral/genetics , DNA, Viral/physiology , Epithelial Cells/virology , Epstein-Barr Virus Infections/virology , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/physiology , Humans , Immunoglobulin A/biosynthesis , Lactoferrin/genetics , Lactoferrin/immunology , Lactoferrin/metabolism , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/immunology , Nasopharyngeal Neoplasms/virology , Nasopharynx/cytology , Nasopharynx/virology , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL