Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Cytokine ; 137: 155314, 2021 01.
Article in English | MEDLINE | ID: mdl-33002743

ABSTRACT

Senescent thymic stromal cells (TSCs) producing senescence-associated secretory phenotype (SASP) may play a role at later phases of thymic involution. However, the etiology and mechanisms responsible for TSC senescence remain to be elucidated. In the present study, the effects of oxidative stress on TSCs and role of miRNA-146a-5p in stress-induced premature senescence (SIPS) were identified. D-galactose (D-gal) induced oxidative stress in primary TSCs and a limited cumulative oxidative stress induced premature senescence but not apoptosis of TSCs. miRNA-146a-5p overexpression can mitigate the SIPS by targeting tumor necrosis factor receptor-associated factor 6 (TRAF6) instead of increasing autophagy clearance. Furthermore, exogenous miRNA-146a-5p reversed the upregulation of chemokines including Cxcl5, pro-inflammatory cytokines, and antimicrobial peptides in TSCs with SIPS. In conclusion, the accumulated oxidative stress may be partially responsible for senescence in TSCs and modulation of miRNA-146a-5p may attenuate this process.


Subject(s)
Cellular Senescence/drug effects , Galactose/pharmacology , MicroRNAs/genetics , Oxidative Stress/physiology , Stromal Cells/drug effects , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Survival/drug effects , Cell Survival/genetics , Cells, Cultured , Cellular Senescence/genetics , Cellular Senescence/physiology , Gene Expression Regulation , Mice, Inbred C57BL , Primary Cell Culture , Reactive Oxygen Species/metabolism , Stromal Cells/metabolism , TNF Receptor-Associated Factor 6/genetics , TNF Receptor-Associated Factor 6/metabolism , Thymus Gland/cytology
2.
Mol Med Rep ; 21(6): 2553-2559, 2020 06.
Article in English | MEDLINE | ID: mdl-32323791

ABSTRACT

Cathepsin A (CTSA) is a lysosomal protease that is abnormally expressed in various types of cancer; however, the function of CTSA in lung adenocarcinoma (LUAD) is unknown. The aim of the present study was to investigate the role of CTSA during LUAD development in vitro. The Cancer Genome Atlas (TCGA) database was used to analyze the expression of CTSA mRNA in LUAD tissues. CTSA was significantly upregulated in LUAD tissues compared with normal lung tissues. To explore the effect of CTSA on LUAD in vitro, LUAD A549 cells were transfected with CTSA small interfering RNA and the hallmarks of tumorigenesis were investigated using cell proliferation, cell cycle, wound healing, invasion and western blot assays. Following CTSA knockdown, proliferation of LUAD cells was decreased and an increased proportion of LUAD cells were arrested at the G0/G1 phase, with altered expression of critical cell cycle and proliferative marker proteins, including p53, p21 and proliferating cell nuclear antigen. Moreover, CTSA knockdown decreased the migration and invasion of A549 cells, as determined by wound healing, invasion, and western blotting assays. The expression levels of key proteins involved in epithelial­mesenchymal transition were analyzed by western blotting. CTSA knockdown enhanced the expression of E­cadherin, but decreased the expression of N­cadherin and ß­catenin in A549 cells. To the best of our knowledge, the present study suggested for the first time it has been identified that CTSA may serve as a tumor promoter in LUAD, enhancing the malignant progression of LUAD cells by promoting cell proliferation, migration and invasion. The results suggested that CTSA may serve as a novel therapeutic target for LUAD.


Subject(s)
Cathepsin A/metabolism , Cell Movement , Cell Proliferation , A549 Cells , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Cadherins/genetics , Cadherins/metabolism , Cathepsin A/antagonists & inhibitors , Cathepsin A/genetics , Cell Cycle Checkpoints , Epithelial-Mesenchymal Transition , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , beta Catenin/genetics , beta Catenin/metabolism
3.
Cell Biosci ; 9: 48, 2019.
Article in English | MEDLINE | ID: mdl-31249661

ABSTRACT

BACKGROUND: Adipocyte accumulation is a predominant feature of age-related thymic involution, but the mechanisms responsible for thymic adipogenesis remain to be elucidated. The aim of this study was to identify key regulators in thymic adipogenesis. We used rosiglitazone, a potent peroxisome proliferator-activated receptor γ (PPARγ) agonist, to induce adipogenic differentiation of OP9-DL1 cells, and investigated the differentially expressed proteins during adipogenic differentiation by using label-free quantitative proteomics. Furthermore, the effects of transforming growth factor ß1 (TGF-ß1) on rosiglitazone-induced adipogenic differentiation of OP9-DL1 cells as well as the underlying mechanisms were also investigated. RESULTS: Proteomic analysis identified 139 proteins differed significantly in rosiglitazone-treated cells compared with dimethyl sulphoxide (DMSO)-treated cells. Rosiglitazone-induced adipogenic differentiation was inhibited by TGF-ß1 treatment in OP9-DL1 cells and primary thymic stromal cells. Real-time PCR analysis showed significant increases in PPARγ and fatty acid binding protein 4 mRNA levels in rosiglitazone-treated cells, which were inhibited by TGF-ß1 treatment. TGF-ß1 down-regulated PPARγ expression at both mRNA and protein levels in OP9-DL1 cells. Chromatin immunoprecipitation analysis demonstrated that TGF-ß1 enhanced the binding of Smad2/3 and histone deacetylase 1, but reduced the binding of H3K14ac to the promoter of PPARγ gene. TGF-ß1 partially reversed the inhibitory effects of rosiglitazone on the expression of Axin2 and ß-catenin protein levels. TGF-ß1 inhibited rosiglitazone-induced adipogenic transformation in OP9-DL1 cells by down-regulation of PPARγ and activation of the canonical Wnt/ß-catenin signaling pathway. CONCLUSION: Taken together, activation of TGF-ß pathway may serve as a useful strategy to prevent thymic adiposity in age-related thymic involution.

4.
Mol Med Rep ; 19(1): 177-186, 2019 01.
Article in English | MEDLINE | ID: mdl-30483746

ABSTRACT

The influence and underlying mechanisms of human adipose­derived stem cells (Hu­ADSCs) on breast cancer cells in the tumor microenvironment remain unclear. Understanding the association between Hu­ADSCs and cancer cells may provide targets for breast cancer treatment and reference for the clinical application of stem cells. Therefore, a Hu­ADSC and breast cancer MCF7 cell coculture system was established to investigate the paracrine effects of Hu­ADSCs on MCF7 cell migration and invasion, in addition to the potential mechanism of action by reverse transcription­quantitative polymerase chain reaction and western blotting. Hu­ADSCs enhanced MCF7 cell migration and invasion by decreasing the expression of epithelial marker E­cadherin, and increasing the expression of interstitial marker N­cadherin and epithelial­mesenchymal transition (EMT) transcription factors in vitro. The EMT effect of cocultured MCF7 cells was inhibited with the addition of anti­transforming growth factor (TGF)­ß1 or phosphoinositide 3­kinase (PI3K) inhibitor LY294002, accompanied by a significant decrease in phosphorylated (p)­mothers against decapentaplegic homolog (Smad) and p­protein kinase B (AKT) expression. The data suggested that the paracrine effect of Hu­ADSCs in the tumor microenvironment promoted the EMT of MCF7 cells by cross interacting with the TGF­ß/Smad and PI3K/AKT pathways.


Subject(s)
Adipose Tissue/metabolism , Breast Neoplasms/metabolism , Epithelial-Mesenchymal Transition/physiology , Mesenchymal Stem Cells/metabolism , Signal Transduction/physiology , Adipose Tissue/pathology , Adult , Biomarkers, Tumor/metabolism , Breast Neoplasms/pathology , Cadherins/metabolism , Cell Line, Tumor , Cell Movement/physiology , Coculture Techniques/methods , Female , Humans , MCF-7 Cells , Mesenchymal Stem Cells/pathology , Neoplasm Invasiveness/pathology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Smad Proteins/metabolism , Transforming Growth Factor beta/metabolism , Tumor Microenvironment/physiology , Young Adult
5.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-793111

ABSTRACT

@#Objective: To investigate the effects of umbilical cord-derived mesenchymal stem cells (UC-MSCs) on apoptosis and proliferation of human lung adenocarcinoma A549 cells via PI3K/AKT signaling pathway, and to explore the mechanism. Methods: UCMSCs were isolated from human umbilical cord tissues by enzyme digestion method and cultured in vitro. The immunophenotypes of the obtained MSCs were identified by flow cytometry. The culture supernatant of UC-MSCs was collected to establish an indirect in vitro co-culture system of UC-MSCs conditioned medium and lung adenocarcinomaA549 cell line. Proliferation ofA549 cells was detected by CCK-8 assay; apoptosis ofA549 cells was determined byAnnexin V/PI double staining, and cell cycle distribution of tumor cells was determined by PI staining. The transcription levels of apoptosis and proliferation associated downstream genes in the PI3K/AKT pathway, such as CyclinD1, BAX and Bcl-2, were detected by quantitative polymerase chain reaction (qPCR). Moreover, Wb was utilized to detect the expression levels of PI3K/AKT pathway-related proteins. Results: The culture flask was filled with fibroblast-like cells, arranged in parallel, with spiral growth after three weeks of isolation and culture of human umbilical cord tissues. The flow cytometry results revealed that the MSC markers CD73, CD90 and CD105, but not CD45 and HLA-DR, were expressed on obtained cells.After indirect in vitro co-culture of UC-MSCs conditioned medium and lung adenocarcinoma A549 cells, the proliferation rate of A549 cells was significantly decreased; the apoptosis rate was significantly increased, and the cell cycle was obviously arrested at the G1 phase as compared with the control group (all P<0.01). The transcription levels of PI3K/AKT signaling pathway-related factors, CyclinD1 and Bcl-2 were down-regulated, and the transcription level of BAX was up-regulated (all P<0.01). The total AKT was not changed, but p-AKT protein expression was decreased in a dose-dependent manner inA549 cells cultured in UC-MSCs conditioned medium (P<0.01). Conclusion: UC-MSCs can affect the proliferation and the apoptosis of A549 cells, and arrest cells in G1 phase. The main mechanism is that UC-MSCs can inhibit the PI3K/AKT signaling pathway in A549 cells, providing an experimental basis for exploring the safety and effectiveness of clinical application of UC-MSCs.

6.
Front Pharmacol ; 9: 1284, 2018.
Article in English | MEDLINE | ID: mdl-30483127

ABSTRACT

Adipocyte deposition is believed to be a primary characteristic of age-related thymic involution. Herein, we cultured primary thymic stromal cells (TSCs), used rosiglitazone, a potent peroxisome proliferator-activated receptor γ (PPARγ) agonist, to induce adipogenic differentiation, and investigated the differentially expressed genes during adipogenic differentiation by using RNA-sequencing analysis. Furthermore, the effects of Notch1 on rosiglitazone-induced adipogenic differentiation of TSCs as well as the underlying mechanisms were also investigated. As a result, we identified a total of 1737 differentially expressed genes, among which 965 genes were up-regulated and 772 genes were down-regulated in rosiglitazone-treated cells compared with control cells. Gene ontology (GO) enrichment analysis showed that the GO terms were enriched in metabolic process, intracellular, and protein binding. Kyoto encyclopedia of genes and genomes (KEGG) analysis showed that a number of pathways, including ubiquitin mediated proteolysis, PPAR signaling pathway, and mammalian target of rapamycin (mTOR) signaling pathway were predominantly over-represented. Meanwhile, overexpression of Notch1 suppressed and inhibition of Notch1 promoted rosiglitazone-induced adipogenic differentiation in TSCs, and the pro-adipogenic effects of the Notch inhibitor DAPT were associated with the activation of autophagy. Taken together, our results suggest that Notch1 is a key regulator in thymic adipogenesis and may serve as a potential target to hinder thymic adiposity in age-related thymic involution.

7.
Cell Biol Int ; 41(4): 447-456, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28185342

ABSTRACT

Adipocyte deposition is a key feature of age-related thymic involution, but the underlying mechanisms responsible for thymic adiposity remain to be elucidated. In the present study, we utilized rosiglitazone, a potent peroxisome proliferator-activated receptor γ agonist, to induce adipogenic differentiation of OP9-DL1 cells, and detected the metabolomics alterations during adipogenic differentiation by using liquid chromatography-mass spectrometry. The obtained metabolites were further processed by multivariate statistical analysis, including principal component analysis, partial least squares discriminant analysis, and orthogonal projection on latent-structures discriminant analysis. As a result, we identified a total of 33 significantly differential metabolites between dimethyl sulphoxide- and rosiglitazone-treated OP9-DL1 cells, which were closely related to the dysregulation of phospholipid metabolism pathway, oxidative stress, and associated amino acid metabolism. Meanwhile, two pathways including glycerophospholipid metabolism and nitrogen metabolism were significantly perturbed (P < 0.05). Collectively, our results may provide some heuristic guidance for addressing the underlying mechanism of thymic adipogenesis, and future studies are warranted to unravel the functions of these altered metabolites in thymic adipogenesis.


Subject(s)
Adipocytes/physiology , Adipogenesis , Animals , Cell Line , Chromatography, High Pressure Liquid , Chromatography, Liquid , Metabolome , Metabolomics , Mice , Oxidative Stress , Rosiglitazone , Spectrometry, Mass, Electrospray Ionization , Thiazolidinediones/pharmacology
8.
Onco Targets Ther ; 9: 2709-16, 2016.
Article in English | MEDLINE | ID: mdl-27217781

ABSTRACT

HER2 positivity has been well studied in various cancers, but its importance in non-small-cell lung cancer (NSCLC) is still being explored. In this study, quantitative reverse transcription polymerase chain reaction (qRT-PCR) was performed to detect HER2 and COX-2 expression in NSCLC tissues. Then, pcDNA3.1-HER2 was used to overexpress HER2, while HER2 siRNA and COX-2 siRNA were used to silence HER2 and COX-2 expression. MTT assay and invasion assay were used to detect the effects of HER2 on cell proliferation and invasion. Our study revealed that HER2 and COX-2 expression were upregulated in NSCLC tissues and HER2 exhibited a significant positive correlation with the levels of COX-2 expression. Overexpression of HER2 evidently elevated COX-2 expression, while silencing of HER2 evidently decreased COX-2 expression. Furthermore, overexpressed HER2 induced the ERK phosphorylation, and this was abolished by the treatment with U0126, a pharmacological inhibitor of MEK, an upstream kinase of ERK. HER2-induced expression and promoter activity of COX-2 were also suppressed by U0126, suggesting that the MEK/ERK signaling pathway regulates COX-2 expression. In addition, HER2 induced activation of AKT signaling pathway, which was reversed by pretreatment with U0126 and COX-2 siRNA. MTT and invasion assays revealed that HER2 induced cell proliferation and invasion that were reversed by pretreatment with U0126 and COX-2 siRNA. In this study, our results demonstrated for the first time that HER2 elevated COX-2 expression through the activation of MEK/ERK pathway, which subsequently induced cell proliferation and invasion via AKT pathway in NSCLC tissues.

9.
Cell Biol Int ; 40(8): 842-6, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27189906

ABSTRACT

Age-related thymic involution is characterized by a loss of thymic epithelial cells (TECs) and a concomitant increase in adipocytes, but the mechanisms involved in thymic adipogenesis are still not clear. Transforming growth factor ß1 (TGF-ß1) is a pleiotropic cytokine that has been reported to be up-regulated with age in thymic stromal cells in both human and mouse. However, the exact role of TGF-ß1 in age-related thymic involution remains to be further elucidated. On the basis of previous findings, we propose a novel hypothesis that TGF-ß1 functions a dual role in age-related thymic involution. On one hand, up-regulation of TGF-ß1 promotes epithelial to mesenchymal transition (EMT) process in TECs via activating forkhead box protein C2 (FoxC2). On the other hand, TGF-ß1 inhibits the transdifferentiation of EMT-derived mesenchymal cells to adipocytes in the thymus. If confirmed, our hypothesis will not only provide further evidence supporting that the transdifferentiation of TECs into pre-adipocytes represents a source of thymic adiposity during age-related thymic involution, but also uncover a unique role of TGF-ß1 in the transdifferentiation of TECs into pre-adipocytes. Collectively, the inhibition of TGF-ß1 may serve as a strategy to hinder age-related thymic involution or even to restore thymic function in the elderly.


Subject(s)
Adipogenesis/physiology , Epithelial-Mesenchymal Transition/physiology , Thymus Gland/physiology , Transforming Growth Factor beta1/metabolism , Adipocytes/drug effects , Age Factors , Animals , Cell Differentiation/physiology , Cell Movement/physiology , Cell Transdifferentiation/physiology , Epithelial Cells/metabolism , Forkhead Transcription Factors/metabolism , Humans , Mice , Signal Transduction , Stromal Cells/cytology , Thymus Gland/cytology , Thymus Gland/metabolism , Transcriptional Activation , Up-Regulation
10.
Sheng Li Xue Bao ; 68(1): 75-86, 2016 Feb 25.
Article in Chinese | MEDLINE | ID: mdl-26915325

ABSTRACT

The thymus is a vital primary lymphoid organ that provides unique microenvironments for the proliferation, differentiation, and maturation of T cells. With advancing age, however, the thymus gradually undergoes age-related involution and reduction in immune function, which are characterized by decreases in tissue size, cellularity, and naïve T cell output. This dynamic process leads to the reduced efficacy of the immune system with age and contributes to the increased susceptibility to infection, autoimmune disease, and cancer. In addition, bone marrow transplantation, radio-chemotherapy and virus infection also impair the thymus and give rise to the decline in immune function. Therefore, understanding the molecular mechanisms involved in age-related thymic involution and development of novel therapeutic strategies for thymic rejuvenation have gained considerable interests in recent years. This review emphasizes thymic microenvironments and thymocyte-stromal cell interactions and summarizes our current knowledge about thymic rejuvenation in terms of sex steroid, cytokines, growth factors, hormones, transcription factors, cell graft, and microRNAs. At the end of each discussion, we also highlight unanswered issues and describe possible future research directions.


Subject(s)
Rejuvenation , Aging , Cell Differentiation , Cytokines , Gonadal Steroid Hormones , Hormones , Humans , Intercellular Signaling Peptides and Proteins , Stromal Cells , T-Lymphocytes , Thymus Gland
11.
Mol Med Rep ; 12(5): 7568-76, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26397044

ABSTRACT

The thymus is a central lymphoid organ that is responsible for T-lymphocyte development and maturation. Through negative and positive selection, lymphoid progenitor cells, which initiate from the bone marrow, develop into mature T cells in the thymus, and are subsequently involved in peripheral cell immunity. It has been reported that the Wnt signaling pathway exists widely in thymic epithelial cells and T lymphocytes. Wnt signaling affects the shape and function of thymic epithelial cells and has an important role in maintaining pro­T­cells, and in the subsequent T­cell differentiation. Previous studies have demonstrated that the Wnt signaling pathway participates in age­associated thymic involution. In the present study alterations in proliferation and apoptosis were investigated in murine thymic cells during aging. The results of the present study demonstrated that the aged thymus was characterized by markedly decreased cell numbers, as well as decreased proliferation and increased apoptosis. Concurrently, age­associated changes in thymic cell number and function were accompanied by a decrease in the transcription levels of Wnt4, and downregulation of forkhead box N1 and B­cell lymphoma­extra large, which are two target genes of the Wnt4 signaling pathway. In vitro studies demonstrated that activation of the Wnt4 signaling pathway promotes mouse thymus epithelial cell 1 (MTEC1) cell proliferation, and that Wnt4 signaling modulation alleviates dexamethasone­mediated MTEC1 cell apoptosis. These results suggest that normal expression levels of Wnt4 have a critical role in maintaining the balance between cell proliferation and apoptosis. Alterations in the Wnt signaling pathway may disrupt the epithelial network structure of the thymus, eventually leading to microenvironmental damage. Therefore, further studies regarding the effects of the Wnt signaling pathway on thymus development and age-related thymic involution, may be beneficial for improving the health conditions of the elderly.


Subject(s)
Apoptosis , Cell Proliferation , Wnt4 Protein/metabolism , Animals , Cell Line , Female , Male , Mice, Inbred C57BL , Thymocytes/physiology , Thymus Gland/cytology , Thymus Gland/physiology , Wnt Signaling Pathway
12.
Urol Int ; 92(1): 104-15, 2014.
Article in English | MEDLINE | ID: mdl-24051497

ABSTRACT

INTRODUCTION: During bladder tumorigenesis, thymopoiesis is usually downregulated. Considering that the thymus is the site of most T-cell development, this phenomenon may be related to thymic involution. However, the mechanisms involved in this phenomenon remain to be elucidated. MATERIALS AND METHODS: An MB 49 murine bladder tumor model was used to identify mechanisms that might underlie this process. RESULTS: The thymuses of tumor-bearing mice showed less cellularity than those of healthy mice. Involution was found to be associated with less proliferation and more apoptosis of thymic epithelial cells (TEC). Foxn1, KGF, and IL-7, three factors known to be involved in thymic development, were also downregulated in the thymuses of tumor bearers. When these mice were intravenously injected with KGF, the thymic microenvironment, thymopoiesis, and T-cell differentiation all returned to near normal status. CONCLUSIONS: The decreases in thymopoiesis and impaired T-cell differentiation may be attributable to changes in the thymic microenvironment. Improving the function of TEC, rather than T-cell progenitors, should be the focus of therapy.


Subject(s)
Carcinoma, Transitional Cell/metabolism , Cellular Microenvironment , Signal Transduction , Thymus Gland/metabolism , Urinary Bladder Neoplasms/metabolism , Animals , Apoptosis , Atrophy , Carcinoma, Transitional Cell/immunology , Carcinoma, Transitional Cell/pathology , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Fibroblast Growth Factor 7/metabolism , Forkhead Transcription Factors/metabolism , Interleukin-7/metabolism , Male , Mice , Mice, Inbred C57BL , T-Lymphocytes/immunology , Thymus Gland/immunology , Thymus Gland/pathology , Time Factors , Urinary Bladder Neoplasms/immunology , Urinary Bladder Neoplasms/pathology
13.
Int J Mol Med ; 32(4): 835-42, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23969555

ABSTRACT

Age-related thymic involution is accompanied by a decrease in thymopoiesis and, thus, a deficiency in T cell-mediated immunity in the elderly. A number of events involved in thymic involution have been discovered; however, it remains unclear as to whether they are causes or consequences of thymic involution. These events include the degeneration of T cell progenitors, as well as the deterioration of the thymic stroma, which is a characteristic of thymic epithelial cell loss due to increased apoptosis and decreased cell proliferation. MicroRNAs (miRNAs) are believed to play important roles in the regulation of cell death and proliferation during the aging process. In the present study, we compared the transcriptional levels of various miRNAs in TECs from young and aged mice using microarray analysis. Quantitative PCR was performed to confirm the changes in the expression of miRNAs in the different age groups. Possible downstream targets and pathways of these miRNAs were predicted by performing bioinformatics analysis. To the best of our knowledge, this is the first study to systematically analyze the expression of miRNAs in mouse TECs and to demonstrate that miRNA expression is altered with thymic aging.


Subject(s)
Aging/genetics , Epithelial Cells/metabolism , Gene Expression Profiling , MicroRNAs/genetics , Thymus Gland/cytology , Aging/metabolism , Animals , Apoptosis/physiology , Cell Proliferation , Computational Biology , Humans , Immunity, Cellular/immunology , Mice , Mice, Inbred C57BL , MicroRNAs/metabolism , Microarray Analysis , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Thymus Gland/metabolism
14.
J Exp Clin Cancer Res ; 28: 125, 2009 Sep 09.
Article in English | MEDLINE | ID: mdl-19737423

ABSTRACT

BACKGROUND: The aim of this study was to determine the clinicopathological features of gastric cancers with loss of imprinting (LOI) of LIT1. Insulin-like growth factor 2 (IGF2) and H19 in Chinese patients. METHODS: DNA and RNA from tumours were amplified and then digested with RsaI, ApaI and HinfI, and RsaI respectively to determine the LOI status. The demographic and clinicopathological characteristics in LOI positive and LOI negative patients were compared and tested with Statistical analysis. RESULTS: Of the 89 patients enrolled for analysis, 22, 40 and 35 were heterozygous and thus informative for LIT1, IGF2 and H19 LOI analyses respectively. The positive rate of LIT1, IGF2 and H19 LOI of gastric cancer tissues were 54.6% (12/22), 45% (18/40) and 8.6% (3/32) in Chinese patients. Gastric corpus cancer (8/10, 80%) were more likely to have LOI of IGF2 in tumours than antrum cancers (10/30, 33.3%){odds ratio (OR) = 8, 95% confidence intervals (CI) = 1.425-44.920, p = 0.018)}. LOI of IGF2 in tumours was also associated with the lymph node metastasis (LNM) (OR = 4.5, 95% CI = 1.084-18.689, p = 0.038). CONCLUSION: IGF2 LOI is present in high frequency in Chinese gastric cancer patients, especially those with gastric corpus cancer.


Subject(s)
Genetic Predisposition to Disease , Insulin-Like Growth Factor II/genetics , Lymphatic Metastasis/genetics , Stomach Neoplasms/genetics , Aged , Asian People , Female , Genomic Imprinting , Humans , Loss of Heterozygosity , Male , Middle Aged , Potassium Channels, Voltage-Gated/genetics , RNA, Long Noncoding , RNA, Untranslated/genetics , Risk Factors , Stomach Neoplasms/pathology
15.
Int Immunol ; 19(10): 1201-11, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17804689

ABSTRACT

Aging is associated with reduced numbers of all thymocyte sub-populations, including early T-cell progenitors. However, it is unclear if this is due to inadequate recruitment of lymphohematopoietic progenitor cells (LPCs) to the aged thymus or to abnormal development of T cells within the thymus. We found that LPCs from young mice were recruited equally well to the thymi of young or aged mice and that thymic stromal cells (TSCs) from young and old mice expressed similar levels of P-selectin and CCL25, which are believed to mediate recruitment of LPCs to the adult thymus. However, the number of recruited thymocytes in old thymus was markedly reduced after two weeks, indicating that T-cell development or proliferation is defective in the aged thymus. We also found that LPCs from aged and young mice have similar capacities to seed a fetal thymus that was transplanted under the kidney capsule. Thymic epithelial cells (TECs) in aged mice had lower proliferative capacity and higher rate of apoptosis, compared with findings in young animals. In addition, immunofluorescence staining with antibodies to cortical and medullary TECs revealed that aged thymi had a disorganized thymic stromal architecture, combined with reduced cellularity of the medulla, and apoptosis of thymocyte sub-populations in the medullary microenvironment was increased, compared with that in young mice. We conclude that aging does not impair recruitment of LPCs to the thymus, but is characterized by abnormalities in thymic epithelial architecture, especially medullary TEC function that may provide sub-optimal support for thymic development of LPCs.


Subject(s)
Aging/immunology , Epithelial Cells/immunology , Hematopoietic Stem Cells/immunology , Lymphoid Progenitor Cells/immunology , Thymus Gland/immunology , Animals , Apoptosis , Cell Proliferation , Chemokines, CC/genetics , Chemokines, CC/metabolism , Epithelial Cells/cytology , Fetus/immunology , Mice , Mice, Inbred C57BL , P-Selectin/genetics , P-Selectin/metabolism , Thymus Gland/cytology
16.
Aging Cell ; 6(5): 663-72, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17681038

ABSTRACT

It has been speculated that aging lymphohematopoietic progenitor cells (LPC) including hematopoietic stem cells (HSC) and early T-cell progenitors (ETP) have intrinsic defects that trigger age-related thymic involution. However, using a different approach, we suggest that that is not the case. We provided a young thymic microenvironment to aged mice by transplanting a fetal thymus into the kidney capsule of aged animals, and demonstrated that old mouse-derived LPCs could re-establish normal thymic lymphopoiesis and all thymocyte subpopulations, including ETPs, double negative subsets, double positive, and CD4(+) and CD8(+) single positive T cells. LPCs derived from aged mice could turn over young RAG(-/-) thymic architecture by interactions, as well as elevate percentage of peripheral CD4(+)IL-2(+) T cells in response to costimulator in aged mice. Conversely, intrathymic injection of ETPs sorted from young animals into old mice did not restore normal thymic lymphopoiesis, implying that a shortage and/or defect of ETPs in aged thymus do not account for age-related thymic involution. Together, our findings suggest that the underlying cause of age-related thymic involution results primarily from changes in the thymic microenvironment, causing extrinsic, rather than intrinsic, defects in T-lymphocyte progenitors.


Subject(s)
Aging , Lymphopoiesis , Precursor Cells, T-Lymphoid/physiology , T-Lymphocyte Subsets/physiology , Thymus Gland/cytology , Thymus Gland/physiology , Animals , Antigens, CD/immunology , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/immunology , Antigens, Differentiation, T-Lymphocyte/metabolism , Bone Marrow Cells/cytology , Fetal Tissue Transplantation , Mice , Mice, Inbred C57BL , Precursor Cells, T-Lymphoid/immunology , T-Lymphocyte Subsets/immunology , Thymus Gland/immunology , Thymus Gland/transplantation
17.
Genomics ; 80(6): 575-84, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12504849

ABSTRACT

Human 11p15.5, as well as its orthologous mouse 7F4/F5, is known as the imprinting domain extending from IPL/Ipl to H19. OBPH1 and Obph1 are located beyond the presumed imprinting boundary on the IPL/Ipl side. We determined full-length cDNAs and complete genomic structures of both orthologues. We also investigated their precise imprinting and methylation status. The orthologues resembled each other in genomic structure and in the position of the 5' CpG island and were expressed ubiquitously. OBPH1 and Obph1 were predominantly expressed from the maternal allele only in placenta, with hypo- and not differentially methylated 5' CpG islands in both species. These results suggested that the imprinting domain would extend beyond the presumed imprinting boundary and that methylation of the 5' CpG island was not associated with the imprinting status in either species. It remains to be elucidated whether the gene is under the control of the KIP2/LIT1 subdomain or is regulated by a specific mechanism. Analysis of the precise genomic sequence around the region should help resolve this question.


Subject(s)
Genomic Imprinting , Receptors, Steroid/genetics , Alleles , Amino Acid Sequence , Animals , Base Sequence , Chromosome Mapping , Chromosomes, Human, Pair 11/genetics , CpG Islands/genetics , DNA/chemistry , DNA/genetics , DNA/metabolism , DNA Methylation , Female , Gene Expression , Humans , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Polymorphism, Single Nucleotide , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Synteny
SELECTION OF CITATIONS
SEARCH DETAIL
...