Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Plant Cell Rep ; 43(5): 119, 2024 Apr 17.
Article in English | MEDLINE | ID: mdl-38632145

ABSTRACT

KEY MESSAGE: Mutants lacking functional HYD2 homoeologs showed improved seedling growth, but comparable or increased susceptibility to salt stress in tillering plants, suggesting a developmentally restricted role of HYD2 in salt response. Salinity stress threatens global food security by reducing the yield of staple crops such as wheat (Triticum ssp.). Understanding how wheat responds to salinity stress is crucial for developing climate resilient varieties. In this study, we examined the interplay between carotenoid metabolism and the response to salt (NaCl) stress, a specific form of salinity stress, in tetraploid wheat plants with mutations in carotenoid ß-hydroxylase 1 (HYD1) and HYD2. Our investigation encompassed both the vulnerable seedling stage and the more developed tillering stage of wheat plant growth. Mutant combinations lacking functional HYD2 homoeologs, including hyd-A2 hyd-B2, hyd-A1 hyd-A2 hyd-B2, hyd-B1 hyd-A2 hyd-B2, and hyd-A1 hyd-B1 hyd-A2 hyd-B2, had longer first true leaves and slightly enhanced root growth during germination under salt stress compared to the segregate wild-type (control) plants. Interestingly, these mutant seedlings also showed decreased levels of neoxanthin and violaxanthin (xanthophylls derived from ß-carotene) and an increase in ß-carotene in roots. However, tillering hyd mutant and segregate wild-type plants generally did not differ in their height, tiller count, and biomass production under acute or prolonged salt stress, except for decreases in these parameters observed in the hyd-A1 hyd-B1 hyd-A2 hyd-B2 mutant that indicate its heightened susceptibility to salt stress. Taken together, these findings suggest a significant, yet developmentally restricted role of HYD2 homoeologs in salt-stress response in tetraploid wheat. They also show that hyd-A2 hyd-B2 mutant plants, previously demonstrated for possessing enriched nutritional (ß-carotene) content, maintain an unimpaired ability to withstand salt stress.


Subject(s)
Salt Stress , Seedlings , Triticum , beta Carotene , beta Carotene/metabolism , Carotenoids/metabolism , Salinity , Salt Stress/genetics , Seedlings/metabolism , Tetraploidy , Triticum/genetics , Plant Proteins/genetics , Plant Proteins/metabolism , Gene Expression Regulation, Plant
2.
Sci Rep ; 9(1): 17267, 2019 11 21.
Article in English | MEDLINE | ID: mdl-31754130

ABSTRACT

The secreted frizzled-related protein 5 gene (SFRP5) that antagonize the Wnt/ß-catenin signaling is frequently inactivated by promoter methylation and oncogenic activation of the Wnt signaling pathway is common in many cancers. The curcumin-rich Curcuma longa has been reported to potent anti-cancer property involved in epigenetic regulation to inhibit tumor suppressor gene methylation and re-expression. In a compounds screening, we found that curcumin can inhibit Wnt/ß-catenin signaling. Therefore, the aim of this study was to investigate the effects of curcumin on SFRP5 DNA methylation modification in an ovarian cancer cell line (SKOV3). SKOV3 cells were treated with DMSO, 10 µM 5-aza-2'-deoxycytidine (DAC), 5 µM DAC, 20 µM curcumin, and 20 µM curcumin combined with 5 µM DAC for 96 hours, following which RNA and proteins were extracted for further analysis. The results showed that curcumin combined with 5 µM DAC may inhibit cancer cell colony formation, migration through EMT (epithelial-mesenchymal transition) process regulation, total DNMT activity, especially in DNMT3a protein expression, and may also regulate tumor suppressor gene SFRP5 expression involved in the Wnt/ß-catenin signaling pathway. The combined treatment attenuated ovarian cancer development.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Curcumin/pharmacology , Ovarian Neoplasms/metabolism , Adaptor Proteins, Signal Transducing/drug effects , Carcinogenesis/drug effects , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Methylation/drug effects , DNA Modification Methylases/genetics , Epigenesis, Genetic/drug effects , Epigenomics/methods , Epithelial-Mesenchymal Transition/drug effects , Female , Humans , Promoter Regions, Genetic/drug effects , Wnt Signaling Pathway/drug effects , Wnt Signaling Pathway/genetics , beta Catenin/metabolism
3.
Int J Mol Sci ; 20(19)2019 09 20.
Article in English | MEDLINE | ID: mdl-31547144

ABSTRACT

Colorectal cancer (CRC) is one of the most common cancers and the second leading cause of cancer-related deaths. Discrepancies in clinical outcomes are observed even among patients with same-stage CRC due to molecular heterogeneity. Thus, biomarkers for predicting prognosis in CRC patients are urgently needed. We previously demonstrated that stage II CRC patients with NKX6.1 methylation had poor 5-year overall survival. However, the methylation frequency of NKX6.1 was only 23% in 151 pairs of CRC tissues. Thus, we aimed to develop a more robust prognostic panel for CRC using NKX6.1 in combination with three genes: LIM homeobox transcription factor 1α (LMX1A), sex-determining region Y-box 1 (SOX1), and zinc finger protein 177 (ZNF177). Through quantitative methylation analysis, we found that LMX1A, SOX1, and ZNF177 were hypermethylated in CRC tissues. LMX1A methylation was significantly associated with poor 5-year overall, and disease-free survivals in stage I and II CRC patients. Sensitivity and specificity analyses of the four-gene combination revealed the best sensitivity and optimal specificity. Moreover, patients with the four-gene methylation profile exhibited poorer disease-free survival than those without methylation. A significant effect of the four-gene methylation status on overall survival and disease-free survival was observed in early stage I and II CRC patients (p = 0.0016 and p = 0.0230, respectively). Taken together, these results demonstrate that the combination of the methylation statuses of NKX6.1, LMX1A, SOX1, and ZNF177 creates a novel prognostic panel that could be considered a molecular marker for outcomes in CRC patients.


Subject(s)
Colorectal Neoplasms , DNA Methylation , DNA, Neoplasm , Neoplasm Proteins , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , Disease-Free Survival , Female , HCT116 Cells , HT29 Cells , Humans , Male , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasm Staging , Survival Rate
4.
Exp Cell Res ; 368(1): 42-49, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29684384

ABSTRACT

The main problem in the treatment of non-small cell lung cancer (NSCLC) is metastasis. Epithelial-mesenchymal transition (EMT) is known as the critical signaling in tumor progression, metastasis, and also the drug resistance. In this study, we reported a novel gene Polymerase delta-interacting protein 2 (POLDIP2) was downregulated in NSCLC tissues and first demonstrated that overexpression of POLDIP2 increased the anchorage-independent growth (AIG) and invasiveness of H1299 cells. In addition, we examined that knockdown of POLDIP2 in H1299 and A549 cells reduced tumorigenicity and metastatic capacity in vitro and also in vivo. Moreover, downregulation of the cell proliferation marker cyclin D1 and EMT markers CDH2, Slug, and Twist was showed in H1299 cells by POLDIP2 knockdown, suggesting that the inhibition of malignancy was affected by modulating key genes for tumor growth and invasiveness. Taken together, our study is the first study that demonstrated that POLDIP2 gene was function as an oncogene in NSCLC and implied the oncogenic ability might be through promoting cell proliferation or EMT.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Gene Expression Regulation, Neoplastic , Neoplasm Invasiveness/genetics , Nuclear Proteins/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/physiology , Epithelial-Mesenchymal Transition/physiology , Gene Knockdown Techniques , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Neoplasm Invasiveness/pathology , Nuclear Proteins/metabolism
5.
Int J Mol Sci ; 19(2)2018 Feb 01.
Article in English | MEDLINE | ID: mdl-29389859

ABSTRACT

Galectins are glycan-binding proteins that contain one or two carbohydrate domains and mediate multiple biological functions. By analyzing clinical tumor samples, the abnormal expression of galectins is known to be linked to the development, progression and metastasis of cancers. Galectins also have diverse functions on different immune cells that either promote inflammation or dampen T cell-mediated immune responses, depending on cognate receptors on target cells. Thus, tumor-derived galectins can have bifunctional effects on tumor and immune cells. This review focuses on the biological effects of galectin-1, galectin-3 and galectin-9 in various cancers and discusses anticancer therapies that target these molecules.


Subject(s)
Galectin 1/metabolism , Galectin 3/metabolism , Galectins/metabolism , Neoplasms/metabolism , Animals , Antineoplastic Agents/therapeutic use , Blood Proteins , Galectin 1/antagonists & inhibitors , Galectin 1/genetics , Galectin 3/antagonists & inhibitors , Galectin 3/genetics , Galectins/antagonists & inhibitors , Galectins/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Molecular Targeted Therapy/methods , Neoplasms/drug therapy , Neoplasms/genetics
6.
Genes Chromosomes Cancer ; 57(5): 268-277, 2018 05.
Article in English | MEDLINE | ID: mdl-29363224

ABSTRACT

Colorectal cancer (CRC) is a common malignancy worldwide. CRC patients in the same stage often present with dramatically different clinical scenarios. Thus, robust prognostic biomarkers are urgently needed to guide therapies and improve treatment outcomes. The NKX6.1 gene has been identified as a hypermethylation marker in cervical cancer, functioning as a metastasis suppressor by regulating epithelial-mesenchymal transition. Here, we investigated whether hypermethylation of NKX6.1 might be a prognostic biomarker for CRC. By analyzing the methylation and expression of NKX6.1 in CRC tissues and CRC cell lines. We quantitatively examined the NKX6.1 methylation levels in 151 pairs of CRC tissues by using methylation-specific polymerase chain reaction analysis and found that NKX6.1 was hypermethylated in 35 of 151 CRC tissues (23%). NKX6.1 gene expression was inversely correlated with the DNA methylation level in CRC cell lines in vitro. Then, we analyzed the association of NKX6.1 methylation with clinical characteristics of these CRC patients. Our data demonstrated that patients with NKX6.1 methylation presented poorer 5-year overall survival (P = 0.0167) and disease-free survival (P = 0.0083) than patients without NKX6.1 methylation after receiving adjuvant chemotherapy. Most importantly, these data revealed that stage II CRC patients with NKX6.1 methylation had poorer 5-year disease-free survival (P = 0.0322) than patients without NKX6.1 methylation after adjuvant chemotherapy. Our results demonstrate that methylation of NKX6.1 is a novel prognostic biomarker in CRC and that it may be used as a predictor of the response to chemotherapy.


Subject(s)
Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , DNA Methylation , Homeodomain Proteins/genetics , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Chemotherapy, Adjuvant , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Disease-Free Survival , Female , Homeodomain Proteins/metabolism , Humans , Male , Middle Aged , Neoplasm Staging , Predictive Value of Tests , Prognosis , Promoter Regions, Genetic , Treatment Outcome , Young Adult
7.
Clin Epigenetics ; 8: 41, 2016.
Article in English | MEDLINE | ID: mdl-27110298

ABSTRACT

BACKGROUND: Related to genetic alteration, frequent promoter hypermethylation is also a contributing factor in the development of human cancers. Recently, we discovered numerous novel genes that were aberrantly methylated in hepatocellular carcinoma (HCC) by using Infinium HumanMethylation27 BeadChip array. We utilized a quantitative methylation-specific PCR (Q-MSP) system for the evaluation of PAX6 methylation in 29 normal controls and 160 paired HCC tissues and their adjacent non-tumor tissues. We verified the correlation between the methylation status of PAX6 and clinical characteristics with different viral status. RESULTS: Paired-box 6 promoter methylation was observed in 39.4 %, 15.6 %, and 3.4 % in primary HCCs, adjacent non-tumors, and normal control tissues, respectively. Methylation of the PAX6 promoter region in HCCs significantly increased compared with control tissues. PAX6 was frequently methylated in HCV-positive HCC tissues (61.3 %) and rarely methylated in HBV-positive (22.1 %) and double-negative HCC tissues (33.3 %). CONCLUSIONS: Our data suggests that promoter hypermethylation of PAX6 is a common event in HCCs and the association of PAX6 methylation in clinicopathological features is divergent with different viral status.


Subject(s)
Carcinoma, Hepatocellular/virology , DNA Methylation , Hepatitis B/genetics , Hepatitis C/genetics , Liver Neoplasms/virology , PAX6 Transcription Factor/genetics , Aged , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , Middle Aged , Promoter Regions, Genetic
8.
Taiwan J Obstet Gynecol ; 54(5): 572-9, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26522113

ABSTRACT

OBJECTIVE: Most endometrial carcinomas appear to develop from precursors (e.g., endometrial hyperplasia) that progress for several years. Patients who are ultimately diagnosed with carcinoma often present clinically with complaints of abnormal vaginal bleeding years before diagnosis, which offers an opportunity for early diagnosis and curative treatment. The analysis of DNA methylation may be used as a method for detecting endometrial cancer (EC). To test the potential clinical application of this method, we used quantitative methylation analysis of five genes in a full spectrum of endometrial lesions. MATERIALS AND METHODS: This hospital-based, prospective, case-controlled study was conducted on 68 patients, which included patients who had a normal endometrium (n = 18), hyperplasia of the endometrium (n = 24), and EC (n = 26). Methylation levels of the following genes were determined by using real-time methylation-specific polymerase chain reaction (PCR) amplification: zinc finger protein 177 (ZNF177), collagen type XIV α1 (COL14A1), dihydropyrimidinase-like 4 (DPYSL4), homeobox A9 (HOXA9), transmembrane protein with epidermal growth factor-like and two follistatin-like domains 2 (TMEFF2). The methylation index (MI) cutoff values for the different diagnoses were determined to test the sensitivity and specificity of the method and to generate the receiver operating characteristic (ROC) curves. The Mann-Whitney U test was used to test between-group differences in the MI. RESULTS: The MI of the five genes was significantly higher in EC than the MIs in specimens of hyperplasia of endometrium and normal appearance (p < 0.001). The ROC analysis demonstrated that the sensitivity, specificity, and accuracy for detecting EC were 92.3%, 94.4%, and 95.1%, respectively, for ZNF177; 92.3%, 94.4%, and 95.7%, respectively, for COL14A1; 80.8%, 94.4%, and 81.4%, respectively, for HOXA9; 65.4%, 94.4%, and 89.5%, respectively, for TMEFF2; and 61.5%, 94.4%, and 63.3%, respectively, for DPYSL4. The combined testing of ZNF177 and COL14A1 had the best specificity (100%), but compromised sensitivity (88.5%). CONCLUSION: Promoter methylation of ZNF177, COL14A1, HOXA9, DPYSL4, and TMEFF2 genes is a frequent epigenetic event in EC. Furthermore, the epigenetic hypermethylation of TMEFF2 may be a valuable marker for identifying undetected EC within endometrial hyperplasia.


Subject(s)
Carcinogenesis/genetics , DNA, Neoplasm/analysis , Early Detection of Cancer/methods , Endometrial Neoplasms/genetics , Genes, Neoplasm/genetics , Case-Control Studies , DNA Methylation , Endometrial Neoplasms/diagnosis , Female , Humans , Promoter Regions, Genetic , Prospective Studies , ROC Curve , Real-Time Polymerase Chain Reaction , Taiwan
9.
World J Gastroenterol ; 21(13): 3960-9, 2015 Apr 07.
Article in English | MEDLINE | ID: mdl-25852282

ABSTRACT

AIM: To examine the methylation levels of interleukin-1 receptor-associated kinase 3 (IRAK3) and GLOXD1 and their potential clinical applications in hepatocellular carcinoma (HCC). METHODS: mRNA expression and promoter methylation of IRAK3 and GLOXD1 in HCC cells were analyzed by reverse transcription-polymerase chain reaction (RT-PCR) and methylation-specific PCR (MSP), respectively. Using pyrosequencing results, we further established a quantitative MSP (Q-MSP) system for the evaluation of IRAK3 and GLOXD1 methylation in 29 normal controls and 160 paired HCC tissues and their adjacent nontumor tissues. We also calculated Kaplan-Meier survival curves to determine the applications of gene methylation in the prognosis of HCC. RESULTS: IRAK3 and GLOXD1 expression was partially restored in several HCC cell lines after treatment with 5-aza-2'-deoxycytidine (DNA methyltransferase inhibitor; 5DAC). A partial decrease in the methylated band was also observed in the HCC cell lines after 5DAC treatment. Using GLOXD1 as an example, we found a significant correlation between the data obtained from the methylation array and from pyrosequencing. The methylation frequency of IRAK3 and GLOXD1 in HCC tissues was 46.9% and 63.8%, respectively. Methylation of IRAK3 was statistically associated with tumor stage. Moreover, HCC patients with IRAK3 methylation had a trend toward poor 3-year disease-free survival (P < 0.05). CONCLUSION: IRAK3 and GLOXD1 were frequently methylated in HCC tissues compared to normal controls and nontumor tissues. IRAK3 methylation was associated with tumor stage and poor prognosis of patients. These data suggest that IRAK3 methylation is a novel prognostic marker in HCC.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Hepatocellular/genetics , DNA Methylation , Interleukin-1 Receptor-Associated Kinases/genetics , Liver Neoplasms/genetics , Adult , Aged , Base Sequence , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/pathology , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Hep G2 Cells , Humans , Kaplan-Meier Estimate , Liver Neoplasms/enzymology , Liver Neoplasms/mortality , Liver Neoplasms/pathology , Male , Middle Aged , Molecular Sequence Data , Neoplasm Staging , Predictive Value of Tests , Promoter Regions, Genetic , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Retrospective Studies , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA
10.
Clin Chem Lab Med ; 52(8): 1235-45, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24681432

ABSTRACT

BACKGROUND: Aberrant DNA methylation is associated with the development of hepatocellular carcinoma (HCC), suggesting that gene methylation could be a potential biomarker for detection of HCC. The aim of this study is to identify potential biomarkers in HCC. METHODS: We used the Infinium methylation array and a DNA-pooling strategy to analyze the genome-wide methylation profile in HCC. Quantitative methylation-specific PCR (Q-MSP) was used to validate homeobox A9 (HOXA9) methylation in 29 normal controls, 100 HCC samples and adjacent non-tumor tissues and in 74 plasma samples, including 40 patients with HCC. RESULTS: Ten genes (HOXA9, NEUROG1, TNFRSF10C, IRAK3, GFPT2, ZNF177, DPYSL4, ELOVL4, FSD1, and CACNA1G) showed differences in methylation between controls and HCCs. Of these, HOXA9 was significantly hypermethylated in HCCs (76.7%; 23/30) compared with controls (3.4%; 1/29). In addition, combination analysis of two- and three-gene sets for HCC detection showed greater sensitivity (90%-96.7%) and comparable specificity (93.1%-96.6%) to each individual gene (33.3%-76.7% and 55.2%-100.0%). HOXA9 methylation was further validated by Q-MSP in two independent set of clinical samples including 100 HCC and paired non-tumor tissues. Further, HOXA9 methylation could be detected in plasma from HCC patients (n=40) but not in normal plasma (n=34) (p<0.0005). Combined testing (either parameter positive) for α-fetoprotein (AFP, a plasma protein biomarker) and HOXA9 methylation showed greater sensitivity (94.6%) for detection of HCC than AFP alone (75.7%). CONCLUSIONS: These data suggest that methylation of HOXA9 could be a helpful biomarker to assist in HCC detection.


Subject(s)
Carcinoma, Hepatocellular/genetics , DNA Methylation , Homeodomain Proteins/genetics , Liver Neoplasms/genetics , Carcinoma, Hepatocellular/blood , Carcinoma, Hepatocellular/pathology , Cell Line , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/blood , Liver Neoplasms/pathology , Male , Promoter Regions, Genetic
11.
Biochim Biophys Acta ; 1833(12): 3206-3217, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24018208

ABSTRACT

We have reported previously that LIM homeobox transcription factor 1α (LMX1A) is hypermethylated and functions as a metastasis suppressor in cervical cancer cells. However, the regulation of LMX1A in carcinogenesis has not been reported. We aim to clarify whether specificity protein 1 (Sp1) and enhancer of zeste homolog 2 (EZH2) are involved in the regulation of LMX1A in cervical cancer. First we characterized the LMX1A promoter and used overexpression, knockdown, and reporter assays to show that Sp1 increased LMX1A promoter activity. Next, we used site-directed mutagenesis and electrophoresis mobility shift assays (EMSAs) to demonstrate that Sp1-binding sites were important for Sp1-mediated activation of the LMX1A promoter. Chromatin immunoprecipitation data demonstrated that Sp1 could bind directly to the LMX1A promoter and activate endogenous LMX1A expression in cells pretreated with 5-aza-2'-deoxycytidine (5-aza-dC). Knockdown of EZH2 decreased H3K27me3 histone modification but was insufficient to restore LMX1A expression. To explore the effect of EZH2 on the endogenous LMX1A promoter, we treated EZH2-knockdown cells with 5-aza-dC and trichostatin A (TSA) and then depleted the cells of drugs for 3days. H3K14ac was enriched at the LMX1A promoter in EZH2-knockdown cells and LMX1A mRNA was still expressed. Taken together, these data imply that Sp1 may activate LMX1A expression upon oncogenic stress during cervical cancer development. Moreover, suppression of EZH2 may delay resilencing of LMX1A after the removal of 5-aza-dC and TSA.


Subject(s)
LIM-Homeodomain Proteins/genetics , Polycomb Repressive Complex 2/metabolism , Sp1 Transcription Factor/metabolism , Transcription Factors/genetics , Uterine Cervical Neoplasms/genetics , Azacitidine/pharmacology , Base Sequence , Binding Sites , Cell Line, Tumor , Enhancer of Zeste Homolog 2 Protein , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Gene Silencing/drug effects , Histone Deacetylase Inhibitors/pharmacology , Histones/metabolism , Humans , LIM-Homeodomain Proteins/metabolism , Methylation/drug effects , Molecular Sequence Data , Promoter Regions, Genetic/genetics , Protein Binding/genetics , Sp1 Transcription Factor/antagonists & inhibitors , Transcription Factors/metabolism
12.
Cell Transplant ; 22(11): 2135-45, 2013.
Article in English | MEDLINE | ID: mdl-23067523

ABSTRACT

The differential activation of T helper (Th) cells and production of cytokines contribute to graft rejection or tolerance. In general, the Th1-type cytokines and cytotoxic T-cells are detected consistently in a host who is undergoing rejection, whereas Th2 responses are linked to a tolerance condition. Galectin-9 modulates Th1 cell immunity by binding to the T-cell immunoglobulin mucin-3 (Tim-3) molecule expressed on the Th1 cells. We investigate whether overexpression of galectin-9 in islets prolongs grafts survival in diabetic recipients. Islets were transduced with lentiviruses carrying galectin-9 and were then transplanted to streptozotocin-induced diabetic NOD/SCID recipients. The normoglycemic recipients then received splenocytes from diabetic NOD mice. Blood glucose concentration was monitored daily after adoptive transfer. The histology of the islet grafts and flow cytometric analyses were assessed at the end of the study. Overexpression of galectin-9 in islets prolonged graft survival in NOD/SCID mice after challenge with diabetogenic splenocytes (mean graft survival, 38.5 vs. 26.0 days, n=10, respectively; p=0.0096). The galectin-9-overexpressed grafts showed decreased infiltration of IFN-γ-producing CD4(+) and CD8(+) T-cells, but not of IL-17-producing CD4(+) T-cells. Strikingly, this islet-specific genetic manipulation did not affect the systemic lymphocyte composition, indicating that galectin-9 may regulate T-cell-mediated inflammation in situ. We demonstrate that galectin-9 protects grafts from Th1 and Tc1 cell-mediated rejections, suggesting that galectin-9 has preventive and/or therapeutic benefit in transplant therapy for autoimmune diabetes and may be applied further to the transplantation of other organs or tissues.


Subject(s)
Galectins/metabolism , Graft Survival/immunology , Islets of Langerhans Transplantation , Islets of Langerhans/cytology , Th1 Cells/immunology , Animals , Blood Glucose/analysis , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Experimental/surgery , Down-Regulation , Galectins/genetics , Interleukin-17/metabolism , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Lentivirus/genetics , Male , Mice , Mice, Inbred NOD , Mice, SCID , Spleen/cytology , Spleen/immunology , Spleen/transplantation , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/immunology , Th1 Cells/cytology , Transplantation, Homologous
13.
Hepatology ; 56(6): 2277-87, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22767186

ABSTRACT

UNLABELLED: Oncogenic activation of the Wnt/ß-catenin signaling pathway is common in hepatocellular carcinoma (HCC). Our recent studies have demonstrated that SRY (sex determining region Y)-box 1 (SOX1) and secreted frizzled-related proteins are concomitantly promoter-hypermethylated, and this might lead to abnormal activation of the Wnt signaling pathway in HCC. SOX1 encodes a transcription factor involved in the regulation of embryonic development and cell fate determination. However, the expression and functional role of SOX1 in HCC remains unclear. In this study, we confirmed via quantitative methylation-specific polymerase chain reaction that SOX1 was frequently downregulated through promoter hypermethylation in HCC cells and tissues. Overexpression of SOX1 by a constitutive or inducible approach could suppress cell proliferation, colony formation, and invasion ability in HCC cell lines, as well as tumor growth in nonobese diabetic/severe combined immunodeficiency mice. Conversely, knockdown of SOX1 by withdrawal of doxycycline could partially restore cell proliferation and colony formation in HCC cells. We used a T cell factor (TCF)-responsive luciferase reporter assay and western blot analysis to prove that SOX1 could regulate TCF-responsive transcriptional activity and inhibit the expression of Wnt downstream genes. Furthermore, we used glutathione S-transferase pull-down, co-immunoprecipitation, and confocal microscopy to demonstrate that SOX1 could interact with ß-catenin but not with the ß-catenin/TCF complex. Moreover, restoration of the expression of SOX1 induces significant cellular senescence in Hep3B cells. CONCLUSION: Our data show that a developmental gene, SOX1, may function as a tumor suppressor by interfering with Wnt/ß-catenin signaling in the development of HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , Genes, Tumor Suppressor , Liver Neoplasms/genetics , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Wnt Signaling Pathway , Animals , Anti-Bacterial Agents/pharmacology , Antigens, CD , Cadherins/genetics , Cadherins/metabolism , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Cycle Checkpoints , Cell Proliferation , Cyclin D1/genetics , Cyclin D1/metabolism , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/metabolism , DNA Methylation , Down-Regulation , Doxycycline/pharmacology , Genes, bcl-1 , Genes, myc , Hep G2 Cells , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Proliferating Cell Nuclear Antigen/metabolism , Promoter Regions, Genetic , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , RNA, Messenger/metabolism , SOXB1 Transcription Factors/drug effects , Snail Family Transcription Factors , Transcription Factors/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...