Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
1.
Cancer Lett ; 491: 22-35, 2020 10 28.
Article in English | MEDLINE | ID: mdl-32730779

ABSTRACT

Cancer-associated fibroblasts (CAFs) are the main stromal cells in the tumour microenvironment (TME). We found that the distribution of CAFs was significantly increased with tumour progression and led to a poor prognosis. In vitro and in vivo assays revealed that CAFs enhanced colorectal cancer (CRC) metastasis. Based on extraction and identification of exosomes of CAFs and normal fibroblasts (NFs), CAFs-exo showed higher expression of miR-17-5p than NFs-exo and could deliver exosomal miR-17-5p from parental CAFs to CRC cells. Further exploration verified that miR-17-5p influenced CRC metastasis capacity and directly targeted 3'-untranslated regions (UTRs) of RUNX family transcription factor 3(RUNX3). Our findings further revealed that RUNX3 interacted with MYC proto-oncogene(MYC) and that both RUNX3 and MYC bound to the promoter of transforming growth factor beta1(TGF-ß1) at base pairs 1005-1296, thereby activating the TGF-ß signalling pathway and contributing to tumour progression. In addition, RUNX3/MYC/TGF-ß1 signalling sustained autocrine TGF-ß1 to activate CAFs, and activated CAFs released more exosomal miR-17-5p to CRC cells, forming a positive feedback loop for CRC progression. Taken together, these data provide a new understanding of the potential diagnostic value of exosomal miR-17-5p in CRC.


Subject(s)
Cancer-Associated Fibroblasts/physiology , Colorectal Neoplasms/pathology , Core Binding Factor Alpha 3 Subunit/physiology , Exosomes/physiology , MicroRNAs/physiology , Proto-Oncogene Proteins c-myc/physiology , Transforming Growth Factor beta1/physiology , Adult , Aged , Animals , Cell Line, Tumor , Colorectal Neoplasms/etiology , Feedback, Physiological , Female , Humans , Male , Mice , Mice, Inbred BALB C , Middle Aged , Phenotype , Proto-Oncogene Mas , Signal Transduction , Transforming Growth Factor beta1/genetics , Tumor Microenvironment
2.
Cell Stress Chaperones ; 25(5): 793-802, 2020 09.
Article in English | MEDLINE | ID: mdl-32681471

ABSTRACT

Lung toxicity is the main cause of the death from methamphetamine (MA) abuse, but its mechanism has remained unclear. The purpose of our study was to investigate if MA can induce epithelial-to-mesenchymal transition (EMT) and if RUNX3 is involved in oxidative EMT in MA-induced chronic lung injury. The rats were divided into the control group and MA group. Extracted lungs were used for morphological measurements and Western blot. The alveolar epithelial cells were cultured or transfected and then treated with MA or/and N-acetyl cysteine (NAC) followed by flow cytometry, Western blot, and immunohistochemistry. Chronic exposure to MA resulted in the lower growth ratio of weight, increased right ventricular index, thickened alveolar walls, and reduced number of alveolar sacs. Long-term administration with MA caused oxidative stress and pulmonary EMT. NAC increased RUNX3 and alleviated EMT. However, after knockdown of RUNX3, reactive oxygen species (ROS) levels were significantly upregulated, indicating that RUNX3 was closely related to oxidative stress. Knockdown of RUNX3 aggravated MA-induced EMT by activating RUNX3-dependent TGF-ß signaling. Therefore, RUNX3 may be the key to oxidative EMT in methamphetamine-induced chronic lung injury.


Subject(s)
Alveolar Epithelial Cells/drug effects , Core Binding Factor Alpha 3 Subunit/physiology , Epithelial-Mesenchymal Transition , Lung Injury/chemically induced , Methamphetamine/toxicity , Oxidative Stress/drug effects , A549 Cells , Animals , Chronic Disease , Humans , Lung/metabolism , Lung/pathology , Male , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism
3.
Urol Oncol ; 38(9): 740.e11-740.e20, 2020 09.
Article in English | MEDLINE | ID: mdl-32600926

ABSTRACT

BACKGROUND: Kidney cancer usually shows no symptoms until the tumor is relatively large, and current drugs fail to stop the tumor recurrence. The transcriptional factor Runt-related transcription factor 3 (RUNX3) has been reported to function as a tumor suppressor in many types of cancers. METHODS: Kidney cancer and adjacent normal tissues were collected from 12 patients to test the expression of RUNX3 by real-time quantitative PCR, immunoblotting, and immunohistochemistry. Promoter methylation status of RUNX3 was determined using methylation analysis from 103 patient samples. Kidney cancer cell lines and xenograft mouse model were used to investigate the promoter methylation and cancer progression through inhibitor treatment and loss/gain-of-function experiments. RESULTS: RUNX3 was significantly downregulated in kidney cancer tissues and cells, which could be elevated by higher methylation status at its promoter region. RUNX3 promoter methylation was positively correlated with poor prognosis of kidney cancer. RUNX3 loss-of-function promoted the cell proliferation, migration, and invasion of kidney cancer cells, in contrast, RUNX3 overexpression inhibited the cancer cell progression. This study provides the first instance of the effect of RUNX3 expression and its promoter methylation status on kidney cancer. CONCLUSION: Targeting RUNX3 pathway and its promoter methylation are potential therapeutic strategies to treat kidney cancer.


Subject(s)
Core Binding Factor Alpha 3 Subunit/physiology , Kidney Neoplasms/pathology , Animals , Cells, Cultured , Disease Models, Animal , Disease Progression , Down-Regulation , Humans , Mice , Mice, Nude , Prognosis
4.
Nat Commun ; 10(1): 3306, 2019 07 24.
Article in English | MEDLINE | ID: mdl-31341159

ABSTRACT

Memory CD8+ T cells have the ability to provide lifelong immunity against pathogens. Although memory features generally arise after challenge with a foreign antigen, naïve CD8 single positive (SP) thymocytes may acquire phenotypic and functional characteristics of memory cells in response to cytokines such as interleukin-4. This process is associated with the induction of the T-box transcription factor Eomesodermin (EOMES). However, the underlying molecular mechanisms remain ill-defined. Using epigenomic profiling, we show that these innate memory CD8SP cells acquire only a portion of the active enhancer repertoire of conventional memory cells. This reprograming is secondary to EOMES recruitment, mostly to RUNX3-bound enhancers. Furthermore, EOMES is found within chromatin-associated complexes containing BRG1 and promotes the recruitment of this chromatin remodelling factor. Also, the in vivo acquisition of EOMES-dependent program is BRG1-dependent. In conclusion, our results support a strong epigenetic basis for the EOMES-driven establishment of CD8+ T cell innate memory program.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Core Binding Factor Alpha 3 Subunit/physiology , DNA Helicases/physiology , Epigenesis, Genetic , Immunologic Memory , Nuclear Proteins/physiology , T-Box Domain Proteins/metabolism , Transcription Factors/physiology , Animals , Core Binding Factor Alpha 3 Subunit/immunology , Core Binding Factor Alpha 3 Subunit/metabolism , DNA Helicases/immunology , DNA Helicases/metabolism , Female , Gene Expression Profiling , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Nuclear Proteins/immunology , Nuclear Proteins/metabolism , T-Box Domain Proteins/genetics , Transcription Factors/immunology , Transcription Factors/metabolism
5.
Am J Pathol ; 188(2): 392-403, 2018 02.
Article in English | MEDLINE | ID: mdl-29137952

ABSTRACT

The pro-chondrogenic function of runt-related transcription factor 2 (Runx2) was previously considered to be dependent on direct binding with the promoter of Indian hedgehog (Ihh)-the major regulator of chondrocyte differentiation, proliferation, and maturation. The authors' previous studies identified neural EGFL like 1 (Nell-1) as a Runx2-responsive growth factor for chondrogenic differentiation and maturation. In this study, it was further revealed that the pro-chondrogenic activities of Nell-1 also rely on Ihh signaling, by showing: i) Nell-1 significantly elevated Ihh signal transduction; ii) Nell-1 deficiency markedly reduced Ihh activation in chondrocytes; and iii) Nell-1-stimulated chondrogenesis was significantly reduced by the specific hedgehog inhibitor cyclopamine. Importantly, the authors demonstrated that Nell-1-responsive Ihh signaling and chondrogenic differentiation extended to Runx2-/- models in vitro and in vivo. In Runx2-/- chondrocytes, Nell-1 stimulated the expression and signal transduction of Runx3, another transcription factor required for complete chondrogenic differentiation and maturation. Furthermore, knocking down Runx3 in Runx2-/- chondrocytes abolished Nell-1's stimulation of Ihh-associated molecule expression, which validates Runx3 as a major mediator of Nell-1-stimulated Ihh activation. For the first time, the Runx2→Nell-1→Runx3→Ihh signaling cascade during chondrogenic differentiation and maturation has been identified as an alternative, but critical, pathway for Runx2 to function as a pro-chondrogenic molecule via Nell-1.


Subject(s)
Calcium-Binding Proteins/physiology , Chondrocytes/physiology , Core Binding Factor Alpha 1 Subunit/physiology , Glycoproteins/physiology , Hedgehog Proteins/physiology , Animals , Cartilage/cytology , Cartilage/physiology , Cell Differentiation/physiology , Cells, Cultured , Chondrocytes/cytology , Chondrogenesis/physiology , Core Binding Factor Alpha 1 Subunit/deficiency , Core Binding Factor Alpha 3 Subunit/physiology , Mice, Knockout , Signal Transduction/physiology
6.
Sci Rep ; 7(1): 17947, 2017 12 20.
Article in English | MEDLINE | ID: mdl-29263341

ABSTRACT

Knowledge concerning expression and function of Suppression of Tumorigenicity 2 (ST2) in chondrocytes is at present, limited. Analysis of murine growth plates and ATDC5 chondrocytes indicated peak expression of the ST2 transmembrane receptor (ST2L) and soluble (sST2) isoforms during the hypertrophic differentiation concomitant with the expression of the hypertrophic markers Collagen X (Col X), Runx2 and MMP-13. Gain- and loss-of-function experiments in ATDC5 and primary human growth plate chondrocytes (PHCs), confirmed regulation of ST2 by the key transcription factor Runx2, indicating ST2 to be a novel Runx2 target. ST2 knock-out mice (ST2-/-) exhibited noticeable hypertrophic zone (HZ) reduction in murine growth plates, accompanied by lower expression of Col X and Osteocalcin (OSC) compared to wild-type (WT) mice. Likewise, ST2 knockdown resulted in decreased Col X expression and downregulation of OSC and Vascular Endothelial Growth Factor (VEGF) in ATDC5 cells. The ST2 suppression was also associated with upregulation of the proliferative stage markers Sox9 and Collagen II (Col II), indicating ST2 to be a new regulator of ATDC5 chondrocyte differentiation. Runx3 was, furthermore, identified as a novel Runx2 target in chondrocytes. This study suggests that Runx2 mediates ST2 and Runx3 induction to cooperatively regulate hypertrophic differentiation of ATDC5 chondrocytes.


Subject(s)
Chondrocytes/metabolism , Core Binding Factor Alpha 1 Subunit/metabolism , Core Binding Factor Alpha 3 Subunit/metabolism , Interleukin-1 Receptor-Like 1 Protein/metabolism , Animals , Cell Differentiation , Cell Line , Child , Child, Preschool , Chondrocytes/pathology , Core Binding Factor Alpha 1 Subunit/physiology , Core Binding Factor Alpha 3 Subunit/physiology , Female , Humans , Hypertrophy , Immunoblotting , Infant , Interleukin-1 Receptor-Like 1 Protein/physiology , Male , Mice , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction
7.
Oncogene ; 36(50): 6884-6894, 2017 12 14.
Article in English | MEDLINE | ID: mdl-28846108

ABSTRACT

The restriction (R)-point decision is fundamental to normal differentiation and the G1-S transition, and the decision-making machinery is perturbed in nearly all cancer cells. The mechanisms underlying the cellular context-dependent R-point decision remain poorly understood. We found that the R-point was dysregulated in Runx3-/-mouse embryonic fibroblasts (MEFs), which formed tumors in nude mice. Ectopic expression of Runx3 restored the R-point and abolished the tumorigenicity of Runx3-/-MEFs and K-Ras-activated Runx3-/-MEFs (Runx3-/-;K-RasG12D/+). During the R-point, Runx3 transiently formed a complex with pRb and Brd2 and induced Cdkn1a (p21Waf1/Cip1/Sdi1; p21), a key regulator of the R-point transition. Cyclin D-CDK4/6 promoted dissociation of the pRb-Runx3-Brd2 complex, thus turning off p21 expression. However, cells harboring oncogenic K-Ras maintained the pRb-Runx3-Brd2 complex and p21 expression even after introduction of Cyclin D1. Thus, Runx3 plays a critical role in R-point regulation and defense against cellular transformation.


Subject(s)
Cell Transformation, Neoplastic , Core Binding Factor Alpha 3 Subunit/physiology , Animals , Carcinogenesis , Cyclin-Dependent Kinase Inhibitor p21/genetics , Genes, ras , HEK293 Cells , Humans , Mice , Protein Serine-Threonine Kinases/physiology , Retinoblastoma Protein/physiology , Transcription Factors
8.
Leukemia ; 31(10): 2219-2227, 2017 10.
Article in English | MEDLINE | ID: mdl-28119527

ABSTRACT

RUNX3, runt-domain transcription factor, is a master regulator of gene expression in major developmental pathways. It acts as a tumor suppressor in many cancers but is oncogenic in certain tumors. We observed upregulation of RUNX3 mRNA and protein expression in nasal-type extranodal natural killer (NK)/T-cell lymphoma (NKTL) patient samples and NKTL cell lines compared to normal NK cells. RUNX3 silenced NKTL cells showed increased apoptosis and reduced cell proliferation. Potential binding sites for MYC were identified in the RUNX3 enhancer region. Chromatin immunoprecipitation-quantitative PCR revealed binding activity between MYC and RUNX3. Co-transfection of the MYC expression vector with RUNX3 enhancer reporter plasmid resulted in activation of RUNX3 enhancer indicating that MYC positively regulates RUNX3 transcription in NKTL cell lines. Treatment with a small-molecule MYC inhibitor (JQ1) caused significant downregulation of MYC and RUNX3, leading to apoptosis in NKTL cells. The growth inhibition resulting from depletion of MYC by JQ1 was rescued by ectopic MYC expression. In summary, our study identified RUNX3 overexpression in NKTL with functional oncogenic properties. We further delineate that MYC may be an important upstream driver of RUNX3 upregulation and since MYC is upregulated in NKTL, further study on the employment of MYC inhibition as a therapeutic strategy is warranted.


Subject(s)
Cell Transformation, Neoplastic/genetics , Core Binding Factor Alpha 3 Subunit/physiology , Gene Expression Regulation, Neoplastic , Lymphoma, Extranodal NK-T-Cell/genetics , Nose Neoplasms/genetics , Proto-Oncogene Proteins c-myc/physiology , Transcription, Genetic/genetics , Apoptosis , Azepines/pharmacology , Binding Sites , Cell Division , Cell Line, Tumor , Core Binding Factor Alpha 3 Subunit/antagonists & inhibitors , Core Binding Factor Alpha 3 Subunit/genetics , Enhancer Elements, Genetic , Genes, Reporter , Genetic Vectors , Humans , Lymphoma, Extranodal NK-T-Cell/etiology , Lymphoma, Extranodal NK-T-Cell/metabolism , Lymphoma, Extranodal NK-T-Cell/pathology , Molecular Targeted Therapy , Nose Neoplasms/etiology , Nose Neoplasms/metabolism , Nose Neoplasms/pathology , Protein Interaction Mapping , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , RNA Interference , RNA, Small Interfering/genetics , Recombinant Fusion Proteins/metabolism , Triazoles/pharmacology , Up-Regulation
9.
Nucleic Acids Res ; 45(5): 2368-2383, 2017 03 17.
Article in English | MEDLINE | ID: mdl-27903901

ABSTRACT

ChIP-seq performed on lymphoblastoid cell lines (LCLs), expressing epitope-tagged EBNA3A, EBNA3B or EBNA3C from EBV-recombinants, revealed important principles of EBNA3 binding to chromatin. When combined with global chromatin looping data, EBNA3-bound loci were found to have a singular character, each directly associating with either EBNA3-repressed or EBNA3-activated genes, but not with both. EBNA3A and EBNA3C showed significant association with repressed and activated genes. Significant direct association for EBNA3B loci could only be shown with EBNA3B-repressed genes. A comparison of EBNA3 binding sites with known transcription factor binding sites in LCL GM12878 revealed substantial co-localization of EBNA3s with RUNX3-a protein induced by EBV during B cell transformation. The beta-subunit of core binding factor (CBFß), that heterodimerizes with RUNX3, could co-immunoprecipitate robustly EBNA3B and EBNA3C, but only weakly EBNA3A. Depletion of either RUNX3 or CBFß with lentivirus-delivered shRNA impaired epitope-tagged EBNA3B and EBNA3C binding at multiple regulated gene loci, indicating a requirement for CBF heterodimers in EBNA3 recruitment during target-gene regulation. ShRNA-mediated depletion of CBFß in an EBNA3C-conditional LCL confirmed the role of CBF in the regulation of EBNA3C-induced and -repressed genes. These results reveal an important role for RUNX3/CBF during B cell transformation and EBV latency that was hitherto unexplored.


Subject(s)
Core Binding Factors/metabolism , Epstein-Barr Virus Nuclear Antigens/metabolism , Gene Expression Regulation , Binding Sites , Cell Line , Chromatin/chemistry , Chromatin/metabolism , Chromatin Immunoprecipitation , Core Binding Factor Alpha 3 Subunit/metabolism , Core Binding Factor Alpha 3 Subunit/physiology , Core Binding Factor beta Subunit/metabolism , Core Binding Factors/physiology , Enhancer Elements, Genetic , Genome, Human , Humans , Transcription Factors/metabolism , Transcription Initiation Site
10.
J Reprod Dev ; 62(5): 479-486, 2016 Oct 18.
Article in English | MEDLINE | ID: mdl-27301496

ABSTRACT

We previously demonstrated that the Runx3 transcription factor is expressed in the hypothalami, pituitaries, and ovaries of mice, and that Runx3 knockout (Runx3-/-) mice are anovulatory and their uteri are atrophic. Runx3 mRNA expression was detected in the granulosa cells of ovarian follicles, and in the anteroventral periventricular nucleus (AVPV) and arcuate nucleus (ARC). In the present study, we examined the effects of Runx3 knockout on the gene expression of enzymes associated with steroidogenesis. We found decreased Cyp11a1 mRNA expression in Runx3-/- mouse ovaries compared with that in wild-type (wt) mouse ovaries at the age of 8 weeks. In situ hybridization analysis showed that the percentages of Cyp11a1 mRNA-expressing theca cells in follicles of Runx3-/- mice were decreased compared with those of wt mice. In accord with the alterations in Runx3-/- mouse ovaries, Kiss1 mRNA levels in ARC were increased, whereas mRNA levels of kisspeptin in AVPV were decreased, and gonadotropin-releasing hormone in the preoptic area and follicle-stimulating hormone ß subunit gene were increased in Runx3-/- mice. Following an ovarian transplantation experiment between Runx3-/- mice and wt mice, corpora lutea were observed when ovaries from Runx3-/- mice were transplanted into wt mice, but not when those from wt mice were transplanted into Runx3-/- mice, suggesting that Runx3 in the hypothalamo-pituitary system may drive gonadotropin release to induce ovulation in the ovary. These findings indicate that Runx3 plays a crucial role in the hypothalamo-pituitary-gonadal axis.


Subject(s)
Core Binding Factor Alpha 3 Subunit/physiology , Ovary/physiology , Ovulation/physiology , Animals , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Core Binding Factor Alpha 3 Subunit/genetics , Female , Gonadotropins/metabolism , Granulosa Cells/cytology , Hypothalamo-Hypophyseal System , Hypothalamus/metabolism , Hypothalamus, Anterior/physiology , In Situ Hybridization , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , Ovarian Follicle/physiology , RNA, Messenger/metabolism , Steroids/chemistry , Transcription Factors/metabolism
11.
Leuk Res ; 39(12): 1405-13, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26475207

ABSTRACT

Internal tandem duplication (ITD) mutations of the FLT3 gene (FLT3-ITD) are well known to correlate with a poor prognosis in acute myeloid leukemia (AML). We previously reported that FLT3-ITD confers resistance to cytosine arabinoside (Ara-C), a key cytotoxic agent in AML treatments. In order to elucidate the detailed molecular mechanisms underlying the Ara-C resistance induced by FLT3-ITD, we performed a microarray gene expression analysis of the human leukemic cell line K562 transduced with FLT3-ITD (K562/FLT3-ITD) and identified RUNX3 as a downstream target of FLT3-ITD. The transcriptional induction of the RUNX3 expression by FLT3-ITD was noted on a Luciferase assay. The knockdown of the RUNX3 expression in the K562/FLT3-ITD cells increased the sensitivity to Ara-C, and the exogenous expression of RUNX3 per se resulted in the enhancement of Ara-C resistance in the K562 cells. A relationship between the FLT3-ITD-induced RUNX3 expression and Ara-C resistance was also observed in AML cells with an endogenous FLT3-ITD expression. Collectively, these findings demonstrate that RUNX3 is a prerequisite for Ara-C resistance via FLT3-ITD signaling.


Subject(s)
Core Binding Factor Alpha 3 Subunit/physiology , Cytarabine/pharmacology , Drug Resistance, Neoplasm/physiology , Leukemia, Myeloid, Acute/pathology , fms-Like Tyrosine Kinase 3/physiology , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Fusion Proteins, bcr-abl/metabolism , Humans , K562 Cells , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , RNA Interference , RNA, Small Interfering/genetics , Recombinant Fusion Proteins/metabolism , Tandem Repeat Sequences , Transcription, Genetic , Transduction, Genetic , fms-Like Tyrosine Kinase 3/genetics
12.
Lasers Med Sci ; 30(2): 499-507, 2015 Feb.
Article in English | MEDLINE | ID: mdl-23700080

ABSTRACT

Photodynamic therapy (PDT) with photosensitizer is one of the promising modalities for cancer treatment. For clinical use of PDT, screening process should be preceded to enhance sensitivity to PDT. Thus, we investigated a molecular biomarker to determine the sensitivity to pheophorbide a (Pa)-PDT in immortalized human oral keratinocytes (IHOK) and oral squamous cell carcinoma (OSCC) cell lines. Two IHOK and several OSCC cell lines were used. After Pa-PDT, cell viability was reduced by more than 50%, and reactive oxygen species were generated in IHOK and OSCC cell lines. Additionally, apoptosis occurred in PDT-treated cells. IHOK(S) and IHOK(P), the two IHOK cell lines derived from the same source, showed a difference in cytotoxicity after Pa-PDT. To explain this difference in cytotoxicity, we looked at the expression of Wnt signaling-related genes in these two cell lines, for the morphology of IHOK(S) which was spindle like and elongated and distinct from IHOK(P) and the parent cell. Among the relevant genes, runt-related transcription factor 3 (RUNX3), an apoptosis-related gene, was selected as a potential marker that confers sensitivity to PDT. We found that the cytotoxicity by Pa-PDT was proportional to RUNX3 expression in OSCC cell lines. Additionally, knockdown of RUNX3 expression reduced cytotoxicity by Pa-PDT, suggesting that RUNX3 might be a biomarker to determine sensitivity to Pa-PDT. This was the first study to find a new target molecule that enhances Pa-PDT effects in IHOK and OSCC cell lines. Hence, the development of a PDT-dependent biomarker could provide a novel approach to improve the effects of PDT on oral precancerous and cancerous lesions.


Subject(s)
Carcinoma, Squamous Cell/therapy , Chlorophyll/analogs & derivatives , Core Binding Factor Alpha 3 Subunit/physiology , Mouth Neoplasms/therapy , Photochemotherapy/methods , Apoptosis/drug effects , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Chlorophyll/chemistry , Gene Expression Regulation, Neoplastic , Humans , Keratinocytes/metabolism , Mouth Neoplasms/metabolism , Photosensitizing Agents/therapeutic use , Reactive Oxygen Species/metabolism , Signal Transduction , Wnt Proteins/metabolism
13.
Nat Immunol ; 15(7): 638-45, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24880459

ABSTRACT

Lineage fate in the thymus is determined by mutually exclusive expression of the transcription factors ThPOK and Runx3, with ThPOK imposing the CD4(+) lineage fate and Runx3 promoting the CD8(+) lineage fate. While it is known that cytokine signals induce thymocytes to express Runx3, it is not known how ThPOK prevents thymocytes from expressing Runx3 and adopting the CD8(+) lineage fate, nor is it understood why ThPOK itself imposes the CD4(+) lineage fate on thymocytes. We now report that genes encoding members of the SOCS (suppressor of cytokine signaling) family are critical targets of ThPOK and that their induction by ThPOK represses Runx3 expression and promotes the CD4(+) lineage fate. Thus, induction of SOCS-encoding genes is the main mechanism by which ThPOK imposes the CD4(+) lineage fate in the thymus.


Subject(s)
CD4-Positive T-Lymphocytes/physiology , Cell Lineage , Core Binding Factor Alpha 3 Subunit/physiology , Suppressor of Cytokine Signaling Proteins/genetics , Transcription Factors/physiology , Animals , CD8-Positive T-Lymphocytes/physiology , Mice , Mice, Inbred C57BL
15.
Nat Immunol ; 15(7): 646-656, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24836425

ABSTRACT

The transcription factors TCF-1 and LEF-1 are essential for early T cell development, but their roles beyond the CD4(+)CD8(+) double-positive (DP) stage are unknown. By specific ablation of these factors in DP thymocytes, we demonstrated that deficiency in TCF-1 and LEF-1 diminished the output of CD4(+) T cells and redirected CD4(+) T cells to a CD8(+) T cell fate. The role of TCF-1 and LEF-1 in the CD4-versus-CD8 lineage 'choice' was mediated in part by direct positive regulation of the transcription factor Th-POK. Furthermore, loss of TCF-1 and LEF-1 unexpectedly caused derepression of CD4 expression in T cells committed to the CD8(+) lineage without affecting the expression of Runx transcription factors. Instead, TCF-1 physically interacted with Runx3 to cooperatively silence Cd4. Thus, TCF-1 and LEF-1 adopted distinct genetic 'wiring' to promote the CD4(+) T cell fate and establish CD8(+) T cell identity.


Subject(s)
CD4 Antigens/physiology , CD4-Positive T-Lymphocytes/physiology , CD8-Positive T-Lymphocytes/physiology , Core Binding Factor Alpha 3 Subunit/physiology , Lymphoid Enhancer-Binding Factor 1/physiology , T Cell Transcription Factor 1/physiology , Transcription Factors/physiology , Animals , Cell Lineage , Female , Hepatocyte Nuclear Factor 1-alpha , Male , Mice
16.
Tumour Biol ; 35(6): 6039-46, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24622886

ABSTRACT

The aim of this study is to investigate the possible roles of runt-related transcription factor 3 (RUNX3) and ß-catenin in the carcinogenesis of sporadic colorectal tubular adenomas. The expression of the RUNX3 and ß-catenin proteins was evaluated by immunohistochemistry in 23 normal colorectal mucosa (NCM), 81 sporadic colorectal tubular adenomas with different dysplasias (SCTA-D) (mild n=33, moderate n=23, and severe n=25 dysplasia), and 48 sporadic colorectal tubular adenomas with cancerous changes (SCTA-Ca). RUNX3 methylation was assessed by methylation-specific polymerase chain reaction (MSP), combined with laser capture microdissection (LCM), in 17 NCM, 41 SCTA-D (mild n=15, moderate n=12, and severe n=14 dysplasia), and 17 SCTA-Ca tissues. Compared to NCM (82.6 %), RUNX3 in SCTA-D (54.3 %) and SCTA-Ca (27.1 %) was significantly downregulated (P<0.05). In NCM, SCTA-D, and SCTA-Ca, the incidence of positive expression for ß-catenin was 13.0, 60.5, and 79.2 %, respectively. A statistically significant difference was observed (P<0.05). RUNX3 levels were markedly higher in adenoma with mild dysplasia (75.8 %) and moderate dysplasia (60.9 %) than in adenoma with severe dysplasia (20.0 %) (both with P<0.05). Likewise, the expression of ß-catenin in severe dysplasia adenoma was 84.0 %, which was significantly higher than that in mild dysplasia adenoma (39.4 %). An inverse correlation was found between the protein expression of RUNX3 and ß-catenin in SCTA-D and SCTA-Ca (P<0.05). MSP results showed that RUNX3 methylation in NCM, SCTA-D, and SCTA-Ca was 5.9, 17.1, and 41.2 %, respectively, with a statistically significant difference between NCM and SCTA-Ca (P<0.05). However, no significant difference of RUNX3 methylation was observed among different dysplasia groups. RUNX3 and ß-catenin play important roles in the carcinogenesis of sporadic colorectal tubular adenomas. In addition, hypermethylation of RUNX3 can downregulate its expression.


Subject(s)
Adenoma/etiology , Colorectal Neoplasms/etiology , Core Binding Factor Alpha 3 Subunit/physiology , beta Catenin/physiology , Adenoma/chemistry , Adenoma/pathology , Cell Transformation, Neoplastic , Colorectal Neoplasms/chemistry , Colorectal Neoplasms/pathology , Core Binding Factor Alpha 3 Subunit/analysis , Core Binding Factor Alpha 3 Subunit/genetics , DNA Methylation , Humans , Promoter Regions, Genetic , Wnt Signaling Pathway , beta Catenin/analysis
17.
Oncogene ; 33(11): 1458-67, 2014 Mar 13.
Article in English | MEDLINE | ID: mdl-23542169

ABSTRACT

RUNX3 is silenced by histone modification and hypoxia-inducible factor (HIF)-1α is stabilized under hypoxia, but little is known of cross-talk between RUNX3 and HIF-1α under hypoxia. In the present study, the authors investigated the effect of RUNX3 on HIF-1α stability in gastric cancer cells. RUNX3 overexpression was found to downregulate HIF-1α stability under normoxic and hypoxic conditions. Furthermore, the activity of a luciferase reporter containing five copies of vascular endothelial growth factor (VEGF) promoter hypoxia-responsive element (5 × HRE) and the amount of secreted VEGF, were diminished in RUNX3-expressing but increased in RUNX3-knockdown cells. When expression of RUNX3 was recovered using epigenetic reagents the expressions of HIF-1α and VEGF were clearly suppressed under hypoxic conditions. RUNX3 also significantly attenuated the half-life of HIF-1α protein, and induced the cytosolic localization and ubiquitination of HIF-1α. In addition, RUNX3 directly interacted with the C-terminal activation domain of HIF-1α and prolyl hydroxylase (PHD) 2 and enhanced the interaction between HIF-1α and PHD2, which potentiated proline hydroxylation and promoted the degradation of HIF-1α. Furthermore, RUNX3 overexpression significantly inhibited hypoxia-induced angiogenesis in vitro and in vivo. Taken together, these results suggest that RUNX3 destabilizes HIF-1α protein by promoting the proline hydroxylation of HIF-1α through binding to HIF-1α/PHD2. RUNX3 appears to be a novel suppressor of HIF-1α and of hypoxia-mediated angiogenesis in gastric cancer cells.


Subject(s)
Core Binding Factor Alpha 3 Subunit/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Prolyl Hydroxylases/metabolism , Stomach Neoplasms/metabolism , Cell Hypoxia/genetics , Cell Line, Tumor , Humans , Hydroxylation , RNA, Messenger/genetics , Stomach Neoplasms/enzymology , Stomach Neoplasms/pathology , Ubiquitination , Vascular Endothelial Growth Factor A/genetics
18.
Curr Top Microbiol Immunol ; 373: 113-29, 2014.
Article in English | MEDLINE | ID: mdl-23612990

ABSTRACT

CD4(+) helper and CD8(+) cytotoxic T cells, two major subsets of αßTCR expressing lymphocytes, are differentiated from common precursor CD4(+)CD8(+) double-positive (DP) thymocytes. Bifurcation of the CD4(+)/CD8(+) lineages in the thymus is a multilayered process and is thought to culminate in a loss of developmental plasticity between these functional subsets. Advances in the last decade have deepened our understanding of the transcription control mechanisms governing CD4 versus CD8 lineage commitment. Reciprocal expression and antagonistic interplay between two transcription factors, ThPOK and Runx3, is crucial for driving thymocyte decisions between these two cell fates. Here, we first focus on the regulation of ThPOK expression and its role in directing helper T cell development. We then discuss a novel aspect of the ThPOK/Runx3 axis in modifying CD4(+) T cell function upon exposure to gut microenvironment.


Subject(s)
CD4-Positive T-Lymphocytes/physiology , CD8-Positive T-Lymphocytes/physiology , Cell Lineage , Animals , Core Binding Factor Alpha 3 Subunit/physiology , DNA-Binding Proteins/physiology , Humans , Intestines/immunology , Transcription Factors/physiology
19.
Trends Cell Biol ; 23(7): 305-10, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23511112

ABSTRACT

Neurons exhibit great size differences, and must coordinate biosynthesis rates in cell bodies with the growth needs of different lengths of axons. Classically, axon growth has been viewed mainly as a consequence of extrinsic influences. However, recent publications have proposed at least two different intrinsic axon growth-control mechanisms. We suggest that these mechanisms form part of a continuum of axon growth-control mechanisms, wherein initial growth rates are pre-programmed by transcription factor levels, and subsequent elongating growth is dependent on feedback from intrinsic length-sensing enabled by bidirectional motor-dependent oscillating signals. This model might explain intrinsic limits on elongating neuronal growth and provides a mechanistic framework for determining the connections between genome expression and cellular growth rates in neurons.


Subject(s)
Cell Proliferation , Cell Size , Neurons/physiology , Signal Transduction/physiology , Animals , Axons/physiology , Core Binding Factor Alpha 3 Subunit/physiology , Dyneins/physiology , Humans , Kinesins/physiology , Models, Neurological , Neurites/physiology , Neurons/cytology
20.
Oncogene ; 32(12): 1488-96, 2013 Mar 21.
Article in English | MEDLINE | ID: mdl-22580604

ABSTRACT

Emerging evidence demonstrates that RUNX3 is a tumor suppressor in breast cancer. Inactivation of RUNX3 in mice results in spontaneous mammary gland tumors, and decreased or silenced expression of RUNX3 is frequently found in breast cancer cell lines and human breast cancer samples. However, the underlying mechanism for initiating RUNX3 inactivation in breast cancer remains elusive. Here, we identify prolyl isomerase Pin1, which is often overexpressed in breast cancer, as a key regulator of RUNX3 inactivation. In human breast cancer cell lines and breast cancer samples, expression of Pin1 inversely correlates with the expression of RUNX3. In addition, Pin1 recognizes four phosphorylated Ser/Thr-Pro motifs in RUNX3 via its WW domain. Binding of Pin1 to RUNX3 suppresses the transcriptional activity of RUNX3. Furthermore, Pin1 reduces the cellular levels of RUNX3 in an isomerase activity-dependent manner by inducing the ubiquitination and proteasomal degradation of RUNX3. Knocking down Pin1 enhances the cellular levels and transcriptional activity of RUNX3 by inhibiting the ubiquitination and degradation of RUNX3. Our results identify Pin1 as a new regulator of RUNX3 inactivation in breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Core Binding Factor Alpha 3 Subunit/physiology , Peptidylprolyl Isomerase/physiology , Amino Acid Motifs , Breast Neoplasms/pathology , Cell Line, Tumor , Core Binding Factor Alpha 3 Subunit/analysis , Core Binding Factor Alpha 3 Subunit/chemistry , Core Binding Factor Alpha 3 Subunit/genetics , Down-Regulation , Female , Humans , NIMA-Interacting Peptidylprolyl Isomerase , Peptidylprolyl Isomerase/analysis , Phosphorylation , Protein Stability , Tumor Suppressor Proteins , Ubiquitination
SELECTION OF CITATIONS
SEARCH DETAIL
...