Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add filters








Year range
1.
Chinese Medical Journal ; (24): 1572-1581, 2019.
Article in English | WPRIM | ID: wpr-802556

ABSTRACT

Background@#Our previous studies have shown that regulatory factor X5 (RFX5), a classical transcription regulator of MHCII genes, was obviously overexpressed in hepatocellular carcinoma (HCC) tumors. However, the role of RFX5 in the carcinogenesis and progress of HCC remains unknown. This study aimed to reveal its biological significance and the underlying mechanism in HCC.@*Methods@#RFX5 mRNA expression level and copy number variation in HCC tumors and cell lines were determined by analyzing deposited data sets in the Cancer Genome Atlas and Gene Expression Omnibus database. The biological significance of RFX5 in HCC was investigated by monitoring the colony formation and subcutaneous tumor growth capacity when RFX5 was silenced with lentiviral short hairpin RNA and CRISPR/Cas9 system in HCC cell lines. The downstream gene transcriptionally activated by RFX5 in HCC cells was determined by chromatin immunoprecipitation and luciferase reporter assay. The involvement of tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein theta (YWHAQ) in HCC development was further determined by performing colony formation rescue assay and subcutaneous tumor growth rescue experiment. The association of YWHAQ with recurrence-free survival of patients with HCC was assessed by Kaplan-Meier analysis. Moreover, apoptosis level and the protein level of p53 pathway were determined to reveal the mechanism of RFX5 in driving HCC development.@*Results@#RFX5 was amplified and highly overexpressed in HCC tumor tissues compared with the corresponding non-tumor tissues. The mRNA expression level of RFX5 was significantly correlated with its DNA copy number (r = 0.4, P < 0.001). Functional study demonstrated that RFX5 was required for both clonogenic forming in vitro and subcutaneous tumor growth in vivo of HCC cells. Further study identified YWHAQ, namely 14-3-3 tau, as a key downstream transcriptional target gene of RFX5, which was tightly regulated by RFX5 in HCC. Moreover, overexpression of YWHAQ largely rescued the clonogenic growth of HCC cells that was suppressed by RFX5 knockdown. In addition, overexpression of YWHAQ in primary tumor was linked to poor prognosis of patients with HCC. These results demonstrated that YWHAQ was a downstream effector of RFX5 in HCC. Notably, RFX5-YWHAQ pathway could protect cells from apoptosis by suppressing the p53 and Bax in HCC.@*Conclusion@#RFX5 is a putative HCC driver gene that plays an important role in the development and progression of HCC by transactivating YWHAQ and suppressing apoptosis.

2.
Chinese Medical Journal ; (24): 1572-1581, 2019.
Article in English | WPRIM | ID: wpr-771229

ABSTRACT

BACKGROUND@#Our previous studies have shown that regulatory factor X5 (RFX5), a classical transcription regulator of MHCII genes, was obviously overexpressed in hepatocellular carcinoma (HCC) tumors. However, the role of RFX5 in the carcinogenesis and progress of HCC remains unknown. This study aimed to reveal its biological significance and the underlying mechanism in HCC.@*METHODS@#RFX5 mRNA expression level and copy number variation in HCC tumors and cell lines were determined by analyzing deposited data sets in the Cancer Genome Atlas and Gene Expression Omnibus database. The biological significance of RFX5 in HCC was investigated by monitoring the colony formation and subcutaneous tumor growth capacity when RFX5 was silenced with lentiviral short hairpin RNA and CRISPR/Cas9 system in HCC cell lines. The downstream gene transcriptionally activated by RFX5 in HCC cells was determined by chromatin immunoprecipitation and luciferase reporter assay. The involvement of tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein theta (YWHAQ) in HCC development was further determined by performing colony formation rescue assay and subcutaneous tumor growth rescue experiment. The association of YWHAQ with recurrence-free survival of patients with HCC was assessed by Kaplan-Meier analysis. Moreover, apoptosis level and the protein level of p53 pathway were determined to reveal the mechanism of RFX5 in driving HCC development.@*RESULTS@#RFX5 was amplified and highly overexpressed in HCC tumor tissues compared with the corresponding non-tumor tissues. The mRNA expression level of RFX5 was significantly correlated with its DNA copy number (r = 0.4, P < 0.001). Functional study demonstrated that RFX5 was required for both clonogenic forming in vitro and subcutaneous tumor growth in vivo of HCC cells. Further study identified YWHAQ, namely 14-3-3 tau, as a key downstream transcriptional target gene of RFX5, which was tightly regulated by RFX5 in HCC. Moreover, overexpression of YWHAQ largely rescued the clonogenic growth of HCC cells that was suppressed by RFX5 knockdown. In addition, overexpression of YWHAQ in primary tumor was linked to poor prognosis of patients with HCC. These results demonstrated that YWHAQ was a downstream effector of RFX5 in HCC. Notably, RFX5-YWHAQ pathway could protect cells from apoptosis by suppressing the p53 and Bax in HCC.@*CONCLUSION@#RFX5 is a putative HCC driver gene that plays an important role in the development and progression of HCC by transactivating YWHAQ and suppressing apoptosis.

3.
Journal of Experimental Hematology ; (6): 1195-1199, 2011.
Article in Chinese | WPRIM | ID: wpr-261902

ABSTRACT

To analyze hematopoietic kinetics of mouse embryonic aorta-gonad-mesonephros (AGM) region, an in vitro tissue culture method was developed in this study, partly based on previous reports. After 2 days of tissue culture, a significant number of erythro myeloid progenitors, as quantitated by colony forming assay was detected in the AGM region. Moreover, the cells from cultured E10.5 AGM region could generate 10.8 ± 3.5 colony-forming unit in spleen (CFU-S) per tissue on average. Transplantation of cultured E10.5-E11.0 AGM cells resulted in efficient (85.7% repopulated) and long-term (> 4 months) reconstitution of lethally irradiated adult recipients with remarkable chimerism [(51.12 ± 21.17)%]. The multilineage contribution of donor cells was validated by significant engraftment of myeloid and/or lymphoid cells in peripheral blood, bone marrow, spleen and thymus of recipients. Taken together, the tissue culture method can enable us to manipulate the AGM region in vitro, fulfilling a systematic evaluation of developmental kinetics of various hematopoietic precursor cells, particularly HSC, in normal and mutant mid-gestation mouse embryos.


Subject(s)
Animals , Female , Mice , Gonads , Cell Biology , Hematopoiesis , Hematopoietic Stem Cells , Cell Biology , Hematopoietic System , Embryology , Mice, Inbred C57BL , Mice, Transgenic , Tissue Culture Techniques , Methods
4.
Journal of Experimental Hematology ; (6): 1282-1285, 2010.
Article in Chinese | WPRIM | ID: wpr-332376

ABSTRACT

The anatomical location of lymphocyte ontogeny in the developing mouse embryo remains controversial. To define the site that can generate lymphocytes de novo, the intraembryonic splanchnopleura (SP) and extraembryonic yolk sac (YS) at 8.5 days postcoitum, when systemic circulation is not established, were investigated. The results indicated that in standard colony forming assay, the cells from both splanchnopleura and yolk sac formed typical myeloerythroid colonies, but their types were distinct. When cocultured with the OP9, the splanchnopleura produced B cells expressing B220, CD19 and surface IgM. Using a three-step culture protocols with the OP9 expressing Delta-like 1 as feeders, the splanchnopleura produced immature T precursor cells (CD44-/CD25+) and more mature single positive T cells (CD4+/CD8-) after 16 days of incubation. However, the yolk sac failed to generate B and T lymphocytes under identical conditions. It is concluded that prior to linked embryonic circulation, the splanchnopleura other than the yolk sac had robust lymphoid potential in vitro. In the future, more reliable evidence from novel model animals will ultimately delineate the embryonic origin of lymphocytes in vivo.


Subject(s)
Animals , Female , Mice , Pregnancy , B-Lymphocytes , Cell Biology , Cell Differentiation , Coculture Techniques , Hematopoietic Stem Cells , Cell Biology , Mice, Inbred C57BL , T-Lymphocytes , Cell Biology , Yolk Sac , Cell Biology
5.
Journal of Southern Medical University ; (12): 374-376, 2007.
Article in Chinese | WPRIM | ID: wpr-268130

ABSTRACT

<p><b>OBJECTIVE</b>To investigate the effect of tumor necrosis factor alpha (TNFalpha) on radiosensitivity of nasopharyngeal carcinoma (NPC) in relation to TNFalpha-induced cell cycle synchronization.</p><p><b>METHODS</b>The radio-resistance of a NPC cell line subclone CNE-2Z-S1 was verified by in vivo experiments and flow cytometry was performed to evaluate cell cycle synchronization in TNFalpha-treated CNE-2Z-S1 cells. The radiosensitivity of the cell synchronized CNE-2Z-S1 cells was determined by clone formation in vitro and in vivo experiment in nude mice.</p><p><b>RESULTS</b>TNFalpha was capable of inducing cell cycle arrest and synchronization of CNE-2Z-S1 cells. Pretreatment with TNFalpha remarkably enhanced the radiosensitivity of CNE-2Z-S1 in vitro, and in vivo experiments with nude mice also suggested the role of TNFalpha in enhancing the radiosensitivity of NPC.</p><p><b>CONCLUSION</b>TNFalpha can enhance the radiosensitivity of NPC cells by inducing cell cycle synchronization.</p>


Subject(s)
Animals , Humans , Mice , Cell Cycle , Radiation Effects , Cell Line, Tumor , Mice, Nude , Nasopharyngeal Neoplasms , Drug Therapy , Pathology , Radiotherapy , Radiation-Sensitizing Agents , Pharmacology , Tumor Necrosis Factor-alpha , Pharmacology , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL