1.Molecular mechanisms of TPT1-AS1 in regulating epithelial ovarian cancer cell invasion, migration, and angiogenesis by targeting the miR-324/TWIST1 axis.
Chinese Journal of Cellular and Molecular Immunology 2025;41(6):536-543
Objective To explore the mechanism of TPT1-AS1 targeting miR-324/TWIST1 axis to regulate the proliferation, invasion, migration and angiogenesis of epithelial ovarian cancer (EOC) cells, thereby affecting ovarian cancer (OC) progression. Methods RT-qPCR was used to detect the expression of TPT1-AS1 and miR-324 in 29 OC lesions and adjacent tissue samples. The two OC cell models of TPT1-AS1 overexpression and miRNA324 knockdown were constructed, and the cell proliferation, invasion and migration abilities were detected by CCK-8, TranswellTM and scratch test. Western blot analysis was used to detect the protein expression levels of TWIST1, epithelial cadherin (E-cadherin), Vimentin, and vascular endothelial growth factor A (VEGF-A) in OC cells. Fluorescence in situ hybridization (FISH) and RNA pull-down experiments were used to verify the interaction between TPT1-AS1 and miR-324. Immunohistochemistry and Targetscan bioinformatics analysis were used to verify the negative regulatory role of miR-324 in the epithelial-mesenchymal transition (EMT) process. Results The TPT1-AS1 expression was significantly higher in OC tissues than that in para-cancerous tissues, while the miR-324 expression was significantly lower. In SKOV3 cells with TPT1-AS1 overexpression, the miR-324 expression decreased significantly, and TPT1-AS1 was negatively correlated with miR-324. It was also found that TPT1-AS1 and miR-324 were co-expressed in OC cells, and there was a direct binding relationship between them. Down-regulation of miR-324 significantly promoted the proliferation, invasion and migration of SKOV3 cells. Further studies revealed that miR-324 had a binding site at the 3'-UTR end of the TWIST1, a key transcription factor for EMT. Inhibiting miR-324 expression increased the transcription level of TWIST1, leading to a decrease in E-cadherin protein expression and an increase in Vimentin protein expression. Additionally, the downregulation of miR-324 resulted in an increased expression level of VEGF-A protein, which in turn enhanced angiogenesis of OC. Conclusion TPT1-AS1 promotes EOC cell proliferation, invasion, migration and angiogenesis by negatively regulating the miR-324/TWIST1 axis, thus promoting the development of OC. These findings provide new potential targets for the diagnosis and treatment of OC.
Humans
;
MicroRNAs/metabolism*
;
Female
;
Cell Movement/genetics*
;
Ovarian Neoplasms/blood supply*
;
Twist-Related Protein 1/metabolism*
;
Cell Line, Tumor
;
Neovascularization, Pathologic/genetics*
;
Neoplasm Invasiveness
;
Carcinoma, Ovarian Epithelial/metabolism*
;
Nuclear Proteins/metabolism*
;
Cell Proliferation/genetics*
;
Epithelial-Mesenchymal Transition/genetics*
;
Gene Expression Regulation, Neoplastic
;
RNA, Long Noncoding/metabolism*
;
Cadherins/genetics*
;
Vascular Endothelial Growth Factor A/genetics*
;
Vimentin/genetics*
;
Angiogenesis
2.Role of Brg1 in regulating the Wnt/β-catenin signaling pathway in a bronchopulmonary dysplasia model.
Ling GUAN ; Mao-Zhu XU ; Yao-Zheng LING ; Li-Li YANG ; Ling-Huan ZHANG ; Sha LIU ; Wen-Jing ZOU ; Zhou FU
Chinese Journal of Contemporary Pediatrics 2025;27(6):731-739
OBJECTIVES:
To investigate the role and mechanism of Brahma-related gene 1 (Brg1) in regulating the Wnt/β-catenin signaling pathway in a bronchopulmonary dysplasia (BPD) model.
METHODS:
Wild-type C57BL/6 and Brg1f1/f1 mice were randomly divided into four groups: wild-type control, wild-type BPD, Brg1f1/f1 control, and Brg1f1/f1 BPD (n=5 each). Immortalized mouse pulmonary alveolar type 2 cells (imPAC2) were cultured, and Brg1 gene was knocked down using lentivirus transfection technology. Cells were divided into three groups: control, empty vector, and Brg1 knockdown. Hematoxylin and eosin staining and immunofluorescence were used to detect pathological changes in mouse lung tissue. Western blot and real-time fluorescent quantitative PCR were used to measure Brg1 protein and mRNA expression levels in mouse lung tissue. Western blot and immunofluorescence were used to detect the expression of homeodomain-containing protein homeobox (HOPX), surfactant protein C (SPC), and Wnt/β-catenin signaling pathway proteins in mouse lung tissue and imPAC2 cells. The CCK8 assay was used to assess the proliferation of imPAC2 cells, and co-immunoprecipitation was performed to verify the interaction between Brg1 and β-catenin proteins in imPAC2 cells.
RESULTS:
Compared to the Brg1f1/f1 control group and wild-type BPD group, the Brg1f1/f1 BPD group showed increased alveolar diameter and SPC protein expression, and decreased relative density of pulmonary vasculature and HOPX protein expression (P<0.05). Compared to the control group, the Brg1 knockdown group showed increased cell proliferation ability, protein expression levels of SPC, Wnt5a and β-catenin, and β-catenin protein fluorescence intensity, along with decreased HOPX protein expression (P<0.05). An interaction between Brg1 and β-catenin proteins was confirmed.
CONCLUSIONS
The Brg1 gene may promote the proliferation of alveolar type 2 epithelial cells by regulating the Wnt/β-catenin signaling pathway, thus influencing the occurrence and development of BPD.
Animals
;
DNA Helicases/genetics*
;
Transcription Factors/genetics*
;
Wnt Signaling Pathway/physiology*
;
Nuclear Proteins/genetics*
;
Mice
;
Bronchopulmonary Dysplasia/etiology*
;
Mice, Inbred C57BL
;
beta Catenin/physiology*
;
Disease Models, Animal
;
Cell Proliferation
;
Lung/pathology*
;
Male
3.NIP7 upregulates the expression of ubiquitin-conjugating enzyme E2 C to promote tumor growth in anaplastic thyroid cancer.
Yingying GONG ; Ziwen FANG ; Yixuan WANG ; Minghua GE ; Zongfu PAN
Journal of Zhejiang University. Medical sciences 2025;54(3):372-381
OBJECTIVES:
To investigate the role of nucleolar pre-rRNA processing protein NIP7 (NIP7) in maintaining the malignant phenotype of anaplastic thyroid cancer (ATC) and its molecular mechanisms.
METHODS:
NIP7 expression in ATC tissues and its gene knock-out effects in ATC cells were analyzed using gene expression microarray (GSE33630), proteome database (IPX0008941000) and the Dependency Map database, respectively. Expression and localization of NIP7 in normal thyroid cells, papillary thyroid cancer cells, and ATC cells were detected by Western blotting. Small interfering RNA (siRNA) was transfected into ATC cells, and the knockdown efficiency of NIP7 was detected by quantitative reverse transcription polymerase chain reaction (qRT-PCR) and Western blotting. Cell proliferation was assessed by CCK-8 assay, colony formation was evaluated by colony formation assay, and tumor growth was assessed by xenograft tumor model in nude mice. SUnSET (surface sensing of translation) assay combined with co-immunoprecipitation were employed to evaluate the effect of NIP7 silencing on ubiquitin-conjugating enzyme E2 C (UBE2C) translation. Finally, gene set enrichment analysis was used to identify shared pathways of NIP7 and UBE2C, which were validated by qRT-PCR.
RESULTS:
Compared with normal tissues and papillary thyroid cancer, NIP7 was significantly upregulated in ATC tissues, and had a gene knock-out fitness effect on different ATC cell lines. The relative protein levels of NIP7 in ATC cells were significantly higher than those in normal thyroid follicular cells, and the protein was mainly expressed in the nucleus. NIP7 silencing significantly inhibited cell proliferation and reduced colony formation. Xenograft tumor model showed that NIP7 knockdown significantly slowed down the growth of ATC xenograft, and the tumor volume and weight were significantly lower than those in the control group (all P<0.05). NIP7 silencing downregulated the protein level of UBE2C, but did not affect the expression of UBE2C mRNA. Compared to the control group, UBE2C silencing significantly inhibited ATC cells proliferation (P<0.01) and colony formation (P<0.05). UBE2C overexpression reversed the proliferation-inhibitory effect induced by NIP7 silencing (P<0.01). Gene set enrichment analysis indicated that NIP7 and UBE2C were both involved in DNA replication. NIP7 or UBE2C silencing could significantly downregulate the expression levels of DNA polymerase epsilon, catalytic subunit 2 and replication factor C4 in DNA replication pathway.
CONCLUSIONS
NIP7 promotes ATC tumor growth by upregulating UBE2C to mediate DNA replication.
Humans
;
Ubiquitin-Conjugating Enzymes/genetics*
;
Thyroid Neoplasms/genetics*
;
Thyroid Carcinoma, Anaplastic/genetics*
;
Animals
;
Mice, Nude
;
Mice
;
Cell Line, Tumor
;
Cell Proliferation
;
Up-Regulation
;
RNA, Small Interfering/genetics*
;
Nuclear Proteins/metabolism*
;
Gene Expression Regulation, Neoplastic
4.Expression and Biological Function of SPOP in Acute Myeloid Leukemia.
Xue-Ying WAN ; Jing XU ; Xiao-Li LIU ; Hong-Wei WANG
Journal of Experimental Hematology 2025;33(1):32-38
OBJECTIVE:
To study the expression of SPOP in patients with acute myeloid leukemia (AML) and its effect on proliferation, apoptosis and cycle of AML cells.
METHODS:
RT-qPCR was used to detect the expression of SPOP mRNA in bone marrow samples of patients with newly diagnosed AML and normal controls. The stable overexpression of SPOP in AML cell lines THP-1 and U937 were constructed by liposome transfection. The effect of SPOP on cell proliferation was detected by CCK-8, and the effect of SPOP on apoptosis and cell cycle was detected by flow cytometry. The expressions of anti-apoptotic protein Bcl-2 and apoptotic protein Bax, Caspase3 were detected by Western blot.
RESULTS:
The median expression level of SPOP mRNA in normal control group was 0.993 1(0.6303, 1.433), while that in AML group was 0.522 1(0.242 2, 0.723 7). The expression level of SPOP in AML group was significantly lower than that in normal control group ( P < 0.001). After the overexpression of SPOP, the proportion of apoptotic cells in the U937 overexpression group and THP-1 overexpression group was 10.9%±0.3% and 4.6%±015%, which were higher than 8.9%±0.3% and 3.0%±0.30% in the Empty Vector group, respectively (both P < 0.05). The expression of Caspase3 in U937 overexpression group and THP-1 overexpression group was 1.154±0.086 and 1.2±0.077, which were higher than 1 in Empty Vector group, respectively (both P < 0.05). The ratio of Bax/Bcl-2 in U937 overexpression group and THP-1 overexpression group was 1.328±0.057 and 1.669±0.15, which were higher than 1 in Empty Vector group, respectively (both P < 0.05). In the cell proliferation experiment, the number of cells in the U937 overexpression group and THP-1 overexpression group were both slightly lower than those in the Empty Vector group, but the differences were not statistically significant (P >0.05). In the cell cycle experiment, the proportion of G1 cells in the U937 overexpression group and THP-1 overexpression group were both slightly higher than those in the Empty Vector group, but the differences were not statistically significant (P >0.05).
CONCLUSION
SPOP can promote the apoptosis of leukemic cells, and its mechanism may be related to down-regulation of Bcl-2 expression and up-regulation of Bax and Caspase3 expression.
Humans
;
Leukemia, Myeloid, Acute/pathology*
;
Apoptosis
;
Repressor Proteins/genetics*
;
Cell Proliferation
;
Nuclear Proteins/genetics*
;
Cell Cycle
;
Proto-Oncogene Proteins c-bcl-2/metabolism*
;
Caspase 3/metabolism*
;
bcl-2-Associated X Protein/metabolism*
;
U937 Cells
;
Cell Line, Tumor
;
RNA, Messenger/genetics*
5.Effect of TBL1XR1 Mutation on Cell Biological Characteristics of Diffuse Large B-Cell Lymphoma.
Hong-Ming FAN ; Le-Min HONG ; Chun-Qun HUANG ; Jin-Feng LU ; Hong-Hui XU ; Jie CHEN ; Hong-Ming HUANG ; Xin-Feng WANG ; Dan GUO
Journal of Experimental Hematology 2025;33(2):423-430
OBJECTIVE:
To investigate the effect of TBL1XR1 mutation on cell biological characteristics of diffuse large B-cell lymphoma (DLBCL).
METHODS:
The TBL1XR1 overexpression vector was constructed and DNA sequencing was performed to determine the mutation status. The effect of TBL1XR1 mutation on apoptosis of DLBCL cell line was detected by flow cytometry and TUNEL fluorescence assay; CCK-8 assay was used to detect the effect of TBL1XR1 mutation on cell proliferation; Transwell assay was used to detect the effect of TBL1XR1 mutation on cell migration and invasion; Western blot was used to detect the effect of TBL1XR1 mutation on the expression level of epithelial-mesenchymal transition (EMT) related proteins.
RESULTS:
The TBL1XR1 overexpression plasmid was successfully constructed. The in vitro experimental results showed that TBL1XR1 mutation had no significant effect on apoptosis of DLBCL cells. Compared with the control group, TBL1XR1 mutation enhanced cell proliferation, migration and invasion of DLBCL cells. TBL1XR1 gene mutation significantly increased the expression of N-cadherin protein, while the expression of E-cadherin protein decreased.
CONCLUSION
TBL1XR1 mutation plays a role in promoting tumor cell proliferation, migration and invasion in DLBCL. TBL1XR1 could be considered as a potential target for DLBCL therapy in future research.
Humans
;
Lymphoma, Large B-Cell, Diffuse/pathology*
;
Cell Proliferation
;
Mutation
;
Receptors, Cytoplasmic and Nuclear/genetics*
;
Apoptosis
;
Cell Line, Tumor
;
Epithelial-Mesenchymal Transition
;
Cell Movement
;
Repressor Proteins/genetics*
;
Nuclear Proteins/genetics*
;
Cadherins/metabolism*
6.Clinical Analysis of Dyskeratosis Congenita in Children.
Wen-Qi LU ; Shao-Yan HU ; Jing GAO ; Wei GAO ; Jun-Jie FAN
Journal of Experimental Hematology 2025;33(3):906-912
OBJECTIVE:
To summarize the clinical characteristics, diagnosis, treatment and prognosis of dyskeratosis congenita (DC) in children, and to provide clinical experience for the diagnosis and treatment of DC.
METHODS:
The clinical data of children with dyskeratosis congenital admitted to Children's Hospital of Soochow University from May 2016 to May 2024 were retrospectively analyzed. Whole exome sequencing (WES) was performed, the patients were followed up and the related literature was reviewed.
RESULTS:
A total of 4 patients were enrolled. There were 1 male and 3 females. Two patients had spontaneous TINF2 mutation, one had TERT mutation, and one had DKC1 mutation. All of them had bone marrow hypoplasia. Two patients underwent allogeneic hematopoietic stem cell transplantation, and both had good engraftment. Anti-rejection drugs were stopped, and they survived more than 5 years of follow-up. One patient was followed up in outpatient department, and another patient was scheduled to undergo hematopoietic stem cell transplantation.
CONCLUSION
The onset of dyskeratosis congenita in children is insidious, so genetic diagnosis is particularly important. c.853_861delGTCATGCTG (p.285-287del) was a new mutation site of TINF2, which expanded the gene mutation spectrum of DC. Hematopoietic stem cell transplantation is an effective treatment for bone marrow failure, and the treatment of other organ complications depends on further genetic exploration.
Humans
;
Dyskeratosis Congenita/therapy*
;
Hematopoietic Stem Cell Transplantation
;
Male
;
Mutation
;
Female
;
Retrospective Studies
;
Telomerase/genetics*
;
Telomere-Binding Proteins/genetics*
;
Child
;
Cell Cycle Proteins/genetics*
;
Nuclear Proteins/genetics*
;
Child, Preschool
;
Prognosis
;
Exome Sequencing
7.Clinical Characteristics of Adult Acute Myeloid Leukemia Patients with NUP98::HOXA9 Fusion Gene.
Hai-Xia CAO ; Ya-Min WU ; Shu-Juan WANG ; Zhi-Dan CHEN ; Jing-Han HU ; Xiao-Qian GENG ; Fang WANG ; Ling SUN ; Zhong-Xing JIANG ; Zhi-Lei BIAN
Journal of Experimental Hematology 2025;33(5):1241-1247
OBJECTIVE:
To investigate the clinical characteristics, treatment and prognosis of adult AML patients with NUP98::HOXA9 fusion gene.
METHODS:
From May 2017 to October 2023, among 2 113 AML patients who visited the Hematology Department of our hospital, patients with NUP98 rearrangements were screened. The clinical characteristics, chromosome karyotypes, immunophenotypes, gene mutations, treatment efficacy and prognosis of the patients with NUP98::HOXA9 positive were analyzed.
RESULTS:
Among the 2 113 AML patients, there were 18 cases with NUP98 rearrangement, including 14 NUP98::HOXA9 positive cases, with a detection rate of 0.66% (14/2 113). The median age of the NUP98::HOXA9 positive patients was 42.5 (23-64) years old. The most common chromosome karyotype was t(7; 11)(p15; p15). The immunophenotypes of all patients expressed CD13, CD33, CD117 and CD38, and most patients expressed CD34 and cMPO, while only a few expressed HLA-DR. Second-generation sequencing (NGS) was performed to detect genetic mutations associated with leukemia in all 14 patients, and the genes exhibiting a high frequency of mutation were WT1 (10/14), TET2 (7/14), and FLT3-ITD (6/14). Additionally, mutations were also observed in KRAS/NRAS, IDH1, and KIT. Of the 13 patients who received treatment, 9 achieved complete remission (CR), and all 3 patients who received azacytidine(AZA)+ venetoclax (VEN) regimen achieved CR after the first course of treatment. Within this cohort, 6 patients were classified as relapsed/refractory (6/13). 4 patients underwent allogeneic hematopoietic stem cell transplantation (allo-HSCT), of which two achieved long-term survival. The median follow-up time was 12 (2.1-65.0) months, while the median overall survival (OS) and relapse-free survival (RFS) were recorded as 11.4 months and 9.6 months, respectively.
CONCLUSION
The most common type of NUP98 rearrangement in adults AML patients is NUP98::HOXA9 , which is often accompanied by somatic mutations in WT1, TET2, and FLT3-ITD. These patients are prone to relapse, have short survival time, and generally face poor prognoses. Hopefully, utilization of the AZA+VEN regimen is anticipated to enhance the rate of induced remission in the patients, and some patients may prolong their survival through allo-HSCT. However, more effective treatment methods are still needed to improve the overall prognosis of these patients.
Humans
;
Adult
;
Leukemia, Myeloid, Acute/genetics*
;
Middle Aged
;
Prognosis
;
Nuclear Pore Complex Proteins/genetics*
;
Oncogene Proteins, Fusion/genetics*
;
Mutation
;
Male
;
Female
;
Young Adult
;
Homeodomain Proteins/genetics*
8.Characterization of Acute Myeloid Leukemia Patients with DEK-NUP214 Fusion Gene Positive.
Ran HUANG ; Yuan-Bing WU ; Ya-Xue WU ; Xiao-Hui HU
Journal of Experimental Hematology 2025;33(5):1293-1298
OBJECTIVE:
To analyze the clinical features of acute myeloid leukemia patients with DEK-NUP214 fusion gene positive.
METHODS:
The DEK-NUP214 fusion gene was amplified by multi-nested PCR in 26 patients admitted to the First Affiliated Hospital of Soochow University from January 2018 to October 2023, and the disease course and post-transplant survival data were obtained by searching outpatient and inpatient medical records and telephone follow-up.
RESULTS:
The median follow-up time of pateints was 21.25(0.9-60.2) months. Among 26 patients with DEK-NUP214 fusion gene positive AML, 15 patients had FLT3-ITD gene mutation positive. One patient died after abandoning treatment due to non-remission of induction chemotherapy, one died due to infection, and 23 patients received allo-HSCT after achieving CR, of which one patient died within one month after transplantation due to multiple infections and one died due to severe pulmonary infection that did not respond to treatment. One patient received allo-HSCT in non-remission state and later died due to recurrence.
CONCLUSION
DEK-NUP214 fusion gene positive AML is a type of acute leukemia subtype with high risk and poor prognosis. Allo-HSCT treatment at the early stage of disease remission is the most effective way to improve the prognosis of patients.
Humans
;
Leukemia, Myeloid, Acute/genetics*
;
Poly-ADP-Ribose Binding Proteins
;
Oncogene Proteins, Fusion/genetics*
;
Nuclear Pore Complex Proteins/genetics*
;
Oncogene Proteins/genetics*
;
Chromosomal Proteins, Non-Histone/genetics*
;
Male
;
Female
;
Adult
;
Mutation
;
Hematopoietic Stem Cell Transplantation
;
Middle Aged
9.The splicing factor HNRNPH1 regulates Circ-MYOCD back-splicing to modulate the course of cardiac hypertrophy.
Rui CAI ; Zhuo HUANG ; Wenxia HE ; Tianhong AI ; Xiaowei SONG ; Shuting HU
Journal of Southern Medical University 2025;45(3):587-594
OBJECTIVES:
To explore the mechanism of Circ-MYOCD back-splicing and its regulatory role in myocardial hypertrophy.
METHODS:
Sanger sequencing and RNase R assays were performed to verify the circularity and stability of Circ-MYOCD, whose subcellular distribution was determined by nuclear-cytoplasmic fractionation. Bioinformatics analysis and mass spectrometry from pull-down assays were conducted to predict the RNA-binding proteins (RBPs) interacting with Circ-MYOCD. In rat cardiomyocytes H9C2 cells, the effects of HNRNPH1 and HNRNPL knockdown and overexpression on Circ-MYOCD back-splicing were evaluated. In a H9C2 cell model of angiotensin II (Ang II)-induced myocardial hypertrophy, the expression of HNRNPH1 was detected, the effects of HNRNPH1 knockdown and overexpression on progression of myocardial hypertrophy were assessed, and the regulatory effect of HNRNPH1 on Circ-MYOCD back-splicing was analyzed.
RESULTS:
Sanger sequencing confirmed that the junction primers could amplify the correct Circ-MYOCD sequence. RNase R and nuclear-cytoplasmic fractionation assays showed that Circ-MYOCD was stable and predominantly localized in the cytoplasm. Bioinformatics analysis and mass spectrometry from the Circ-MYOCD pull-down assay identified HNRNPH1 and HNRNPL as the RBPs interacting with Circ-MYOCD. In H9C2 cells, HNRNPH1 knockdown significantly enhanced while its overexpression inhibited Circ-MYOCD back-splicing; HNRNPH1 overexpression obviously increased the expressions of myocardial hypertrophy markers ANP and BNP, while its knockdown produced the opposite effect. In Ang II-induced H9C2 cells, which exhibited a significant increase of HNRNPH1 expression and increased expressions of ANP and BNP, HNRNPH1 knockdown obviously increased Circ-MYOCD expression, decreased MYOCD expression and lowered both ANP and BNP expressions.
CONCLUSIONS
HNRNPH1 regulates Circ-MYOCD back-splicing to influence the progression of myocardial hypertrophy.
Animals
;
Rats
;
RNA, Circular/genetics*
;
Cardiomegaly/metabolism*
;
Myocytes, Cardiac/metabolism*
;
Heterogeneous-Nuclear Ribonucleoprotein Group F-H/metabolism*
;
Cell Line
;
RNA Splicing
;
Angiotensin II
;
RNA-Binding Proteins
10.Inhibition of BRD4 promotes migration of esophageal squamous cell carcinoma cells with low ACC1 expression.
Wenxin JIA ; Shuhua HUO ; Jiaping TANG ; Yuzhen LIU ; Baosheng ZHAO
Journal of Southern Medical University 2025;45(10):2258-2269
OBJECTIVES:
To investigate the effect of BRD4 inhibition on migration of esophageal squamous cell carcinoma (ESCC) cells with low acetyl-CoA carboxylase 1 (ACC1) expression.
METHODS:
ESCC cell lines with lentivirus-mediated ACC1 knockdown or transfected with a negative control sequence (shNC) were treated with DMSO, JQ1 (a BRD4 inhibitor), co-transfection with shNC-siBRD4 or siNC with additional DMSO or C646 (an ahistone acetyltransferase inhibitor) treatment, or JQ1combined with 3-MA (an autophagy inhibitor). BRD4 mRNA expression in the cells was detected using RT-qPCR. The changes in cell proliferation, migration, autophagy, and epithelial-mesenchymal transition (EMT) were examined with CCK8 assay, Transwell migration assay, and Western blotting.
RESULTS:
ACC1 knockdown did not significantly affect BRD4 expression in the cells but obviously increased their sensitivity to JQ1. JQ1 treatment at 1 and 2 μmol/L significantly inhibited ESCC cell proliferation, while JQ1 at 0.2 and 2 μmol/L promoted cell migration. The cells with ACC1 knockdown and JQ1 treatment showed increased expresisons of vimentin and Slug and decreased expression of E-cadherin. BRD4 knockdown promoted migration of ESCC cells, and co-transfection with shACC1 and siBRD4 resulted in increased vimentin and Slug expressions and decreased E-cadherin expression in the cells. C646 treatment of the co-transfected cells reduced acetylation levels, decreased vimentin and Slug expressions, and increased E-cadherin expression. Treatment with JQ1 alone obviously increased LC3A/B-II levels in the cells either with or without ACC1 knockdown. In the cells with ACC1 knockdown and JQ1 treatment, additional 3-MA treatment significantly decreased the expressions of vimentin, Slug and LC3A/B-II and increased the expression of E-cadherin.
CONCLUSIONS
BRD4 inhibition promotes autophagy of ESCC cells via a histone acetylation-dependent mechanism, thereby enhancing EMT and ultimately increasing cell migration driven by ACC1 deficiency.
Humans
;
Cell Movement
;
Transcription Factors/metabolism*
;
Esophageal Neoplasms/metabolism*
;
Cell Line, Tumor
;
Cell Cycle Proteins
;
Azepines/pharmacology*
;
Epithelial-Mesenchymal Transition
;
Carcinoma, Squamous Cell/metabolism*
;
Esophageal Squamous Cell Carcinoma
;
Triazoles/pharmacology*
;
Nuclear Proteins/genetics*
;
Cell Proliferation
;
Acetyl-CoA Carboxylase/genetics*
;
Transfection
;
Autophagy
;
Bromodomain Containing Proteins

Result Analysis
Print
Save
E-mail