1.Application and mechanisms of targeting BRD4 in osteosarcoma.
Ding CHEN ; Jiaming TIAN ; Yihe DONG ; Zi LI ; Jun HUANG
Journal of Central South University(Medical Sciences) 2025;50(3):416-429
OBJECTIVES:
Metastasis is the primary cause of death in osteosarcoma, and current clinical treatments remain limited. BRD4, a key epigenetic regulator, has shown therapeutic promise in various cancers through its inhibition. However, the mechanistic role of BRD4 in osteosarcoma remains poorly understood. This study aims to elucidate the molecular mechanisms by which BRD4 regulate osteosarcoma progression and to explore novel therapeutic strategies.
METHODS:
Immunofluorescence was used to assess BRD4 expression levels in a tissue microarray containing 80 osteosarcoma samples from different patients. The Gene Expression Omnibus (GEO) dataset (GSE42352, containing survival data from 88 osteosarcoma patients) was downloaded to perform Kaplan-Meier survival analysis based on BRD4 gene expression levels. In vivo, an orthotopic intramedullary osteosarcoma model was established using HOS cells in C57 mice, followed by treatment with varying doses of the BRD4 inhibitor (+)-JQ1. Micro-CT, 3D reconstruction of bone tissue, and HE staining were employed to evaluate pathological changes in bone and intestinal lymph nodes. In vitro, cell viability was measured using the methyl thiazolyl tetrazolium (MTT) assay, while colony formation and Transwell assays assessed proliferative and invasive capacities. Chromatin-bound BRD4 was analyzed via co-immunoprecipitation combined with mass spectrometry (Co-IP/MS), and O-GlcNAc glycosylation sites and glycan chains of BRD4 were identified using Co-IP with Nano-LC MS/MS. Real-time PCR and Western blotting were used to analyze the relative mRNA and protein expression levels of target genes, respectively.
RESULTS:
BRD4 was positively expressed in 61.25% (49/80) of osteosarcoma tissues. Patients with high BRD4 expression exhibited significantly shorter survival times (P<0.05). In the orthotopic mouse model, intervention with (+)-JQ1, a potent and commonly used BETi, significantly inhibited tumor growth in vivo and reduced bone destruction (P<0.05). (+)-JQ1 treatment significantly suppressed the proliferation (P<0.001), invasion (P<0.001), and migration (P<0.05) of HOS cells. In osteosarcoma cells, BRD4 exhibited O-GlcNAc modifications at both N- and C- C-termini, particularly at Thr73, which is essential for protein stability. This modification also contributed to the activation of the EGFR tyrosine kinase inhibitor resistance pathway (KEGG Pathway: hsa01521). (+)-JQ1 treatment displaced BRD4 from enhancers and downregulated the transcription of pathway-related genes, such as EGFR and PDGFC, thereby suppressing the malignant behavior of osteosarcoma cells.
CONCLUSIONS
BRD4 promotes osteosarcoma progression via O-GlcNAc modification at Thr73 and plays a crucial role in tumor growth and metastasis.
Osteosarcoma/drug therapy*
;
Humans
;
Transcription Factors/metabolism*
;
Animals
;
Cell Cycle Proteins
;
Mice
;
Bone Neoplasms/drug therapy*
;
Azepines/pharmacology*
;
Cell Line, Tumor
;
Cell Proliferation/drug effects*
;
Triazoles/pharmacology*
;
Mice, Inbred C57BL
;
Nuclear Proteins/metabolism*
;
Gene Expression Regulation, Neoplastic
;
Male
;
Bromodomain Containing Proteins
2.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
3.Analysis of the genomic landscape of primary central nervous system lymphoma using whole-genome sequencing in Chinese patients.
Xianggui YUAN ; Teng YU ; Jianzhi ZHAO ; Huawei JIANG ; Yuanyuan HAO ; Wen LEI ; Yun LIANG ; Baizhou LI ; Wenbin QIAN
Frontiers of Medicine 2023;17(5):889-906
Primary central nervous system lymphoma (PCNSL) is an uncommon non-Hodgkin's lymphoma with poor prognosis. This study aimed to depict the genetic landscape of Chinese PCNSLs. Whole-genome sequencing was performed on 68 newly diagnosed Chinese PCNSL samples, whose genomic characteristics and clinicopathologic features were also analyzed. Structural variations were identified in all patients with a mean of 349, which did not significantly influence prognosis. Copy loss occurred in all samples, while gains were detected in 77.9% of the samples. The high level of copy number variations was significantly associated with poor progression-free survival (PFS) and overall survival (OS). A total of 263 genes mutated in coding regions were identified, including 6 newly discovered genes (ROBO2, KMT2C, CXCR4, MYOM2, BCLAF1, and NRXN3) detected in ⩾ 10% of the cases. CD79B mutation was significantly associated with lower PFS, TMSB4X mutation and high expression of TMSB4X protein was associated with lower OS. A prognostic risk scoring system was also established for PCNSL, which included Karnofsky performance status and six mutated genes (BRD4, EBF1, BTG1, CCND3, STAG2, and TMSB4X). Collectively, this study comprehensively reveals the genomic landscape of newly diagnosed Chinese PCNSLs, thereby enriching the present understanding of the genetic mechanisms of PCNSL.
Humans
;
DNA Copy Number Variations
;
Nuclear Proteins/genetics*
;
Central Nervous System Neoplasms/pathology*
;
Transcription Factors/genetics*
;
Prognosis
;
Lymphoma/genetics*
;
Genomics
;
China
;
Central Nervous System/pathology*
;
Bromodomain Containing Proteins
;
Cell Cycle Proteins/genetics*

Result Analysis
Print
Save
E-mail