1.A novel feedback loop: CELF1/circ-CELF1/BRPF3/KAT7 in cardiac fibrosis.
Yuan JIANG ; Bowen ZHANG ; Bo ZHANG ; Xinhua SONG ; Xiangyu WANG ; Wei ZENG ; Liyang ZUO ; Xinqi LIU ; Zheng DONG ; Wenzheng CHENG ; Yang QIAO ; Saidi JIN ; Dongni JI ; Xiaofei GUO ; Rong ZHANG ; Xieyang GONG ; Lihua SUN ; Lina XUAN ; Berezhnova Tatjana ALEXANDROVNA ; Xiaoxiang GUAN ; Mingyu ZHANG ; Baofeng YANG ; Chaoqian XU
Acta Pharmaceutica Sinica B 2025;15(10):5192-5211
Cardiac fibrosis is characterized by an elevated amount of extracellular matrix (ECM) within the heart. However, the persistence of cardiac fibrosis ultimately diminishes contractility and precipitates cardiac dysfunction. Circular RNAs (circRNAs) are emerging as important regulators of cardiac fibrosis. Here, we elucidate the functional role of a specific circular RNA CELF1 in cardiac fibrosis and delineate a novel feedback loop mechanism. Functionally, circ-CELF1 was involved in enhancing fibrosis-related markers' expression and promoting the proliferation of cardiac fibroblasts (CFs), thereby exacerbating cardiac fibrosis. Mechanistically, circ-CELF1 reduced the ubiquitination-degradation rate of BRPF3, leading to an elevation of BRPF3 protein levels. Additionally, BRPF3 acted as a modular scaffold for the recruitment of histone acetyltransferase KAT7 to facilitate the induction of H3K14 acetylation within the promoters of the Celf1 gene. Thus, the transcription of Celf1 was dramatically activated, thereby inhibiting the subsequent response of their downstream target gene Smad7 expression to promote cardiac fibrosis. Moreover, Celf1 further promoted Celf1 pre-mRNA transcription and back-splicing, thereby establishing a feedback loop for circ-CELF1 production. Consequently, a novel feedback loop involving CELF1/circ-CELF1/BRPF3/KAT7 was established, suggesting that circ-CELF1 may serve as a potential novel therapeutic target for cardiac fibrosis.
2.Moslosooflavone ameliorates dextran sulfate sodium-induced colitis in mice by suppressing intestinal epithelium apoptosis via inhibiting the PI3K/AKT signaling pathway.
Fei CHU ; Xiaohua CHEN ; Bowen SONG ; Jingjing YANG ; Lugen ZUO
Journal of Southern Medical University 2025;45(4):819-828
OBJECTIVES:
To investigate the effect of moslosooflavone (MOS) for ameliorating dextran sulfate sodium (DSS)-induced colitis in mice and the underlying molecular mechanism.
METHODS:
C57BL/6J mice with or without DSS exposure in the drinking water were both randomized into two groups for treatment with intraperitoneal injections with MOS (200 mg/kg) or normal saline for 7 days (n=6). Disease severity of the mice was assessed by observing changes in body weight, colon length, histopathology (HE staining), intestinal barrier function, and TUNEL staining. In the in vitro studies, lipopolysaccharide (LPS)-stimulated mouse colon organoids were treated with MOS (120 μmol/L) for 24 h, and the changes in barrier dysfunction and inflammation were analyzed. Network pharmacology and Western blotting were employed to identify functional pathways and apoptotic protein regulation associated with the therapeutic effect of MOS on colitis.
RESULTS:
In the mouse models of DSS-indcued colitis, MOS treatment significantly reduced body weight loss, disease activity index (DAI) scores and colon shortening, ameliorated colonic histopathological changes and inflammation, and lowered pro-inflammatory cytokine levels (TNF-α, IL-1β, IL-6, and IFN-γ). MOS effectively restored intestinal barrier integrity in the mice by reducing serum FITC-dextran and I-FABP concentrations while enhancing the tight junction proteins (ZO-1 and claudin-1). In the colon organoids, MOS significantly suppressed LPS-induced inflammatory responses and epithelial barrier disruption. Western blotting revealed that MOS downregulated C-caspase-3 and BAX and upregulated Bcl-2 expressions in both models. Mechanistically, MOS suppressed PI3K and AKT phosphorylation in both DSS-treated mouse colonic tissues and LPS-stimulated organoids.
CONCLUSIONS
MOS alleviates experimental colitis in mice by inhibiting intestinal epithelial apoptosis via inhibiting the PI3K/AKT pathway, thereby restoring intestinal barrier integrity and reducing inflammation.
Animals
;
Dextran Sulfate
;
Mice, Inbred C57BL
;
Colitis/metabolism*
;
Mice
;
Signal Transduction/drug effects*
;
Intestinal Mucosa/metabolism*
;
Apoptosis/drug effects*
;
Proto-Oncogene Proteins c-akt/metabolism*
;
Phosphatidylinositol 3-Kinases/metabolism*
;
Flavones/pharmacology*
;
Male
3.SF3B3 overexpression promotes proliferation of gastric cancer cells and correlates with poor patient prognosis.
Hui LU ; Bowen SONG ; Jinran SHI ; Shunyin WANG ; Xiaohua CHEN ; Jingjing YANG ; Sitang GE ; Lugen ZUO
Journal of Southern Medical University 2025;45(10):2240-2249
OBJECTIVES:
To investigate the role of SF3B3 in gastric cancer (GC) progression and prognosis and its possible mechanisms.
METHODS:
SF3B3 expression levels in pan-cancer and GC were analyzed using TIMER2.0, GEPIA, and UALCAN databases and validated using immunohistochemistry in GC tissues. Survival curves of GC patients were established using Kaplan-Meier Plotter and the data of a patient cohort our hospital. The independent risk factors for 5-year postoperative survival were identified using Cox regression, and their predictive values were evaluated using ROC analysis. SF3B3-associated biological processes were predicted by bioinformatics enrichment analyses. In GC HGC-27 cells, the effects of lentivirus-mediated SF3B3 knockdown and overexpression on cell proliferation and migration were investigated, and the changes in the key glycolytic proteins and extracellular acidification rate (ECAR) were detected. The influence of SF3B3 expression level on tumorigenesis and glycolytic protein expression in vivo were evaluated in a nude mouse xenograft model.
RESULTS:
High expression of SF3B3 in GC was associated with poor patient prognosis (P<0.05). The factors affecting 5-year survival outcomes following gastric oncological resection included high SF3B3 expression, a CEA level ≥5μg/L, a CA19-9 level ≥37 kU/L, tumor stage T3-4, and lymph node metastasis stage N2-3 (P<0.05). Bioinformatics analysis showed significant enrichment of SF3B3 in glycolysis. In HGC-27 cells, SF3B3 knockdown significantly inhibited while SF3B3 overexpression enhanced cell proliferation, migration, and invasion. SF3B3 knockdown obviously decreased the expressions of HK2, PKM2 and LDHA proteins and ECAR in HGC-27 cells, whereas SF3B3 overexpression produced the opposite effect. In nude mouse xenograft models, SF3B3 knockdown significantly reduced tumor mass and downregulated expression of HK2, PKM2 and LDHA proteins, and SF3B3 overexpression induced the opposite changes.
CONCLUSIONS
SF3B3 overexpression is associated with poor prognosis of GC patients and promotes GC cell proliferation, migration and invasion possibly by enhancing glycolysis.
Stomach Neoplasms/diagnosis*
;
Humans
;
Cell Proliferation
;
Prognosis
;
Animals
;
Mice, Nude
;
Cell Line, Tumor
;
Mice
;
Cell Movement
;
Male
;
Female
4.Elevated TMCO1 expression in gastric cancer is associated poor prognosis and promotes malignant phenotypes of tumor cells by inhibiting apoptosis.
Bowen SONG ; Renjie ZHOU ; Ying XU ; Jinran SHI ; Zhizhi ZHANG ; Jing LI ; Zhijun GENG ; Xue SONG ; Lian WANG ; Yueyue WANG ; Lugen ZUO
Journal of Southern Medical University 2025;45(11):2385-2393
OBJECTIVES:
To investigate the impact of high expression of transmembrane and coiled helix structural domain 1 (TMCO1) on prognosis of gastric cancer and the possible mechanisms.
METHODS:
TMCO1 expression in gastric cancer and its effect on gastric cancer progression and prognosis were analyzed using publicly available databases and clinical data of patients undergoing radical surgery in our hospital, and its possible biological functions were explored using KEGG and GO analyses. In gastric cancer HGC-27 cells, the effects of lentivirus-mediated TMCO1 overexpression and TMCO1 silencing on cell apoptosis, proliferation, invasion and migration were examined.
RESULTS:
TMCO1 expression was significantly elevated in gastric cancer tissues (P<0.05), and its high expression was positively correlated with cancer progression (P<0.001) and a lowered postoperative 5-year survival rate of the patients (P<0.05). Bioinformatic analyses suggested that TMCO1 may affect gastric cancer cell apoptosis via Wnt signaling. In HGC-27 cells, TMCO1 overexpression significantly promoted tumor cell proliferation, inhibited cell apoptosis, and enhanced cell migration and invasion, whereas TMCO1 silencing produced the opposite effects. Western blotting showed that β-catenin levels were significantly upregulated in TMCO1-overexpressing cells and downregulated in cells with TMCO1 silencing.
CONCLUSIONS
TMCO1 is overexpressed in gastric cancer tissues, and its high expression promotes gastric cancer progression and affects long-term prognosis of the patients possibly by activating the Wnt/ β-catenin signaling pathway to inhibit apoptosis of gastric cancer cells.
Humans
;
Stomach Neoplasms/metabolism*
;
Apoptosis
;
Prognosis
;
Cell Line, Tumor
;
Cell Proliferation
;
Cell Movement
;
Wnt Signaling Pathway
;
beta Catenin/metabolism*
;
Gene Expression Regulation, Neoplastic
6. Atractyloside targets the area of action of oncoprotein BORIS to inhibit cancer cell proliferation
Chen LIU ; Mengdie FANG ; Hao XU ; Chao LI ; Juan REN ; Bowen ZUO ; Yanmei ZHANG
Chinese Journal of Clinical Pharmacology and Therapeutics 2021;26(9):1005-1013
AIM: To analyze the binding area of atractyloside targeting oncoprotein BORIS to inhibit cancer cell proliferation. METHODS: DNAMAN comparison sequences were used to find the conserved regions of BORIS. Conservative regions were elected and the structure were predicted using SWISS-MODEL. ChemBio3D Ultra was used for minimum structure quantification, and Autodocking for molecular docking. The BORIS of the corresponding segment were overexpressed for verification. RESULTS: BORIS-N end had relatively conserved regions and high-level structures in the biological evolution process. The N-terminal of human-derived BORIS was the main action area we speculated, especially the 70th to 97th amino acids, and the site that binded preferentially to atractyloside after molecular docking was the 96th position (Glutamine), and this area would inhibit cell proliferation. CONCLUSION: BORIS-N terminal and atractyloside have an action area, and this segment has an important effect on cell proliferation, which is of great significance for the future screening of targeted drugs.

Result Analysis
Print
Save
E-mail