1.Combination of Components from Tripterygii Radix et Rhizoma-Chuanxiong Rhizoma Affects RA-FLSs by Regulating NF-κB, Nrf2/HO-1 Signaling Pathways and Bcl-2/Caspase-3 Expression
Yongmei GUAN ; Zhiyan WAN ; Shuhui WANG ; Weifeng ZHU ; Zhiyong LIU ; Cheng JIANG ; Zhenzhong ZANG
Chinese Journal of Experimental Traditional Medical Formulae 2025;31(2):17-26
ObjectiveTo investigate the effects of the combination of components from Tripterygii Radix et Rhizoma and Chuanxiong Rhizoma on rheumatoid arthritis fibroblast-like synoviocytes (RA-FLSs) and the underlying mechanism. MethodsRA-FLSs were grouped as follows: blank control, positive control (methotrexate), Tripterygii Radix et Rhizoma components, Chuanxiong Rhizoma components, and components from Tripterygii Radix et Rhizoma+Chuanxiong Rhizoma. The cell-counting kit-8 (CCK-8) assay was employed to the cell proliferation, invasion, and apoptosis. The levels of tumor necrosis factor (TNF)-α, interleukin (IL)-6, reactive oxygen species (ROS), and malondiadehyde (MDA) in cells were measured. Western blot was employed to determine the protein levels of nuclear factor erythroid 2-related factor 2 (Nrf2), heme oxygenase-1 (HO-1), nuclear factor-kappa B (NF-κB) p65, phosphorylated inhibitory subunit of NF-κBα (p-IκBα), cysteinyl aspartate-specific protease-3 (Caspase-3), and B-cell lymphoma 2 (Bcl-2). Real-time PCR was employed to determine the mRNA levels of Nrf2, HO-1, and NF-κB p65. ResultsThe cells in the groups of positive control, Tripterygii Radix et Rhizoma components, Chuanxiong Rhizoma components, and components from Tripterygii Radix et Rhizoma+Chuanxiong Rhizoma were treated with 2.50 mg·L-1 methotrexate, 0.20 mg·L-1 triptolide + 0.20 mg·L-1 celastrol, 5.00 mg·L-1 ferulic acid + 20.00 mg·L-1 ligustrazine, 0.20 mg·L-1 triptolide + 0.20 mg·L-1 celastrol + 5.00 mg·L-1 ferulic acid + 20.00 mg·L-1 ligustrazine, respectively. Compared with the blank control group, drug administration reduced the proliferation and invasion and increased the apoptosis of cells (P<0.01), lowered the levels of TNF-α, IL-6, ROS, and MDA (P<0.01), up-regulated the mRNA and protein levels of Caspase-3, Nrf2, and HO-1 (P<0.01), and down-regulated the mRNA and protein levels of Bcl-2, NF-κB p65, and p-IκBα (P<0.01). Compared with the Tripterygii Radix et Rhizoma components group, the combination of components from Tripterygii Radix et Rhizoma+Chuanxiong Rhizoma inhibited the proliferation and invasion (P<0.05) and promoted the apoptosis of RA-FLSs, up-regulated the mRNA levels of Nrf2 and HO-1 and protein levels of Nrf2 and Caspase-3 (P<0.05), and down-regulated the protein levels of NF-κB p65 and p-IκBα (P<0.05). ConclusionThe combination of components from Chuanxiong Rhizoma and Tripterygii Radix et Rhizoma can inhibit the proliferation and invasion and promote the apoptosis of RA-FLSs and alleviate oxidative stress and inflammation by inhibiting the NF-κB signaling pathway, activating the Nrf2/HO-1 pathway, and regulating the expression of Bcl-2/Caspase-3.
2.Progress in the treatment of giant cell tumors of extremities with pathological fracture
Wenhao YAO ; Daoyang FAN ; Xieyuan JIANG ; Weifeng LIU
Chinese Journal of Surgery 2025;63(1):81-85
Giant cell tumor of bone (GCTB) is a common locally aggressive junctional primary bone tumor, whose clinical treatment becomes more difficult once combined with pathological fracture. Extended curettage and en-bloc resection are common surgical procedures for treating GCTB, and drugs such as receptor activator of nuclear factor-κB ligand(RANKL) inhibitors and bisphosphonates have been successfully used. Curettage is recommended for patients with Campanaccigrade Ⅱor Campanaccigrade Ⅲ with localized soft tissue invasion only and simple fractures with intact bone structure. Resection may be considered for Campanaccigrade Ⅲ with extensive soft tissue invasion or complex fractures with incomplete bone structure. RANKL inhibitors such as denosumab may be recommended if surgery is not possible or before performing resection. This article summarizes the common treatment modalities of pathological fractures combined with giant cell tumors of extremities, including the current status of surgical and pharmacological treatments, analyzing the choice of surgical modalities in different clinical situations, in order to provide clinical inspirations for diagnosis and treatment.
3.Progress in the treatment of giant cell tumors of extremities with pathological fracture
Wenhao YAO ; Daoyang FAN ; Xieyuan JIANG ; Weifeng LIU
Chinese Journal of Surgery 2025;63(1):81-85
Giant cell tumor of bone (GCTB) is a common locally aggressive junctional primary bone tumor, whose clinical treatment becomes more difficult once combined with pathological fracture. Extended curettage and en-bloc resection are common surgical procedures for treating GCTB, and drugs such as receptor activator of nuclear factor-κB ligand(RANKL) inhibitors and bisphosphonates have been successfully used. Curettage is recommended for patients with Campanaccigrade Ⅱor Campanaccigrade Ⅲ with localized soft tissue invasion only and simple fractures with intact bone structure. Resection may be considered for Campanaccigrade Ⅲ with extensive soft tissue invasion or complex fractures with incomplete bone structure. RANKL inhibitors such as denosumab may be recommended if surgery is not possible or before performing resection. This article summarizes the common treatment modalities of pathological fractures combined with giant cell tumors of extremities, including the current status of surgical and pharmacological treatments, analyzing the choice of surgical modalities in different clinical situations, in order to provide clinical inspirations for diagnosis and treatment.
4.Research Progress on the Correlation Between Mitophagy and Vascular Cognitive Impairment
Yan LIU ; Xingang DONG ; Xiaoyuan WANG ; Gege QI ; Yiqin REN ; Lianpeng ZHOU ; Hui LI ; Suqing ZHANG ; Weifeng LI
Medical Journal of Peking Union Medical College Hospital 2025;16(2):338-349
Vascular cognitive impairment (VCI), caused by cerebrovascular dysfunction, severely impacts the quality of life in the elderly population, yet effective therapeutic approaches remain limited. Mitophagy, a selective mitochondrial quality-control mechanism, has emerged as a critical focus in neurological disease research. Accumulating evidence indicates that mitophagy modulates oxidative stress, neuroinflammation, and neuronal apoptosis. Key signaling pathways associated with mitophagy—including PINK1/Parkin, BNIP3/Nix, FUNDC1, PI3K/Akt/mTOR, and AMPK—have been identified as potential therapeutic targets for VCI. This review summarizes the mechanistic roles of mitophagy in VCI pathogenesis and explores emerging therapeutic strategies targeting these pathways, aiming to provide novel insights for clinical intervention and advance the development of effective treatments for VCI.
5.Targeting TM4SF1 promotes tumor senescence enhancing CD8+ T cell cytotoxic function in hepatocellular carcinoma
Weifeng ZENG ; Furong LIU ; Yachong LIU ; Ze ZHANG ; Haofan HU ; Shangwu NING ; Hongwei ZHANG ; Xiaoping CHEN ; Zhibin LIAO ; Zhanguo ZHANG
Clinical and Molecular Hepatology 2025;31(2):489-508
Background/Aims:
Transmembrane 4 L six family member 1 (TM4SF1) is highly expressed and contributes to the progression of various malignancies. However, how it modulates hepatocellular carcinoma (HCC) progression and senescence remains to be elucidated.
Methods:
TM4SF1 expression in HCC samples was evaluated using immunohistochemistry and flow cytometry. Cellular senescence was assessed through SA-β-gal activity assays and Western blot analysis. TM4SF1-related protein interactions were investigated using immunoprecipitation-mass spectrometry, co-immunoprecipitation, bimolecular fluorescence complementation, and immunofluorescence. Tumor-infiltrating immune cells were analyzed by flow cytometry. The HCC mouse model was established via hydrodynamic tail vein injection.
Results:
TM4SF1 was highly expressed in human HCC samples and murine models. Knockdown of TM4SF1 suppressed HCC proliferation both in vitro and in vivo, inducing non-secretory senescence through upregulation of p16 and p21. TM4SF1 enhanced the interaction between AKT1 and PDPK1, thereby promoting AKT phosphorylation, which subsequently downregulated p16 and p21. Meanwhile, TM4SF1-mediated AKT phosphorylation enhanced PD-L1 expression while reducing major histocompatibility complex class I level on tumor cells, leading to impaired cytotoxic function of CD8+ T cells and an increased proportion of exhausted CD8+ T cells. In clinical HCC samples, elevated TM4SF1 expression was associated with resistance to anti-PD-1 immunotherapy. Targeting TM4SF1 via adeno-associated virus induced tumor senescence, reduced tumor burden and synergistically enhanced the efficacy of anti-PD-1 therapy.
Conclusions
Our results revealed that TM4SF1 regulated tumor cell senescence and immune evasion through the AKT pathway, highlighting its potential as a therapeutic target in HCC, particularly in combination with first-line immunotherapy.
7.Targeting TM4SF1 promotes tumor senescence enhancing CD8+ T cell cytotoxic function in hepatocellular carcinoma
Weifeng ZENG ; Furong LIU ; Yachong LIU ; Ze ZHANG ; Haofan HU ; Shangwu NING ; Hongwei ZHANG ; Xiaoping CHEN ; Zhibin LIAO ; Zhanguo ZHANG
Clinical and Molecular Hepatology 2025;31(2):489-508
Background/Aims:
Transmembrane 4 L six family member 1 (TM4SF1) is highly expressed and contributes to the progression of various malignancies. However, how it modulates hepatocellular carcinoma (HCC) progression and senescence remains to be elucidated.
Methods:
TM4SF1 expression in HCC samples was evaluated using immunohistochemistry and flow cytometry. Cellular senescence was assessed through SA-β-gal activity assays and Western blot analysis. TM4SF1-related protein interactions were investigated using immunoprecipitation-mass spectrometry, co-immunoprecipitation, bimolecular fluorescence complementation, and immunofluorescence. Tumor-infiltrating immune cells were analyzed by flow cytometry. The HCC mouse model was established via hydrodynamic tail vein injection.
Results:
TM4SF1 was highly expressed in human HCC samples and murine models. Knockdown of TM4SF1 suppressed HCC proliferation both in vitro and in vivo, inducing non-secretory senescence through upregulation of p16 and p21. TM4SF1 enhanced the interaction between AKT1 and PDPK1, thereby promoting AKT phosphorylation, which subsequently downregulated p16 and p21. Meanwhile, TM4SF1-mediated AKT phosphorylation enhanced PD-L1 expression while reducing major histocompatibility complex class I level on tumor cells, leading to impaired cytotoxic function of CD8+ T cells and an increased proportion of exhausted CD8+ T cells. In clinical HCC samples, elevated TM4SF1 expression was associated with resistance to anti-PD-1 immunotherapy. Targeting TM4SF1 via adeno-associated virus induced tumor senescence, reduced tumor burden and synergistically enhanced the efficacy of anti-PD-1 therapy.
Conclusions
Our results revealed that TM4SF1 regulated tumor cell senescence and immune evasion through the AKT pathway, highlighting its potential as a therapeutic target in HCC, particularly in combination with first-line immunotherapy.
9.Targeting TM4SF1 promotes tumor senescence enhancing CD8+ T cell cytotoxic function in hepatocellular carcinoma
Weifeng ZENG ; Furong LIU ; Yachong LIU ; Ze ZHANG ; Haofan HU ; Shangwu NING ; Hongwei ZHANG ; Xiaoping CHEN ; Zhibin LIAO ; Zhanguo ZHANG
Clinical and Molecular Hepatology 2025;31(2):489-508
Background/Aims:
Transmembrane 4 L six family member 1 (TM4SF1) is highly expressed and contributes to the progression of various malignancies. However, how it modulates hepatocellular carcinoma (HCC) progression and senescence remains to be elucidated.
Methods:
TM4SF1 expression in HCC samples was evaluated using immunohistochemistry and flow cytometry. Cellular senescence was assessed through SA-β-gal activity assays and Western blot analysis. TM4SF1-related protein interactions were investigated using immunoprecipitation-mass spectrometry, co-immunoprecipitation, bimolecular fluorescence complementation, and immunofluorescence. Tumor-infiltrating immune cells were analyzed by flow cytometry. The HCC mouse model was established via hydrodynamic tail vein injection.
Results:
TM4SF1 was highly expressed in human HCC samples and murine models. Knockdown of TM4SF1 suppressed HCC proliferation both in vitro and in vivo, inducing non-secretory senescence through upregulation of p16 and p21. TM4SF1 enhanced the interaction between AKT1 and PDPK1, thereby promoting AKT phosphorylation, which subsequently downregulated p16 and p21. Meanwhile, TM4SF1-mediated AKT phosphorylation enhanced PD-L1 expression while reducing major histocompatibility complex class I level on tumor cells, leading to impaired cytotoxic function of CD8+ T cells and an increased proportion of exhausted CD8+ T cells. In clinical HCC samples, elevated TM4SF1 expression was associated with resistance to anti-PD-1 immunotherapy. Targeting TM4SF1 via adeno-associated virus induced tumor senescence, reduced tumor burden and synergistically enhanced the efficacy of anti-PD-1 therapy.
Conclusions
Our results revealed that TM4SF1 regulated tumor cell senescence and immune evasion through the AKT pathway, highlighting its potential as a therapeutic target in HCC, particularly in combination with first-line immunotherapy.

Result Analysis
Print
Save
E-mail