1.Effect of Go-Ichi-Ni-San complex subunit 1 on disease progression and chemotherapy resistance in hepatocellular carcinoma
Yishan HUO ; Dawei LI ; Xiangbing DUAN ; Yuyu MA ; Guojun ZHANG ; Kainan ZHANG ; Xiumin MA
Journal of Clinical Hepatology 2025;41(3):485-492
ObjectiveTo investigate the role and mechanism of Go-Ichi-Ni-San complex subunit 1 (GINS1) in the progression of hepatocellular carcinoma (HCC) and the development of chemotherapy resistance. MethodsThe tumor database GEPIA2 was used to analyze the differential expression of GINS1 between HCC patients and healthy individuals, and pathological tissue samples were collected from 40 HCC patients who were admitted to The Affiliated Tumor Hospital of Xinjiang Medical University and the First Affiliated Hospital of Xinjiang Medical University from May 2017 to January 2021. Immunohistochemical staining was used to measure the difference in the expression of GINS1 between HCC tissue and corresponding adjacent tissue, and the correlation between the expression level of GINS1 and the clinical TNM stage of HCC was analyzed. Western blot was also used to measure the difference in the expression of GINS1 between HCC Huh7/Hep3B/Li-7/MHCC97H cell lines and normal human QSG7701 hepatocytes. The method of lentivirus transfection was used to establish the MHCC97H cell line with stable GINS1 knockdown and its negative control cell line. CCK-8 assay and colony formation assay were used to measure cell proliferative capacity; scratch assay was used to measure cell migration ability; Transwell assay was used to measure cell invasion ability; cells were treated with oxaliplatin to measure their sensitivity to chemotherapy drugs. Nude mice were used to establish a tumor-bearing model and observe the effect of GINS1 knockdown on the growth of HCC in vivo. Western Blot was used to measure the expression levels of the proteins associated with the Notch pathway and the JAK/STAT pathway. The cells were treated with the Notch receptor agonist Jagged-1 to analyze the association between GINS1 and the Notch/JAK/STAT pathway. The independent-samples t test was used for comparison of continuous data between two groups; a one-way analysis of variance was used for comparison between multiple groups, and the least significant difference t-test was used for further comparison between two groups. ResultsThe expression of GINS1 was upregulated in HCC patients, HCC tissue, and HCC cell lines (all P<0.05), and the expression level of GINS1 was positively correlated with the clinical TNM stage of HCC (r=0.822, P=0.011). Compared with the negative control cells, the GINS1-knockdown MHCC97H cells showed significant reductions in proliferation, migration, and invasion activities (all P<0.01) and a significantly enhanced sensitivity to oxaliplatin (P<0.01). Compared with the nude mice in the control group, GINS1 knockdown caused significant inhibition of tumor weight and volume in vivo in nude mice (all P<0.001). Compared with the negative control cells, the GINS1-knockdown MHCC97H cells showed significant reductions in the expression levels of Notch1, Notch3, p-JAK2, and p-STAT3 (all P<0.05), while there were no significant differences in the overall expression levels of JAK2 and STAT3 (P>0.05). After Jagged-1 treatment, the GINS1-knockdown MHCC97H cells showed significant increases in proliferation, migration, and invasion activities and a significant reduction in sensitivity to oxaliplatin, as well as significant increases in the levels of p-JAK2 and p-STAT3 (all P<0.05). ConclusionGINS1 is upregulated in HCC and can promote HCC progression and chemotherapy resistance through the Notch/JAK2/STAT3 pathway.
2.Inhibitory effect of hydroxy safflower yellow A on neuronal pyroptosis after glucose-oxygen deprivation/reglucose-reoxygenation treatment
Zeqian WANG ; Yanzhe DUAN ; Yige WU ; Dong MA ; Jianjun HUANG ; Yuqing YAN ; Lijuan SONG
Chinese Journal of Tissue Engineering Research 2025;29(19):4044-4051
BACKGROUND:Hydroxy safflower yellow A has anti-ischemia,anti-oxidation,anti-thrombotic and anti-inflammatory effects.Whether it affects neuronal pyroptosis after glucose-oxygen deprivation/reglucose-reoxygenation is still unclear. OBJECTIVE:To investigate the protective effect of hydroxy safflower yellow A on neuronal pyroptosis and its mechanism. METHODS:HT22 cells in logarithmic growth phase were randomly divided into five groups:normal group,model group,hydroxy safflower yellow A group,colivelin group,and colivelin+hydroxy safflower yellow A group.HT22 cells were treated with glucose-oxygen deprivation/reglucose-reoxygenation to establish neuronal pyroptosis model,and then treated with STAT3 agonist Colivelin and hydroxy safflower yellow A.JC-1 probe was employed to assess changes in mitochondrial membrane potential.Reactive oxygen species kit was used to determine the content of reactive oxygen species in cells.GSDMD/TUNEL staining was conducted to observe cell pyroptosis.Immunofluorescence analysis was performed to detect STAT3 and GSDMD protein expression.RT-PCR was utilized for assessing mRNA expression levels of STAT3,NLRP3,and Caspase-1.Western blot assay was utilized to measure the protein expression levels of p-STAT3,NLRP3,GSDMD,Cleaved-caspase-1,and interleukin-1β. RESULTS AND CONCLUSION:(1)Compared with the normal group,the number of pyroptotic cells increased in HT22 cells in the model group along with a significant increase in protein expression levels of p-STAT3,NLRP3,Cleaved-caspase-1,GSDMD,and interleukin-1β.Compared with the model group,the number of pyroptotic cells reduced,and the expression of pyroptosis-related proteins significantly decreased in the hydroxy safflower yellow A group.(2)In comparison with the model group,pyroptosis worsened in the colivelin group where mitochondrial membrane potential decreased along with elevated reactive oxygen species content and increased mRNA expression levels of STAT3,NLRP3,and Caspase-1,as well as increased protein expression levels of p-STAT3,NLRP3,GSDMD,Cleaved-caspase-1,and interleukin-1β.Compared with the Colivelin group,above indexes were improved in the colivelin+hydroxy safflower yellow A group.These results suggest that hydroxy safflower yellow A plays a neuroprotective role through STAT3 signaling pathway to inhibit HT22 pyroptosis after glucose-oxygen deprivation/reglucose-reoxygenation treatment.
3.ST6GAL1 promotes glycolysis, migration and invasion of colorectal cancer HCT116 cells by activating the Notch1/PI3K/AKT/mTORC1 pathway
HUO Yishan1 ; WU Huili1 ; DUAN Xiangbing1 ; MA Xiumin1 ; LI Tao2
Chinese Journal of Cancer Biotherapy 2025;32(5):469-475
[摘 要] 目的:探究β-半乳糖苷α-2-6唾液酸转移酶1(ST6GAL1)对结直肠癌(CRC)HCT116细胞糖酵解和迁移、侵袭的作用及可能的分子机制。方法:通过检索GEPIA2数据库,分析ST6GAL1在CRC患者和健康人群中的表达差异;WB法检测ST6GAL1在CRC细胞HCT116、SW480、Caco-2、HT29、LoVo和人正常结肠上皮细胞NCM460细胞中的表达差异;免疫组织化学法分析ST6GAL1在CRC组织和对应癌旁组织中的表达差异。通过慢病毒转染细胞的方法构建稳定敲低或过表达ST6GAL1的HCT116细胞,通过划痕愈合实验检测细胞迁移能力,Transwell实验检测细胞侵袭能力,WB法检测细胞糖酵解相关蛋白、Notch1受体胞内段(Notch1 ICD)以及PI3K/AKT/mTOR通路磷酸化水平,细胞免疫荧光实验观察Notch1 ICD表达水平和进入细胞核情况;加入Notch1受体激动剂Jagged1处理HCT116细胞,通过WB法检测糖酵解相关蛋白、Notch1 ICD表达水平以及PI3K/AKT/mTOR通路磷酸化水平。结果:ST6GAL1在CRC组织和细胞中均表达上调(均P < 0.05)。与对照组和过表达组相比,敲低ST6GAL1导致HCT116细胞内Notch1 ICD表达水平和PI3K/AKT/mTORC1磷酸化水平显著降低,细胞糖酵解相关蛋白表达水平降低,细胞迁移和侵袭能力减弱(均P < 0.05);过表达ST6GAL1增加了HCT116细胞内Notch1 ICD表达水平并促进其进入细胞核,细胞糖酵解相关蛋白表达水平升高,细胞迁移和侵袭能力增强(均P < 0.05)。结论:ST6GAL1通过活化Notch1受体进而磷酸化激活PI3K/AKT/mTORC1通路,并增强CRC细胞糖酵解水平和迁移、侵袭能力。
4.Association between per- and polyfluoroalkyl substances and serum high-sensitivity C-reactive protein levels: Based on the National Health and Nutrition Examination Survey
Shuang MA ; Qian ZHANG ; Huirong DUAN ; Jinzhu YIN ; Tong WANG ; Qian GAO
Journal of Environmental and Occupational Medicine 2025;42(8):900-907
Background High-sensitivity C-reactive protein (hs-CRP) is a sensitive biomarker for cardiovascular disease (CVD) and can independently predict the risk of cardiovascular events. Although the association between per- and polyfluoroalkyl substances (PFAS) exposure and CVD risk has been widely reported, studies on the association between hs-CRP and PFAS remain limited. Objective To investigate the association between PFAS and hs-CRP levels, to provide a scientific basis for early identification and prevention of environment-related cardiovascular events. Methods This study utilized data from the National Health and Nutrition Examination Survey (NHANES) database (2015–2018). Based on predefined inclusion and exclusion criteria, a total of
5.Hearing loss prevalence and burden of disease in China: Findings from provincial-level analysis.
Yu WANG ; Yang XIE ; Minghao WANG ; Mengdan ZHAO ; Rui GONG ; Ying XIN ; Jia KE ; Ke ZHANG ; Shaoxing ZHANG ; Chen DU ; Qingchuan DUAN ; Fang WANG ; Tao PAN ; Furong MA ; Xiangyang HU
Chinese Medical Journal 2025;138(1):41-48
BACKGROUND:
Without timely and effective rehabilitation, hearing loss may profoundly affect human life quality. China has a large population of hearing-impaired individuals, which imposes a heavy health burden on society. Moreover, this population is projected to increase rapidly owing to China's aging society.
METHODS:
We used data from a population-representative epidemiological investigation of hearing loss and ear diseases in four Chinese provinces. We estimated the national prevalence using multiple linear regression of the age-group proportions and prevalence in 31 provinces with clustering analysis. We used years lived with disability (YLDs) to analyze the disease burden and forecasted the prevalence of hearing loss by 2060 in China.
RESULTS:
An estimated 115 million people had moderate-to-complete hearing loss in 2015 across the 31 provinces of China (8.4% of 1.37 billion people). Of these, 85.7% were older than age 50 years (99 million people) and 2.4% were younger than 20 years old (2.8 million people). Of all YLDs attributable to hearing loss, 68.9% were attributable to moderate-to-complete cases. By 2060, a projected 242 million people in China will have moderate-to-complete hearing loss, a 110.0% increase from 2015.
CONCLUSIONS
The hearing loss prevalence in China is high. Population aging and socioeconomic factors substantially affect the prevalence and severity of hearing loss and the disease burden. The prevalence and severity of hearing loss are unevenly distributed across different provinces. Future public health policies should take these trends and regional variations into account.
Humans
;
China/epidemiology*
;
Hearing Loss/epidemiology*
;
Prevalence
;
Middle Aged
;
Male
;
Female
;
Adult
;
Aged
;
Adolescent
;
Young Adult
;
Child
;
Child, Preschool
;
Infant
;
Aged, 80 and over
;
Cost of Illness
6.Chidamide triggers pyroptosis in T-cell lymphoblastic lymphoma/leukemia via the FOXO1/GSDME axis.
Xinlei LI ; Bangdong LIU ; Dezhi HUANG ; Naya MA ; Jing XIA ; Xianlan ZHAO ; Yishuo DUAN ; Fu LI ; Shijia LIN ; Shuhan TANG ; Qiong LI ; Jun RAO ; Xi ZHANG
Chinese Medical Journal 2025;138(10):1213-1224
BACKGROUND:
T-cell lymphoblastic lymphoma/acute lymphoblastic leukemia (T-LBL/ALL) is an aggressive form of hematological malignancy associated with poor prognosis in adult patients. Histone deacetylases (HDACs) are aberrantly expressed in T-LBL/ALL and are considered potential therapeutic targets. Here, we investigated the antitumor effect of a novel HDAC inhibitor, chidamide, on T-LBL/ALL.
METHODS:
HDAC1, HDAC2 and HDAC3 levels in T-LBL/ALL cell lines and patient samples were compared with those in normal controls. Flow cytometry, transmission electron microscopy, and lactate dehydrogenase release assays were conducted in Jurkat and MOLT-4 cells to assess apoptosis and pyroptosis. A specific forkhead box O1 (FOXO1) inhibitor was used to rescue pyroptosis and upregulated gasdermin E (GSDME) expression caused by chidamide treatment. The role of the FOXO1 transcription factor was evaluated by dual-luciferase reporter and chromatin immunoprecipitation assays. The efficacy of chidamide in vivo was evaluated in a xenograft mouse.
RESULTS:
The expression of HDAC1, HDAC2 and HDAC3 was significantly upregulated in T-LBL/ALL. Cell viability was obviously inhibited after chidamide treatment. Pyroptosis, characterized by cell swelling, pore formation on the plasma membrane and lactate dehydrogenase leakage, was identified as a new mechanism of chidamide treatment. Chidamide triggered pyroptosis through caspase 3 activation and GSDME transcriptional upregulation. Chromatin immunoprecipitation assays confirmed that chidamide led to the increased transcription of GSDME through a more relaxed chromatin structure at the promoter and the upregulation of FOXO1 expression. Moreover, we identified the therapeutic effect of chidamide in vivo .
CONCLUSIONS
This study suggested that chidamide exerts an antitumor effect on T-LBL/ALL and promotes a more inflammatory form of cell death via the FOXO1/GSDME axis, which provides a novel choice of targeted therapy for patients with T-LBL/ALL.
Humans
;
Pyroptosis/drug effects*
;
Forkhead Box Protein O1/genetics*
;
Aminopyridines/pharmacology*
;
Animals
;
Mice
;
Benzamides/pharmacology*
;
Cell Line, Tumor
;
Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy*
;
Phosphate-Binding Proteins/metabolism*
;
Histone Deacetylase Inhibitors/pharmacology*
;
Jurkat Cells
;
Histone Deacetylases/metabolism*
;
Apoptosis/drug effects*
;
Gasdermins
7.Dynamic gait parameters reveal long-term compensatory characteristics in knee joint function recovery following anterior cruciate ligament reconstruction: A retrospective cohort study.
Qitai LIN ; Zehao LI ; Meiming LI ; Yongsheng MA ; Wenming YANG ; Yugang XING ; Yang LIU ; Ruifeng LIANG ; Yixuan ZHANG ; Ruipeng ZHAO ; Wangping DUAN ; Pengcui LI ; Xiaochun WEI
Chinese Medical Journal 2025;138(22):3016-3018
8.Associations between statins and all-cause mortality and cardiovascular events among peritoneal dialysis patients: A multi-center large-scale cohort study.
Shuang GAO ; Lei NAN ; Xinqiu LI ; Shaomei LI ; Huaying PEI ; Jinghong ZHAO ; Ying ZHANG ; Zibo XIONG ; Yumei LIAO ; Ying LI ; Qiongzhen LIN ; Wenbo HU ; Yulin LI ; Liping DUAN ; Zhaoxia ZHENG ; Gang FU ; Shanshan GUO ; Beiru ZHANG ; Rui YU ; Fuyun SUN ; Xiaoying MA ; Li HAO ; Guiling LIU ; Zhanzheng ZHAO ; Jing XIAO ; Yulan SHEN ; Yong ZHANG ; Xuanyi DU ; Tianrong JI ; Yingli YUE ; Shanshan CHEN ; Zhigang MA ; Yingping LI ; Li ZUO ; Huiping ZHAO ; Xianchao ZHANG ; Xuejian WANG ; Yirong LIU ; Xinying GAO ; Xiaoli CHEN ; Hongyi LI ; Shutong DU ; Cui ZHAO ; Zhonggao XU ; Li ZHANG ; Hongyu CHEN ; Li LI ; Lihua WANG ; Yan YAN ; Yingchun MA ; Yuanyuan WEI ; Jingwei ZHOU ; Yan LI ; Caili WANG ; Jie DONG
Chinese Medical Journal 2025;138(21):2856-2858
9.Research progress on natural small molecule compound inhibitors of NLRP3 inflammasome.
Tian-Yuan ZHANG ; Xi-Yu CHEN ; Xin-Yu DUAN ; Qian-Ru ZHAO ; Lin MA ; Yi-Qi YAN ; Yu WANG ; Tao LIU ; Shao-Xia WANG
China Journal of Chinese Materia Medica 2025;50(3):644-657
In recent years, there has been a growing interest in the research on NOD-like receptor thermal protein domain associated protein 3(NLRP3) inflammasome inhibitors in the treatment of inflammatory diseases. The NLRP3 inflammasome is integral to the innate immune response, and its abnormal activation can lead to the release of pro-inflammatory cytokine, consequently facilitating the progression of various pathological conditions. Therefore, investigating the pharmacological inhibition pathway of the NLRP3 inflammasome represents a promising strategy for the treatment of inflammation-related diseases. Currently, the Food and Drug Administration(FDA) has not approved drugs targeting the NLRP3 inflammasome for clinical use due to concerns regarding liver toxicity and gastrointestinal side effects associated with chemical small molecule inhibitors in clinical trials. Natural small molecule compounds such as polyphenols, flavonoids, and alkaloids are ubiquitously found in animals, plants, and other natural substances exhibiting pharmacological activities. Their abundant sources, intricate and diverse structures, high biocompatibility, minimal adverse reactions, and superior biochemical potency in comparison to synthetic compounds have attracted the attention of extensive scholars. Currently, certain natural small molecule compounds have been demonstrated to impede the activation of the NLRP3 inflammasome via various action mechanisms, so they are viewed as the innovative, feasible, and minimally toxic therapeutic agents for inhibiting NLRP3 inflammasome activation in the treatment of both acute and chronic inflammatory diseases. Hence, this study systematically examined the effects and potential mechanisms of natural small molecule compounds derived from traditional Chinese medicine on the activation of NLRP3 inflammasomes at their initiation, assembly, and activation stages. The objection is to furnish theoretical support and practical guidance for the effective clinical application of these natural small molecule inhibitors.
NLR Family, Pyrin Domain-Containing 3 Protein/metabolism*
;
Inflammasomes/metabolism*
;
Inflammation/drug therapy*
;
Anti-Inflammatory Agents/therapeutic use*
;
Humans
;
Animals
;
Disease Models, Animal
;
Biological Products/therapeutic use*
;
Drug Discovery
;
Medicine, Chinese Traditional/methods*
10.The systemic inflammatory response index as a risk factor for all-cause and cardiovascular mortality among individuals with coronary artery disease: evidence from the cohort study of NHANES 1999-2018.
Dao-Shen LIU ; Dan LIU ; Hai-Xu SONG ; Jing LI ; Miao-Han QIU ; Chao-Qun MA ; Xue-Fei MU ; Shang-Xun ZHOU ; Yi-Xuan DUAN ; Yu-Ying LI ; Yi LI ; Ya-Ling HAN
Journal of Geriatric Cardiology 2025;22(7):668-677
BACKGROUND:
The association of systemic inflammatory response index (SIRI) with prognosis of coronary artery disease (CAD) patients has never been investigated in a large sample with long-term follow-up. This study aimed to explore the association of SIRI with all-cause and cause-specific mortality in a nationally representative sample of CAD patients from United States.
METHODS:
A total of 3386 participants with CAD from the National Health and Nutrition Examination Survey (NHANES) 1999-2018 were included in this study. Cox proportional hazards model, restricted cubic spline (RCS), and receiver operating characteristic curve (ROC) were performed to investigate the association of SIRI with all-cause and cause-specific mortality. Piece-wise linear regression and sensitivity analyses were also performed.
RESULTS:
During a median follow-up of 7.7 years, 1454 all-cause mortality occurred. After adjusting for confounding factors, higher lnSIRI was significantly associated with higher risk of all-cause (HR = 1.16, 95% CI: 1.09-1.23) and CVD mortality (HR = 1.17, 95% CI: 1.05-1.30) but not cancer mortality (HR = 1.17, 95% CI: 0.99-1.38). The associations of SIRI with all-cause and CVD mortality were detected as J-shaped with threshold values of 1.05935 and 1.122946 for SIRI, respectively. ROC curves showed that lnSIRI had robust predictive effect both in short and long terms.
CONCLUSIONS
SIRI was independently associated with all-cause and CVD mortality, and the dose-response relationship was J-shaped. SIRI might serve as a valid predictor for all-cause and CVD mortality both in the short and long terms.

Result Analysis
Print
Save
E-mail