1.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
2.A novel fully human LAG-3 monoclonal antibody LBL-007 combined with PD-1 antibody inhibits proliferation, migration and invasion of tumor cells via blocking NF-κB pathway.
Huinan ZHOU ; Jianfei LIU ; Chenglin WU ; Kewei QIN ; Lijun ZHOU
Chinese Journal of Cellular and Molecular Immunology 2025;41(5):398-405
Objective To investigate the effects of LBL-007, a novel fully human lymphocyte activation gene 3 (LAG-3) monoclonal antibody, in combination with programmed cell death protein 1 (PD-1) antibody, on the invasion, migration and proliferation of tumor cells, and to elucidate the underlying mechanisms. Methods Human lymphocyte cells Jurkat were co-cultured with A549 and MGC803 tumor cell lines and treated with the isotype control antibody human IgG, LBL-007, anti-PD-1 antibody BE0188, or tumor necrosis factor-alpha (TNF-α, the NF-κB signaling pathway agonist). Tumor cell proliferation was assessed using a colony formation assay; invasion was measured by TranswellTM assay; migration was evaluated using a wound healing assay. Western blotting was employed to determine the expression levels of NF-κB pathway-related proteins: IκB inhibitor kinase alpha (Ikkα), phosphorylated Ikkα (p-IKKα), NF-κB subunit p65, phosphorylated p65 (p-p65), NF-κB Inhibitor Alpha (IκBα), phosphorylated IκBα (p-IκBα), matrix metalloproteinase 9 (MMP9), and MMP2. Results Compared with the control and IgG isotype groups, LBL-007 and BE0188 significantly reduced tumor cell proliferation, invasion, and migration. They also decreased the phosphorylation of p-IKKα, p-p65 and p-IκBα, and the expression of MMP9 and MMP2 of tumor cells in the co-culture system. The combined treatment of LBL-007 and BE0188 enhanced inhibitory effects. Treatment with the NF-κB signaling pathway agonist TNF-α reversed the suppressive effects of LBL-007 and BE0188 on tumor cell proliferation, invasion, migration, and NF-κB signaling. Conclusion LBL-007 and anti-PD-1 antibody synergistically inhibit the invasion, migration, and proliferation of A549 and MGC803 tumor cells by blocking the NF-κB signaling pathway.
Humans
;
Cell Proliferation/drug effects*
;
Cell Movement/drug effects*
;
Signal Transduction/drug effects*
;
NF-kappa B/metabolism*
;
Neoplasm Invasiveness
;
Antibodies, Monoclonal/pharmacology*
;
Programmed Cell Death 1 Receptor/antagonists & inhibitors*
;
Cell Line, Tumor
;
Antigens, CD/immunology*
;
Lymphocyte Activation Gene 3 Protein
;
A549 Cells
;
I-kappa B Kinase/metabolism*
;
Jurkat Cells
;
Matrix Metalloproteinase 9/metabolism*
3.Effect and mechanism of LncRNA EFRL on homocysteine-induced atherosclerosis in macrophage efferocytosis.
Jiaqi YANG ; Zhenghao ZHANG ; Fang MA ; Tongtong XIA ; Honglin LIU ; Jiantuan XIONG ; Shengchao MA ; Yideng JIANG ; Yinju HAO
Chinese Journal of Cellular and Molecular Immunology 2025;41(7):577-584
Objective To investigate the effect and mechanism of Efferocytosis Relatived LncRNA (EFRL) on homocysteine-induced atherosclerosis in macrophage efferocytosis. Methods RAW264.7 cells were cultured in vitro, and the Control group (0 μmol/L Hcy) and Hcy intervention group (100 μmol/L Hcy) were set up. After GapmeR transfection of macrophages with Hcy intervention, EFRL knockdown negative control group (Hcy combined with LNA-NC) and EFRL knockdown group (Hcy combined with LNA-EFRL) were set up. High-throughput sequencing was applied for different expression of LncRNA MSTRG. 88917.16 (EFRL), UCSC was used to analyze its conservation, CPC and CPAT were used to analyze its ability to encode proteins, and GO and KEGG were used to analyze related biological functions. The localization of LncRNA EFRL in macrophages was analyzed by nucleoplasmic separation and RNA-FISH. Quantitative real-time PCR was used to detect the expression levels of LncRNA EFRL and its target gene SPAST in Hcy-treated macrophages. The apoptosis rate of Jurkat cells induced by UV was detected by flow cytometry. In vitro efferocytosis assay combined with immunofluorescence technique was used to analyze macrophage efferocytosis. ELISA was used to detect the levels of interleukin 1β(IL-1β) and IL-18. Results The new LncRNA MSTRG.88917.16 was identified and named EFRL(Efferocytosis Relatived LncRNA). UCSC, CPC and CPAT analyses showed that LncEFRL is highly conserved and does not have the ability to encode proteins. GO and KEGG analyses suggested that LncEFRL may be involved in macrophage efferocytosis. LncRNA EFRL was localized in the nucleus of macrophages as determined by nucleoplasmic separation and RNA-FISH. In comparison to the Control group, the expression levels of LncRNA EFRL and its target gene SPAST in the Hcy group were increased. In comparison to the Control group (0 min), the apoptosis rate of the experimental group (15, 30 min) Annexin V is more than 85%. Compared with Hcy combined with LNA-NC group, Hcy combined with LNA-EFRL group had enhanced macrophage efferocytosis and reduced levels of inflammatory factors. Compared with Hcy combined with LNA-NC group, the expression level of SPAST in Hcy combined with LNA-EFRL group was decreased. Conclusion Inhibition of EFRL expression can alleviate the process of Hcy inhibiting macrophage efferocytosis, and the mechanism is related to the regulation of the downstream target gene SPAST by EFRL.
RNA, Long Noncoding/physiology*
;
Animals
;
Homocysteine
;
Mice
;
Macrophages/drug effects*
;
Humans
;
RAW 264.7 Cells
;
Atherosclerosis/chemically induced*
;
Apoptosis/genetics*
;
Phagocytosis/genetics*
;
Jurkat Cells
;
Interleukin-1beta/genetics*
;
Efferocytosis
4.Investigation of the Effects of Arsenic Trioxide Combined with Deslorelin on Proliferation and Apoptosis of Jurkat Cells Based on Wnt/β-Catenin Pathway.
Journal of Experimental Hematology 2025;33(3):640-647
OBJECTIVE:
To investigate the effect of Arsenic trioxide (ATO) combined with Norcantharidin (NCTD) on the proliferation and apoptosis of Jurkat cells, and to evaluate its effect on the proliferation and apoptosis of acute T-lymphoblastic leukemia (T-ALL) based on the Wnt/β-catenin signaling pathway.
METHODS:
Jurkat cell lines were used as the study subjects and treated with different concentrations of ATO (0, 2, 4, 8, 16 μmol/L) and NCTD (0, 10, 25, 50, 100 μmol/L) for 72 hours, and the cell proliferation was detected by CCK-8. Meanwhile, flow cytometry was used to detect the apoptosis rate, EdU staining to detect cell proliferation viability, cell clone formation assay to assess cell cloning ability, Transwell assay to assess cell invasion ability, and Western blot to detect apoptosis and the expression of Wnt/β-catenin signaling pathway-related proteins.
RESULTS:
Compared with the control group, both ATO and NCTD effectively inhibited Jurkat cell proliferation when used alone, and the inhibition effect was more significant when used in combination ( P < 0.05). The combination significantly increased the apoptosis rate of Jurkat cells ( P < 0.05). Meanwhile, the combination significantly decreased the proliferation vitality and clone formation ability of the cells ( P < 0.05), and inhibited the invasion ability of Jurkat cells ( P < 0.05). Western blot analysis showed that the combination of ATO and NCTD significantly up-regulated the expression of pro-apoptotic proteins Bax and E-cadherin, and down-regulated the expression of anti-apoptotic proteins Bcl-2, c-myc and Cyclin D1 ( P < 0.05).
CONCLUSION
The combination of ATO and NCTD had a synergistic effect in inhibiting proliferation and promoting apoptosis in Jurkat cells, which may be related to the inhibition of Wnt/β-catenin signaling pathway.
Humans
;
Apoptosis/drug effects*
;
Jurkat Cells
;
Cell Proliferation/drug effects*
;
Arsenic Trioxide
;
Wnt Signaling Pathway/drug effects*
;
Bridged Bicyclo Compounds, Heterocyclic/pharmacology*
;
beta Catenin/metabolism*
;
Arsenicals/pharmacology*
;
Oxides/pharmacology*
5.A reporter gene assay for determining antibody-dependent cell-mediated phagocytosis activity of HER2-targeted antibody drug conjugate.
Ying CHEN ; Can WANG ; Qin ZHAO ; Mingren WANG ; Tiantian LI ; Shanshan DONG ; Hong SHAO ; Weidong XU
Chinese Journal of Biotechnology 2025;41(8):3122-3130
To develop a method for determining the antibody-dependent cell-mediated phagocytosis (ADCP) activity of human epidermal growth factor receptor 2 (HER2)-targeted antibody drug conjugate (ADC) based on the reporter gene assay, we established an ADCP activity assay with Jurkat/NFAT/FcγRIIa cells as the effector cells and BT474 as the target cells. Then, the target cell density, the ratio of effector to target cells, the target cell adhesion time, the incubation time for drug administration, and the induction time after adding effector cells were optimized by the method of design of experiment (DOE). The method showed a significant dose-response relationship, which was complied with the four-parameter equation: y=(A-D)/[1+(x/C)B]+D. The durability ranges of the target cell density, the ratio of effector to target cells, the target cell adhesion time, the incubation time for drug administration, and the induction time after adding effector cells were (2.5-4.0)×105 cells/mL, 3-5, 1.0-2.0 h, 0 h, and 5.0-6.0 h, respectively. The results of the methodological validation showed that the linear equation was y=1.106 8x-0.011 6, r=0.969 2. The established method showed the relative accuracy ranging from -6.59% to 2.98% and the geometric coefficient of variation less than 11% in the intermediate precision test. Furthermore, the method was target-specific. The method was then applied to the determination of ADCP activity of HER2-targeted ADC, demonstrating the result of (103.5±5.7)%. We developed a reporter gene assay for determining the ADCP activity of HER2-targeted ADC and the assay demonstrated high accuracy and good reproducibility, which proposes a highly efficient and approache for evaluating ADCP effect of this HER2-targeted ADC, and also provides a referable technique for characterizing the Fc effector functions of ADCs with diverse targets.
Humans
;
Receptor, ErbB-2/immunology*
;
Phagocytosis/drug effects*
;
Immunoconjugates/immunology*
;
Genes, Reporter
;
Antibody-Dependent Cell Cytotoxicity
;
Jurkat Cells
6.Ku80 Inhibition Affects the Chemotherapeutic Sensitivity of T-Acute Lymphoblastic Leukemia Cell Line Jurkat.
Journal of Experimental Hematology 2024;32(6):1689-1695
OBJECTIVE:
To investigate the influence of Ku80 inhibition on the chemotherapeutic sensitivity of the T-acute lymphoblastic leukemia(T-ALL) cell line Jurkat, and to explore the potential mechanism.
METHODS:
The transcription and expression level of Ku80 in 6 hematological malignant cell lines were detected by RT-qPCR and Western blot, respectively. The expression of Ku80 in Jurkat cells was detected by Western blot after transfection with the recombinant shKu80 lentiviral vector. The proliferation capacity of Jurkat cells was explored by CCK-8 after Ku80 inhibition. The colony formation ability, apoptosis, and γH2AX(a protein marker of DNA damage) expression in Jurkat cells were investigated after Ku80 silencing and co-treated with etoposide(VP16) for 4 hours through soft agar assay, flow cytometry and Western blot, respectively.
RESULTS:
The mRNA level and protein expression of Ku80 were both highest in Jurkat among 6 hematological malignant cell lines. Ku80 expression was successfully down regulated in Jurkat cells after relative plasmid transfected. The proliferative ability of cells was significantly decreased after Ku80 inhibition(P < 0.05). The colony formation capacity of Jurkat cells was obviously reduced and the cells apoptosis and γH2AX expression were increased after Ku80 inhibition, with or without VP16 incubation.
CONCLUSION
Targeted silencing of Ku80 could enhance the sensitivity of VP16 in Jurkat cells, which might be associated with the elevated level of DNA damage accumulation.
Humans
;
Ku Autoantigen/metabolism*
;
Jurkat Cells
;
Apoptosis/drug effects*
;
Cell Proliferation
;
Etoposide/pharmacology*
;
DNA Damage
;
Cell Line, Tumor
;
Precursor T-Cell Lymphoblastic Leukemia-Lymphoma
7.Study on the Regulation of Chidamide on CD8+T Cells in T-cell Acute Lymphoblastic Leukemia.
Miao-Ming YAN ; Zhao-Xuan LI ; Chong CHEN ; Wei ZHANG ; Dao-Bin ZHOU
Journal of Experimental Hematology 2023;31(1):71-75
OBJECTIVE:
To explore the regulatory effect of chidamide on CD8+ T cells in T-cell acute lymphoblastic leukemia.
METHODS:
The expression levels of CXCL9 and CXCL3 mRNA in Jurkat cells, lymphocytes treated with chidamide and lymphocytes co-cultured with chidamide-treated Jurkat cells were detected by fluorescence quantitative PCR. The proportion of CD8+ T cells in lymphocytes treated with chidamide and lymphocytes co-cultured with chidamide-treated Jurkat cells was determined by flow cytometry.
RESULTS:
Chidamide upregulated CXCL9 mRNA expression in Jurkat cell line in a dose-dependent manner (r=0.950). The mRNA expression of CXCL9 in chidamide 5 μmol/L group was 164 times higher than that in control group. Chidamide upregulated CXCL9 mRNA expression in lymphocytes, but the up-regulated level was significantly lower than that in Jurkat cell line treated with the same concentration of chidamide. Co-culture with chidamide treated Jurkat cells upregulated the proportion of CD8+ T cells in lymphocytes.
CONCLUSION
In T-cell acute lymphoblastic leukemia, chidamide may increase the concentration of CXCL9 in the tumor microenvironment by up-regulating the expression of CXCL9 in tumor cells, leading to an increase in the number of CD8+ T cells.
Humans
;
CD8-Positive T-Lymphocytes
;
Precursor T-Cell Lymphoblastic Leukemia-Lymphoma
;
Aminopyridines/pharmacology*
;
Jurkat Cells
;
RNA, Messenger
;
Cell Line, Tumor
;
Apoptosis
;
Tumor Microenvironment
8.Effects of Methionine Restriction on Proliferation, Cell Cycle, and Apoptosis of Human Acute Leukemia Cells.
Yu-Jie HE ; Si-Shu YU ; Bin ZHANG ; Mei-Rong LI ; Li-Jun XU ; Long-Ming LIANG ; Zheng-Gang ZHAO ; Zi-Jian ZHAO ; Su-Jin ZHOU ; Fang-Hong LI
Journal of Experimental Hematology 2023;31(5):1290-1295
OBJECTIVE:
To investigate the effects of methionine restriction on proliferation, cell cycle and apoptosis of human acute leukemia cells.
METHODS:
Cell Counting Kit-8 (CCK-8) assay was used to detect the effect of methionine restriction on HL-60 and Jurkat cells proliferation. The effect of methionine restriction on cell cycle of HL-60 and Jurkat cells was examined by PI staining. Annexin V-FITC / PI double staining was applied to detect apoptosis of HL-60 and Jurkat cells following methionine restriction. The expression of cell cycle-related proteins cyclin B1, CDC2 and apoptosis-related protein Bcl-2 was evaluated by Western blot assay.
RESULTS:
Methionine restriction significantly inhibited the proliferation of HL-60 and Jurkat cells in a time-dependent manner (HL-60: r =0.7773, Jurkat: r =0.8725), arrested the cells at G2/M phase (P < 0.001), and significantly induced apoptosis of HL-60 and Jurkat cells (HL-60: P < 0.001; Jurkat: P < 0.05). Furthermore, Western blot analysis demonstrated that methionine restriction significantly reduced the proteins expression of Cyclin B1 (P < 0.05), CDC2 (P < 0.01) and Bcl-2 (P < 0.001) in HL-60 and Jurkat cells.
CONCLUSION
Acute leukemia cells HL-60 and Jurkat exhibit methionine dependence. Methionine restriction can significantly inhibit the proliferation, promote cell cycle arrest and induce apoptosis of HL-60 and Jurkat cells, which suggests that methionine restriction may be a potential therapeutic strategy for acute leukemia.
Humans
;
Cyclin B1/pharmacology*
;
Cell Proliferation
;
Methionine/pharmacology*
;
Cell Cycle
;
Apoptosis
;
Leukemia, Myeloid, Acute
;
Cell Division
;
Cell Cycle Proteins
;
Jurkat Cells
;
Proto-Oncogene Proteins c-bcl-2/metabolism*
;
HL-60 Cells
9.Deficiency of two-pore segment channel 2 contributes to systemic lupus erythematosus via regulation of apoptosis and cell cycle.
Keke LI ; Jingkai XU ; Ke XUE ; Ruixing YU ; Chengxu LI ; Wenmin FEI ; Xiaoli NING ; Yang HAN ; Ziyi WANG ; Jun SHU ; Yong CUI
Chinese Medical Journal 2022;135(4):447-455
BACKGROUND:
Systemic lupus erythematosus (SLE) is a complex autoimmune disease, and the mechanism of SLE is yet to be fully elucidated. The aim of this study was to explore the role of two-pore segment channel 2 (TPCN2) in SLE pathogenesis.
METHODS:
Quantitative reverse transcription polymerase chain reaction (qRT-PCR) was used to detect the expression of TPCN2 in SLE. We performed a loss-of-function assay by lentiviral construct in Jurkat and THP-1 cell. Knockdown of TPCN2 were confirmed at the RNA level by qRT-PCR and protein level by Western blotting. Cell Count Kit-8 and flow cytometry were used to analyze the cell proliferation, apoptosis, and cell cycle of TPCN2-deficient cells. In addition, gene expression profile of TPCN2-deficient cells was analyzed by RNA sequencing (RNA-seq).
RESULTS:
TPCN2 knockdown with short hairpin RNA (shRNA)-mediated lentiviruses inhibited cell proliferation, and induced apoptosis and cell-cycle arrest of G2/M phase in both Jurkat and THP-1 cells. We analyzed the transcriptome of knockdown-TPCN2-Jurkat cells, and screened the differential genes, which were enriched for the G2/M checkpoint, complement, and interleukin-6-Janus kinase-signal transducer and activator of transcription pathways, as well as changes in levels of forkhead box O, phosphatidylinositol 3-kinase/protein kinase B/mechanistic target of rapamycin, and T cell receptor pathways; moreover, TPCN2 significantly influenced cellular processes and biological regulation.
CONCLUSION
TPCN2 might be a potential protective factor against SLE.
Apoptosis/genetics*
;
Cell Division
;
Humans
;
Jurkat Cells
;
Lupus Erythematosus, Systemic/genetics*
;
RNA, Small Interfering/genetics*
10.Effect of Down-Regulation of LncRNA-HOTAIRM1 to Proliferation, Apoptosis and KIT/AKT Pathway of Jurkat Cells.
Yu-Ru LI ; Wen-Jing YAN ; Li-Li CAI ; Xin-Li DENG
Journal of Experimental Hematology 2021;29(4):1123-1128
OBJECTIVE:
To observe the effects of down-regulation of long non-coding RNA HOX antisense intergenic RNA myeloid 1 (LncRNA-HOTAIRM1) to the proliferation and apoptosis of Jurkat in human leukemia T lymphocytes, and explore its mechanism.
METHODS:
Jurkat cells were cultured in vitro and randomly divided into control group, HOTAIRM1 siRNA-NC group and HOTAIRM1 siRNA group; the expressions of LncRNA-HOTAIRM1 mRNA, KIT receptor tyrosine kinase (KIT) mRNA and serine threonine kinase (AKT) mRNA in Jurkat cells were detected by real-time fluorescence quantification (RT-qPCR); the proliferation of Jurkat cells in each groups was detected by CCK-8 method; the apoptosis of Jurkat cells in each groups was detected by Annexin V-FITC/PI double staining; the expressions of KIT, AKT, p-KIT, p-AKT, B-lymphoma-2 gene (BCL-2) and Caspase-3 were detected by Western blot.
RESULTS:
Compared with the cells in the control group and HOTAIRM1 siRNA-NC group, the expression level of LncRNA-HOTAIRM1 mRNA, cell survival rate, expression levels of KIT mRNA, AKT mRNA, p-KIT, p-AKT and BCL-2 proteins in Jurkat cells in HOTAIRM1 siRNA group were significantly lower (P<0.05), while the expression level of Cleared Caspase-3 protein and Jurkat cell apoptosis rate were significantly higher (P<0.05).
CONCLUSION
LncRNA-HOTAIRM1 may inhibit Jurkat cell proliferation and induce apoptosis through KIT/AKT signaling pathway.
Apoptosis
;
Cell Proliferation
;
Down-Regulation
;
Humans
;
Jurkat Cells
;
Proto-Oncogene Proteins c-akt/metabolism*
;
RNA, Long Noncoding/genetics*

Result Analysis
Print
Save
E-mail