1.Regulatory Mechanism of Mangiferin Combined with Bortezomib on Malignant Biological Behavior of Burkitt Lymphoma and Its Effect on Expression of CXC Chemokine Receptors.
Zhi-Min YAN ; Yan-Quan LIU ; Qing-Lin XU ; Jie LIN ; Xin LIU ; Qiu-Ping ZHU ; Xin-Ji CHEN ; Ting-Bo LIU ; Xiao-Lan LIAN
Journal of Experimental Hematology 2023;31(5):1394-1402
OBJECTIVE:
To analyze the effects of mangiferin combined with bortezomib on the proliferation, invasion, apoptosis and autophagy of human Burkitt lymphoma Raji cells, as well as the expression of CXC chemokine receptors (CXCRs) family, and explore the molecular mechanism between them to provide scientific basis for basic research and clinical work of Burkitt lymphoma.
METHODS:
Raji cells were intervened with different concentrations of mangiferin and bortezomib alone or in combination, then cell proliferation was detected by CCK-8 assay, cell invasion ability was detected by Transwell chamber method, cell apoptosis was detected by Annexin V/PI double-staining flow cytometry, apoptosis, autophagy and Akt/mTOR pathway protein expression were detected by Western blot, and the expression changes of CXCR family was detected by real-time quantitative PCR (RT-qPCR).
RESULTS:
Different concentrations of mangiferin intervened Raji cells for different time could inhibit cell viability in a concentration- and time-dependent manner (r =-0.682, r =-0.836). When Raji cells were intervened by combination of mangiferin and bortezomib, compared with single drug group, the proliferation and invasion abilities were significantly decreased, while the apoptosis level was significantly increased (P <0.01). Mangiferin combined with bortezomib could significantly up-regulate the expression of pro-apoptotic protein Bax and down-regulate the expression of anti-apoptotic protein Bcl-2 after intervention in Raji cells. Caspase-3 was also hydrolyzed and activated, and then induced the apoptosis of Raji cells. Mangiferin combined with bortezomib could up-regulate the expression of LC3Ⅱ protein in Raji cells, and the ratio of LC3Ⅱ/LC3Ⅰ in cells was significantly up-regulated compared with single drug or control group (P <0.01). Mangiferin combined with bortezomib could significantly inhibit the phosphorylation levels of Akt and mTOR, inhibit the proliferation and invasion of Raji cells by inhibiting Akt/mTOR pathway, and induce cell autophagy and apoptosis. Mangiferin and bortezomib could down-regulate the expressions of CXCR4 and CXCR7 mRNA after single-agent intervention in Raji cells, and the down-regulations of CXCR4 and CXCR7 mRNA expression were more significant when the two drugs were combined (P <0.01). Mangiferin alone or combined with bortezomib had no significant effect on CXCR5 mRNA expression in Raji cells (P >0.05), while the combination of the two drugs could down-regulate the expression of CXCR3 (P <0.05).
CONCLUSION
Mangiferin combined with bortezomib can synergistically inhibit the proliferation and invasion of Raji cells, and induce autophagy and apoptosis. The mechanism may be related to the inhibition of Akt/mTOR signaling pathway, down-regulation of anti-apoptotic protein Bcl-2 and up-regulation of pro-apoptotic protein Bax, and the inhibition of the expression of CXCR family.
Humans
;
Antineoplastic Agents/therapeutic use*
;
Apoptosis/drug effects*
;
Apoptosis Regulatory Proteins/immunology*
;
Autophagy/immunology*
;
bcl-2-Associated X Protein/immunology*
;
Bortezomib/therapeutic use*
;
Burkitt Lymphoma/immunology*
;
Cell Line, Tumor
;
Cell Proliferation/drug effects*
;
Drug Therapy, Combination
;
Proto-Oncogene Proteins c-akt
;
Proto-Oncogene Proteins c-bcl-2
;
Receptors, CXCR/immunology*
;
RNA, Messenger
;
TOR Serine-Threonine Kinases
;
Xanthones/therapeutic use*
2.Development and validation of a risk-prediction model for immune-related adverse events in patients with non-small-cell lung cancer receiving PD-1/PD-L1 inhibitors.
Qing QIU ; Chenghao WU ; Wenxiao TANG ; Longfei JI ; Guangwei DAI ; Yuzhen GAO ; Enguo CHEN ; Hanliang JIANG ; Xinyou XIE ; Jun ZHANG
Journal of Zhejiang University. Science. B 2023;24(10):935-942
Lung cancer remains the leading cause of cancer deaths worldwide and is the most common cancer in males. Immune-checkpoint inhibitors (ICIs) that target programmed cell death protein-1 (PD-1) or programmed cell death-ligand 1 (PD-L1) have achieved impressive efficacy in the treatment of non-small-cell lung cancer (NSCLC) (Pardoll, 2012; Champiat et al., 2016; Gao et al., 2022). Although ICIs are usually well tolerated, they are often accompanied by immune-related adverse events (irAEs) (Doroshow et al., 2019). Non-specific activation of the immune system produces off-target immune and inflammatory responses that can affect virtually any organ or system (O'Kane et al., 2017; Puzanov et al., 2017). Compared with adverse events caused by chemotherapy, irAEs are often characterized by delayed onset and prolonged duration and can occur in any organ at any stage of treatment, including after cessation of treatment (Puzanov et al., 2017; von Itzstein et al., 2020). They range from rash, pneumonitis, hypothyroidism, enterocolitis, and autoimmune hepatitis to cardiovascular, hematological, renal, neurological, and ophthalmic irAEs (Nishino et al., 2016; Kumar et al., 2017; Song et al., 2020). Hence, we conducted a retrospective study to identify validated factors that could predict the magnitude of the risk of irAEs in patients receiving PD-1/PD-L1 inhibitors; our approach was to analyze the correlation between the clinical characteristics of patients at the start of treatment and relevant indicators such as hematological indices and the risk of developing irAEs. Then, we developed an economical, practical, rapid, and simple model to assess the risk of irAEs in patients receiving ICI treatment, as early as possible.
Male
;
Humans
;
Carcinoma, Non-Small-Cell Lung/drug therapy*
;
Lung Neoplasms/drug therapy*
;
Immune Checkpoint Inhibitors/adverse effects*
;
Programmed Cell Death 1 Receptor
;
Retrospective Studies
;
Apoptosis
3.MiR -18a -5p aggravates homocysteine -induced myocardial injury via autophagy.
Juan YIN ; Longlong HU ; Xueling HAN ; Lu CHEN ; Lingling YU ; Yinhui LU
Journal of Central South University(Medical Sciences) 2023;48(1):24-33
OBJECTIVES:
Hyperhomocysteinaemia (Hcy) is an independent risk factor for cardiovascular and cerebrovascular diseases. MicroRNA (miR)-18a-5p is closely related to cardiovascular diseases. This study aims to investigate the effects of miR-18a-5p on homocysteine (Hcy)-induced myocardial cells injury.
METHODS:
H9c2 cells were transfected with miR-18a-5p mimic/miR-18a-5p mimic negative control (NC) or combined with Hcy for intervention, and untreated cells were set as a control group. The transfection efficiency was verified by real-time RT-PCR, and cell counting kit-8 (CCK-8) assay was used to determine cell viability. Flow cytometry was used to detect apoptosis and reactive oxygen species (ROS) levels. Western blotting was performed to measure the protein levels of microtubule-associated protein 1 light chain 3 (LC3)-I, LC3-II, Beclin1, p62, Bax, Bcl-2, and Notch2. Dual luciferase reporter assay was used to detect the interaction of miR-18a-5p with Notch2.
RESULTS:
Compared with the control, treatment with Hcy or transfection with miR-18a-5p mimic alone, or combined treatment with Hcy and miR-18a-5p mimic/miR-18a-5p mimic NC significantly reduced the H9c2 cell viability, promoted apoptosis and ROS production, up-regulated the expressions of Bax and Beclin, down-regulated the expressions of Bcl-2, p62, and Notch2, and increased the ratio of LC3-II/LC3-I (all P<0.05). Compared with the combined intervention of miR-18a-5p mimic NC and Hcy group, the above indexes were more significantly changed in the combined intervention of miR-18a-5p mimic and Hcy group, and the difference between the 2 groups was statistically significant (all P<0.05). There is a targeted binding between Notch2 and miR-18a-5p.
CONCLUSIONS
MiR-18a-5p could induce autophagy and apoptosis via increasing ROS production in cardiomyocytes, and aggravate Hcy-induced myocardial injury. Notch2 is a target of miR-18a-5p.
Apoptosis/genetics*
;
Autophagy/genetics*
;
bcl-2-Associated X Protein
;
MicroRNAs/metabolism*
;
Proto-Oncogene Proteins c-bcl-2/genetics*
;
Reactive Oxygen Species
;
Rats
;
Animals
;
Myocytes, Cardiac/drug effects*
;
Homocysteine/adverse effects*
;
Hyperhomocysteinemia
4.Resveratrol improves cognitive function in severely burned rats by inhibiting hippocampal NF-κB/JNK pathway.
Liang XING ; Wei WEI ; Cuina ZHANG ; Bingquan GAO ; Jianke FENG ; Leilei MA
Chinese Journal of Cellular and Molecular Immunology 2023;39(7):604-609
Objective To investigate the protective effect of resveratrol (RSV) on improving cognitive function in severely burned rats and its possible mechanism. Methods 18 male SD rats aged 18-20 months were randomly divided into 3 groups: control group, model group and RSV group, with 6 rats in each group. After successful modeling, the rats in RSV group were gavaged once daily with RSV (20 mg/kg). Meanwhile, the rats in control group and model group were gavaged once daily with an equal volume of sodium chloride solution. After 4 weeks, the cognitive function of all rats was estimated by Step-down Test. The concentration of tumor necrosis factor α (TNF-α) and interleukin 6 (IL-6) protein in serum of rats were detected by ELISA. The expression of IL-6, TNF-α mRNA and protein were estimated by real-time PCR and Western blotting. The apoptosis of hippocampal neurons was tested by terminal deoxynuclectidyl transferase-mediated dUTP-biotin nick end labeling assay (TUNEL). The expression of nuclear transcription factor-κB (NF-κB)/c-Jun N-terminal kinase (JNK) pathway-related proteins in hippocampus were assessed by Western blotting. Results Compared with the rats in model group, rats in RSV group exhibited improved cognitive function. Consistently, the rats in RSV group had a reduced concentration of TNF-α and IL-6 in serum, decreased mRNA and protein expressions of TNF-α and IL-6 in hippocampus, and decreased apoptosis rate and relative expression of p-NF-κB p65/NF-κB p65 and p-JNK/JNK in hippocampal neurons. Conclusion RSV alleviates inflammatory response and hippocampal neuronal apoptosis by inhibiting NF-κB/JNK pathway, thereby improving cognitive function in severely burned rats.
Resveratrol/pharmacology*
;
Male
;
Animals
;
Rats
;
Rats, Sprague-Dawley
;
Burns/drug therapy*
;
Cognition/drug effects*
;
Hippocampus/metabolism*
;
MAP Kinase Signaling System
;
NF-kappa B/metabolism*
;
Tumor Necrosis Factor-alpha/blood*
;
Interleukin-6/blood*
;
Neurons/drug effects*
;
Apoptosis
5.Role of selenoprotein M knockdown in the melatonin antagonism of nickel-induced apoptosis and endoplasmic reticulum stress in mouse heart.
Xintong ZHANG ; Xiaoxue GAI ; Lihua XU ; Wenxue MA ; Qiaohan LIU ; Bendong SHI ; Cheng FANG ; Jingzeng CAI ; Ziwei ZHANG
Journal of Zhejiang University. Science. B 2023;24(5):406-417
The aim of this study was to investigate the role of selenoprotein M (SelM) in endoplasmic reticulum stress and apoptosis in nickel-exposed mouse hearts and to explore the detoxifying effects of melatonin. At 21 d after intraperitoneal injection of nickel chloride (NiCl2) and/or melatonin into male wild-type (WT) and SelM knockout (KO) C57BL/6J mice, NiCl2 was found to induce changes in the microstructure and ultrastructure of the hearts of both WT and SelM KO mice, which were caused by oxidative stress, endoplasmic reticulum stress, and apoptosis, as evidenced by decreases in malondialdehyde (MDA) content and total antioxidant capacity (T-AOC) activity. Changes in the messenger RNA (mRNA) and protein expression of genes related to endoplasmic reticulum stress (activating transcription factor 4 (ATF4), inositol-requiring protein 1 (IRE1), c-Jun N-terminal kinase (JNK), and C/EBP homologous protein (CHOP)) and apoptosis (B-cell lymphoma-2 (Bcl-2), Bcl-2-associated X protein (Bax), Caspase-3, Caspase-9, and Caspase-12) were also observed. Notably, the observed damage was worse in SelM KO mice. Furthermore, melatonin alleviated the heart injury caused by NiCl2 in WT mice but could not exert a good protective effect in the heart of SelM KO mice. Overall, the findings suggested that the antioxidant capacity of SelM, as well as its modulation of endoplasmic reticulum stress and apoptosis, plays important roles in nickel-induced heart injury.
Animals
;
Male
;
Mice
;
Antioxidants/pharmacology*
;
Apoptosis
;
Endoplasmic Reticulum Stress
;
Melatonin/pharmacology*
;
Mice, Inbred C57BL
;
Nickel/adverse effects*
;
Selenoproteins/genetics*
;
Heart/drug effects*
6.Schisandra chinensis Oil Attenuates Aristolochic Acid I-Induced Nephrotoxicity in vivo and in vitro.
Yan YANG ; Fei-Lin GE ; Xiao-Yan ZHAN ; Wen-Qing MU ; Zhi-Yong LI ; Li LIN ; Zi-Ying WEI ; Zhao-Fang BAI ; Qin SUN ; Xiao-He XIAO
Chinese journal of integrative medicine 2022;28(7):603-611
OBJECTIVE:
To investigate the protective effects of Schisandra chinensis oil (SCEO) against aristolochic acid I (AA I)-induced nephrotoxicity in vivo and in vitro and elucidate the underlying mechanism.
METHODS:
C57BL/6 mice were randomly divided into 5 groups according to a random number table, including control group, AA I group, and AA I +SCEO (0.25, 0.5 and 1 g/kg) groups (n=5 per group). Pretreatment with SCEO was done for 2 days by oral administration, while the control and AA I groups were treated with sodium carboxymethyl cellulose. Mice of all groups except for the control group were injected intraperitoneally with AA I (5 mg/kg) from day 3 until day 7. Histopathological examination and apoptosis of kidney tissue were observed by hematoxylin and eosin and TdT-mediated dUTP nick-end labeling (TUNEL) staining, respectively. The levels of serum alanine aminotransferase (ALT), aspartate aminotransferase (AST), blood urea nitrogen (BUN), and serum creatinine (SCr), as well as renal malondialdehyde (MDA), glutathione, r-glutamyl cysteingl+glycine (GSH), and superoxide dismutase (SOD) were analyzed using enzyme-linked immunosorbent assay (ELISA). Expressions of hepatic cytochrome P450 1A1 (CYP1A1), CYP1A2, and nad(p)hquinonedehydrogenase1 (NQO1) were analyzed using ELISA, quantitative real-time polymerase chain reaction (qPCR) and Western blot, respectively. In vitro, SCEO (40 µ g/mL) was added 12 h before treatment with AA I (40 µ mol/mL for 48 h) in human renal proximal tubule cell line (HK-2), then apoptosis and reactive oxygen species (ROS) were analyzed by flow cytometry.
RESULTS:
SCEO 0.5 and 1 g/kg ameliorated histopathological changes and TUNEL+ staining in the kidney tissues of mice with AA I-induced nephrotoxicity, and reduced serum levels of ALT, AST, BUN and SCr (P<0.01 or P<0.05). SCEO 0.5 and 1 g/kg alleviated the ROS generation in kidney, containing MDA, GSH and SOD (P<0.01 or P<0.05). SCEO 1 g/kg increased the expressions of CYP1A1 and CYP1A2 and decreased NQO1 level in the liver tissues (P<0.01 or P<0.05). Besides, in vitro studies also demonstrated that SCEO 40 µ g/mL inhibited apoptosis and ROS generation (P<0.05 or P<0.01).
CONCLUSIONS
SCEO can alleviate AA I-induced kidney damage both in vivo and in vitro. The protective mechanism may be closely related to the regulation of metabolic enzymes, thereby inhibiting apoptosis and ROS production.
Animals
;
Apoptosis
;
Aristolochic Acids/toxicity*
;
Cytochrome P-450 CYP1A1/metabolism*
;
Cytochrome P-450 CYP1A2/metabolism*
;
Glutathione/metabolism*
;
Kidney/drug effects*
;
Kidney Diseases/drug therapy*
;
Mice
;
Mice, Inbred C57BL
;
Oxidative Stress
;
Plant Oils/therapeutic use*
;
Protective Agents/therapeutic use*
;
Reactive Oxygen Species/metabolism*
;
Schisandra
;
Superoxide Dismutase/metabolism*
7.Role of DNMT3a in Hydroquinone-Induced Hematopoietic Stem Cell Toxicity.
Kun WU ; Bo NIE ; Jin-Rong YANG ; Zheng-Xin HE ; Shen-Ju CHENG ; Yan-Hong LI ; Zhen JIN ; Ming-Xia SHI
Journal of Experimental Hematology 2022;30(2):607-612
OBJECTIVE:
To investigate the regulatory effect and mechanism of DNA methyltransferase 3A (DNMT3a) in hydroquinone-induced hematopoietic stem cell toxicity.
METHODS:
Cells (HSPC-1) were divided into 4 groups, that is A: normal HSPC-1; B: HQ-intervented HSPC-1; C: group B + pcDNA3 empty vector; D: group B + pcDNA3- DNMT3a. RT-qPCR and Western blot were used to detect the expression levels of DNMT3a and PARP-1 mRNA and protein, respectively. Cell morphology was observe; Cell viability and apoptosis rate of HSPC-1 were detected by MTT and flow cytometry, respectively.
RESULTS:
Compared with group A, the expression levels of DNMT3a mRNA and protein in HSPC-1 of group B were decreased, while PARP-1 mRNA and protein were increased (P<0.05); there was no significant difference in the above indexes between group C and group B; compared with group B, the expression levels of DNMT3a mRNA and protein showed increased, while PARP-1 mRNA and protein were decreased significantly in cells of group D transfected with DNMT3a (P<0.05). Cells in each group were transfected with DNMT3a and cultured for 24 h, HSPC-1 in group A showed high density growth and mononuclear fusion growth, while the number of HSPC-1 in group B and C decreased and grew slowly. Compared with group B and C, the cell growth rate of group D was accelerated. The MTT analysis showed that cell viability of HSPC-1 in group B were lower than that of group A at 24 h, 48 h and 72 h (P<0.05); after transfected with DNMT3a, the cell viability of HSPC-1 in group D were higher than that of group B at 24 h, 48 h and 72 h (P<0.05). The apoptosis rate of cells in group B was significantly higher than that of group A (P<0.001), while the apoptosis rate in group D was lower than that of group B (P<0.001).
CONCLUSION
DNMT3a may be involved in the damage of hematopoietic stem cells induced by hydroquinone, which may be related to the regulation of PARP-1 activity by hydroquinone-inhibited DNMT3a.
Apoptosis
;
Cell Proliferation
;
DNA Methyltransferase 3A
;
Hematopoietic Stem Cells/drug effects*
;
Humans
;
Hydroquinones/toxicity*
;
Poly (ADP-Ribose) Polymerase-1
;
RNA, Messenger/metabolism*
8.Human Serum-derived Extracellular Vesicles Protect A549 from PM
Qiu Lian ZHOU ; Yu Zheng BAI ; Juan GAO ; Yi DUAN ; Yi Cheng LYU ; Long Fei GUAN ; Kenneth ELKIN ; Yu Ling XIE ; Zheng JIAO ; Hong Yun WANG
Biomedical and Environmental Sciences 2021;34(1):40-49
Objective:
Epidemiological studies reveal that exposure to fine particulate matter (aerodynamic diameter ≤ 2.5 μm, PM
Methods:
EVs were isolated from the serum of healthy subjects, quantified
Results:
PM
Conclusions
EVs treatment promotes cell survival and attenuates PM
A549 Cells
;
Air Pollutants/toxicity*
;
Apoptosis/drug effects*
;
Cell Survival/drug effects*
;
Extracellular Vesicles
;
Humans
;
Male
;
Middle Aged
;
Particulate Matter/toxicity*
;
Protective Agents/pharmacology*
;
Proto-Oncogene Proteins c-akt/metabolism*
;
Serum
9.The Effect and Mechanism of Novel Telomerase Inhibitor Nilo 22 on Leukemia Cells.
Jing-Jing YIN ; Qian TANG ; Jia-Li GU ; Ya-Fang LI ; Hui-Er GAO ; Mei HE ; Ming YANG ; Wen-Shan ZHANG ; Hui XU ; Chao-Qun WANG ; Ying-Hui LI ; Cui-Gai BAI ; Ying-Dai GAO
Journal of Experimental Hematology 2021;29(4):1056-1064
OBJECTIVE:
To investigate the cytotoxic effect and its mechanism of the micromolecule compound on the leukemia cells.
METHODS:
The cytotoxic effects of 28 Nilotinib derivatives on K562, KA, KG, HA and 32D cell lines were detected by MTT assays, and the compound Nilo 22 was screen out. Cell apoptosis and cell cycle on leukemia cells were detected by flow cytometry. The effect of compound screened out on leukemogenesis potential of MLL-AF9 leukemia mice GFP
RESULTS:
Nilo 22 serves as the most outstanding candidate out of 28 Nilotinib derivatives, which impairs leukemia cell lines, but spares normal hematopoietic cell line. Comparing with Nilotinib, Nilo 22 could induce the apoptosis of GFP
CONCLUSION
Nilo 22 shows a significant cytotoxic effect on mice and human leukemia cells, especially for drug resistance cells. Nilo 22 is a promising anti-leukemia agent to solve the common clinical problems of drug resistance and relapse of leukemia.
Animals
;
Apoptosis/drug effects*
;
Cell Cycle/drug effects*
;
Cell Line, Tumor
;
Humans
;
Leukemia
;
Mice
;
Myeloid-Lymphoid Leukemia Protein/genetics*
;
Telomerase/metabolism*
;
Telomere/metabolism*

Result Analysis
Print
Save
E-mail