1.Lysosomal membrane protein Sidt2 knockout induces apoptosis of human hepatocytes in vitro independent of the autophagy-lysosomal pathway.
Jiating XU ; Mengya GENG ; Haijun LIU ; Wenjun PEI ; Jing GU ; Mengxiang QI ; Yao ZHANG ; Kun LÜ ; Yingying SONG ; Miaomiao LIU ; Xin HU ; Cui YU ; Chunling HE ; Lizhuo WANG ; Jialin GAO
Journal of Southern Medical University 2023;43(4):637-643
OBJECTIVE:
To explore the regulatory mechanism of human hepatocyte apoptosis induced by lysosomal membrane protein Sidt2 knockout.
METHODS:
The Sidt2 knockout (Sidt2-/-) cell model was constructed in human hepatocyte HL7702 cells using Crispr-Cas9 technology.The protein levels of Sidt2 and key autophagy proteins LC3-II/I and P62 in the cell model were detected using Western blotting, and the formation of autophagosomes was observed with MDC staining.EdU incorporation assay and flow cytometry were performed to observe the effect of Sidt2 knockout on cell proliferation and apoptosis.The effect of chloroquine at the saturating concentration on autophagic flux, proliferation and apoptosis of Sidt2 knockout cells were observed.
RESULTS:
Sidt2-/- HL7702 cells were successfully constructed.Sidt2 knockout significantly inhibited the proliferation and increased apoptosis of the cells, causing also increased protein expressions of LC3-II/I and P62(P < 0.05) and increased number of autophagosomes.Autophagy of the cells reached a saturated state following treatment with 50 μmol/L chloroquine, and at this concentration, chloroquine significantly increased the expressions of LC3B and P62 in Sidt2-/- HL7702 cells.
CONCLUSION
Sidt2 gene knockout causes dysregulation of the autophagy pathway and induces apoptosis of HL7702 cells, and the latter effect is not mediated by inhibiting the autophagy-lysosomal pathway.
Humans
;
Lysosome-Associated Membrane Glycoproteins/metabolism*
;
Autophagy
;
Apoptosis
;
Hepatocytes
;
Lysosomes/metabolism*
;
Chloroquine/pharmacology*
;
Nucleotide Transport Proteins/metabolism*
2.BGB-A445, a novel non-ligand-blocking agonistic anti-OX40 antibody, exhibits superior immune activation and antitumor effects in preclinical models.
Beibei JIANG ; Tong ZHANG ; Minjuan DENG ; Wei JIN ; Yuan HONG ; Xiaotong CHEN ; Xin CHEN ; Jing WANG ; Hongjia HOU ; Yajuan GAO ; Wenfeng GONG ; Xing WANG ; Haiying LI ; Xiaosui ZHOU ; Yingcai FENG ; Bo ZHANG ; Bin JIANG ; Xueping LU ; Lijie ZHANG ; Yang LI ; Weiwei SONG ; Hanzi SUN ; Zuobai WANG ; Xiaomin SONG ; Zhirong SHEN ; Xuesong LIU ; Kang LI ; Lai WANG ; Ye LIU
Frontiers of Medicine 2023;17(6):1170-1185
OX40 is a costimulatory receptor that is expressed primarily on activated CD4+, CD8+, and regulatory T cells. The ligation of OX40 to its sole ligand OX40L potentiates T cell expansion, differentiation, and activation and also promotes dendritic cells to mature to enhance their cytokine production. Therefore, the use of agonistic anti-OX40 antibodies for cancer immunotherapy has gained great interest. However, most of the agonistic anti-OX40 antibodies in the clinic are OX40L-competitive and show limited efficacy. Here, we discovered that BGB-A445, a non-ligand-competitive agonistic anti-OX40 antibody currently under clinical investigation, induced optimal T cell activation without impairing dendritic cell function. In addition, BGB-A445 dose-dependently and significantly depleted regulatory T cells in vitro and in vivo via antibody-dependent cellular cytotoxicity. In the MC38 syngeneic model established in humanized OX40 knock-in mice, BGB-A445 demonstrated robust and dose-dependent antitumor efficacy, whereas the ligand-competitive anti-OX40 antibody showed antitumor efficacy characterized by a hook effect. Furthermore, BGB-A445 demonstrated a strong combination antitumor effect with an anti-PD-1 antibody. Taken together, our findings show that BGB-A445, which does not block OX40-OX40L interaction in contrast to clinical-stage anti-OX40 antibodies, shows superior immune-stimulating effects and antitumor efficacy and thus warrants further clinical investigation.
Mice
;
Animals
;
Receptors, Tumor Necrosis Factor/physiology*
;
Receptors, OX40
;
Membrane Glycoproteins
;
Ligands
;
Antibodies, Monoclonal/pharmacology*
;
Antineoplastic Agents/pharmacology*
3.Melatonin Augments the Effects of Fluoxetine on Depression-Like Behavior and Hippocampal BDNF-TrkB Signaling.
Kun LI ; Si SHEN ; Yu-Tian JI ; Xu-Yun LI ; Li-San ZHANG ; Xiao-Dong WANG
Neuroscience Bulletin 2018;34(2):303-311
Depression is a debilitating psychiatric disorder with a huge socioeconomic burden, and its treatment relies on antidepressants including selective serotonin reuptake inhibitors (SSRIs). Recently, the melatonergic system that is closely associated with the serotonergic system has been implicated in the pathophysiology and treatment of depression. However, it remains unknown whether combined treatment with SSRI and melatonin has synergistic antidepressant effects. In this study, we applied a sub-chronic restraint stress paradigm, and evaluated the potential antidepressant effects of combined fluoxetine and melatonin in adult male mice. Sub-chronic restraint stress (6 h/day for 10 days) induced depression-like behavior as shown by deteriorated fur state, increased latency to groom in the splash test, and increased immobility time in the forced-swim test. Repeated administration of either fluoxetine or melatonin at 10 mg/kg during stress exposure failed to prevent depression-like phenotypes. However, combined treatment with fluoxetine and melatonin at the selected dose attenuated stress-induced behavioral abnormalities. Moreover, we found that the antidepressant effects of combined treatment were associated with the normalization of brain-derived neurotrophic factor (BDNF)-tropomyosin receptor kinase B (TrkB) signaling in the hippocampus, but not in the prefrontal cortex. Our findings suggest that combined fluoxetine and melatonin treatment exerts synergistic antidepressant effects possibly by restoring hippocampal BDNF-TrkB signaling.
Animals
;
Antidepressive Agents
;
pharmacology
;
Behavior, Animal
;
drug effects
;
Brain-Derived Neurotrophic Factor
;
drug effects
;
metabolism
;
Depression
;
Drug Synergism
;
Drug Therapy, Combination
;
Fluoxetine
;
pharmacology
;
Hippocampus
;
drug effects
;
metabolism
;
Male
;
Melatonin
;
pharmacology
;
Membrane Glycoproteins
;
drug effects
;
metabolism
;
Mice, Inbred C57BL
;
Protein-Tyrosine Kinases
;
drug effects
;
metabolism
;
Restraint, Physical
;
Signal Transduction
;
drug effects
4.Effects of paeonol on the function of bone marrow-derived macrophage from Porphyromonas gingivalis-induced mice.
West China Journal of Stomatology 2017;35(2):139-144
OBJECTIVEThis work aims to examine the effects of paeonol treatment on the ability of bone marrow-derived macrophage (BMM) to excrete inflammatory factors and to differentiate into osteoclasts upon induction with Porphyromonas gingivalis (P. gingivalis). This work also aims to investigate the underlying mechanisms of these abilities.
METHODSBMM culture was treated with different paeonol concentrations at for 1 h and then stimulated with P. gingivalis for 24 h before programmed death-ligand 1 (PD-L1) was quantified with flow cytometry. Tumor necrosis factor-α (TNF-α), interleukin (IL)-1β, and IL-6 were detected by enzyme-linked immunosorbent assay (ELISA). The BMM culture was treated with the receptor activator for nuclear factor-κB ligand (RANKL) and macrophage colony-stimulating factor (M-CSF), and then with paeonol for 1 h prior to induction with P. gingivalis. Then, osteoclast formation was assessed using tartrate resistant acid phosphatase (TRAP) staining. The osteoclast-related proteins TRAP and receptor activator of nuclear factor-κB (RANK) were quantified by Western blotting.
RESULTSPaeonol was nontoxic to BMM within a range of 10-50 μmol·L⁻¹. Flow cytometry showed that paeonol inhibited PD-L1 expression in P. gingivalis-induced BMM in a dose-dependent manner. ELISA indicated that paeonol dose-dependently inhibited the excretion of TNF-α, IL-1β, and IL-6 by P. gingivalis-induced BMM (P<0.01). TRAP staining revealed that paenol treatment inhibited the differentiation of P. gingivalis-induced BMM into osteoclasts. Western blot results suggested that paeonol decreased the expression of TRAP and RANK in BMM.
CONCLUSIONSPaeonol dose-dependently inhibited the excretion of the inflammatory factors TNF-α, IL-1β, and IL-6 by P. gingivalis-induced BMM in a dose-dependent manner. Moreover, paenol treatment prevented the differentiation of P. gingivalis-induced BMM differentiation into osteoclasts. .
Acetophenones ; pharmacology ; Acid Phosphatase ; Animals ; Carrier Proteins ; Cell Differentiation ; Interleukin-1beta ; Interleukin-6 ; Isoenzymes ; Macrophage Colony-Stimulating Factor ; Macrophages ; Membrane Glycoproteins ; Mice ; Osteoclasts ; Porphyromonas gingivalis ; RANK Ligand ; Receptor Activator of Nuclear Factor-kappa B ; Tumor Necrosis Factor-alpha
5.Salidroside protects PC12 cells from HO-induced apoptosis via suppressing NOX2-ROS-MAPKs signaling pathway.
Zhi-Lin QI ; Yin-Hua LIU ; Shi-Mei QI ; Lie-Feng LING ; Zun-Yong FENG ; Qiang LI
Journal of Southern Medical University 2016;37(2):178-183
OBJECTIVETo investigate the molecular mechanism by which salidroside protects PC12 cells from HO-induced apoptosis.
METHODSPC12 cells cultured in DMEM supplemented with 10% horse serum and 5% fetal bovine serum were pretreated with different doses of salidroside for 2 h and then stimulated with HOfor different lengths of time. The expression levels of PARP and caspase 3 and the phosphorylation of p38, ERK and JNK were determined with Western blotting. The cell nuclear morphology was observed after DAPI staining. The production of ROS was detected using a ROS detection kit, and the levels of gp91and p47in the membrane and cytoplasm were detected by membrane-cytoplasm separation experiment; the binding between gp91and p47was assayed by coimmunoprecipitation experiment.
RESULTSSalidroside dose-dependently suppressed cell apoptosis, lowered phosphorylation levels of p38, ERK and JNK, inhibited the production of ROS, reduced the binding between gp91and p47, and inhibited the activity of NOX2 in PC12 cells exposed to HO.
CONCLUSIONSalidroside protects PC12 cells from HO-induced apoptosis at least partly by suppressing NOX2-ROS-MAPKs signaling pathway.
Animals ; Apoptosis ; Caspase 3 ; metabolism ; Glucosides ; pharmacology ; Hydrogen Peroxide ; MAP Kinase Signaling System ; drug effects ; Membrane Glycoproteins ; metabolism ; NADPH Oxidase 2 ; NADPH Oxidases ; metabolism ; Neuroprotective Agents ; pharmacology ; PC12 Cells ; Phenols ; pharmacology ; Phosphorylation ; Rats ; Reactive Oxygen Species ; metabolism
6.Equol protects PC12 neuronal cells against hypoxia/reoxygenation injury in vitro by reducing reactive oxygen species production.
Wei YU ; Xiuling DENG ; Zhen MA ; Yan WANG
Journal of Southern Medical University 2016;36(1):1-7
OBJECTIVEBoth of gp91(phox) (an isoform of nicotinamide adenine dinucleotide phosphate-reduced oxidases) and Src (a non-receptor protein tyrosine kinase) play a prominent role in mediating hypoxia/reoxygenation injury of neurons. The present study was designed to investigate the neuroprotective effect of equol, a predominant active metabolite of daidzein, against hypoxia/reoxygenation injury in rat pheochromocytoma cell line (PC12) and explore the underlying mechanisms.
METHODSPC12 cells exposed to hypoxia/reoxygenation injury were examined for reactive oxygen species (ROS) using dihydroethidium and 2', 7'-dichlorofluorescein diacetate and analyzed for changes in lactate dehydrogenase (LDH) activity and malondialdehyde (MDA) content. The expression levels of gp91(phox) and phosphorylated Src-Tyr416 (p-Src) were measured using Western blotting.
RESULTSEquol dose-dependently restored the cell viability and decreased LDH activity and MDA content in culture medium of PC12 cells exposed to hypoxia/reoxygenation. Pretreatment of the cells with 10(-5) and 10(-6) mol/L equol inhibited hypoxia/reoxygenation-induced increase of ROS. PC12 cells treated with equol prior to hypoxia/reoxygenation injury showed significant enhancement of the protein levels of gp91(phox) and p-Src.
CONCLUSIONEquol confers neuroprotection against hypoxia/reoxygenation injury in PC12 cells by inhibiting the generation of ROS very likely as a result of down-regulation of gp91(phox) and inhibition of Src phosphorylation.
Animals ; Cell Hypoxia ; Cell Survival ; Down-Regulation ; Equol ; pharmacology ; L-Lactate Dehydrogenase ; metabolism ; Malondialdehyde ; metabolism ; Membrane Glycoproteins ; metabolism ; NADPH Oxidase 2 ; NADPH Oxidases ; metabolism ; Neurons ; drug effects ; metabolism ; Neuroprotective Agents ; pharmacology ; PC12 Cells ; Phosphorylation ; Rats ; Reactive Oxygen Species ; metabolism ; src-Family Kinases ; metabolism
7.Synaptic vesicle protein2A decreases in amygdaloid-kindling pharmcoresistant epileptic rats.
Jing SHI ; Feng ZHOU ; Li-kun WANG ; Guo-feng WU
Journal of Huazhong University of Science and Technology (Medical Sciences) 2015;35(5):716-722
Synaptic vesicle protein 2A (SV2A) involvement has been reported in the animal models of epilepsy and in human intractable epilepsy. The difference between pharmacosensitive epilepsy and pharmacoresistant epilepsy remains poorly understood. The present study aimed to observe the hippocampus SV2A protein expression in amygdale-kindling pharmacoresistant epileptic rats. The pharmacosensitive epileptic rats served as control. Amygdaloid-kindling model of epilepsy was established in 100 healthy adult male Sprague-Dawley rats. The kindled rat model of epilepsy was used to select pharmacoresistance by testing their seizure response to phenytoin and phenobarbital. The selected pharmacoresistant rats were assigned to a pharmacoresistant epileptic group (PRE group). Another 12 pharmacosensitive epileptic rats (PSE group) served as control. Immunohistochemistry, real-time PCR and Western blotting were used to determine SV2A expression in the hippocampus tissue samples from both the PRE and the PSE rats. Immunohistochemistry staining showed that SV2A was mainly accumulated in the cytoplasm of the neurons, as well as along their dendrites throughout all subfields of the hippocampus. Immunoreactive staining level of SV2A-positive cells was 0.483 ± 0.304 in the PRE group and 0.866 ± 0.090 in the PSE group (P < 0.05). Real-time PCR analysis demonstrated that 2(-ΔΔCt) value of SV2A mRNA was 0.30 ± 0.43 in the PRE group and 0.76 ± 0.18 in the PSE group (P < 0.05). Western blotting analysis obtained the similar findings (0.27 ± 0.21 versus 1.12 ± 0.21, P < 0.05). PRE rats displayed a significant decrease of SV2A in the brain. SV2A may be associated with the pathogenesis of intractable epilepsy of the amygdaloid-kindling rats.
Amygdala
;
drug effects
;
metabolism
;
physiopathology
;
Animals
;
Anticonvulsants
;
pharmacology
;
Disease Models, Animal
;
Drug Resistance
;
Electric Stimulation
;
Epilepsy
;
drug therapy
;
genetics
;
metabolism
;
pathology
;
Gene Expression Regulation
;
Hippocampus
;
drug effects
;
metabolism
;
physiopathology
;
Kindling, Neurologic
;
drug effects
;
genetics
;
metabolism
;
pathology
;
Male
;
Membrane Glycoproteins
;
genetics
;
metabolism
;
Nerve Tissue Proteins
;
genetics
;
metabolism
;
Phenobarbital
;
pharmacology
;
Phenytoin
;
pharmacology
;
RNA, Messenger
;
genetics
;
metabolism
;
Rats
;
Rats, Sprague-Dawley
;
Synaptic Transmission
;
Synaptic Vesicles
;
drug effects
;
metabolism
;
pathology
8.CYP2C8-derived epoxyeicosatrienoic acids decrease oxidative stress-induced endothelial apoptosis in development of atherosclerosis: Role of Nrf2 activation.
Wan-jun LIU ; Tao WANG ; Bei WANG ; Xin-tian LIU ; Xing-wei HE ; Yu-jian LIU ; Zhu-xi LI ; Rong TAN ; He-song ZENG
Journal of Huazhong University of Science and Technology (Medical Sciences) 2015;35(5):640-645
The aim of the present study is to investigate how cytochrome P450 enzymes (CYP) 2C8-derived epoxyeicosatrienoic acids (EETs) regulate the nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway and protect against oxidative stress-induced endothelial injuries in the development and progression of atherosclerosis. In this study, cultured human umbilical vein endothelial cells (HUVECs) were transfected with CYP2C8 or pretreated with exogenous EETs (1 μmol/L) before TNF-α (20 ng/mL) stimulation. Apoptosis and intracellular ROS production were determined by flow cytometry. The expression levels of ROS-associated NAD(P)H subunits gp91 and p47, the anti-oxidative enzyme catalase (CAT), Nrf2, heme oxygenase-1 (HO-1) and endothelial nitric oxide synthase (eNOS) were detected by Western blotting. The results showed that CYP2C8-derived EETs decreased apoptosis of HUVECs treated with TNF-α. Pretreatment with 11, 12-EET also significantly blocked TNF-α-induced ROS production. In addition, 11, 12-EET decreased oxidative stress-induced apoptosis. Furthermore, the ability of 11, 12-EET to protect cells against TNF-α-induced apoptosis via oxidative stress was abrogated by transient transfection with Nrf2-specific small interfering RNA (siRNA). In conclusion, CYP2C8-derived EETs prevented TNF-α-induced HUVECs apoptosis via inhibition of oxidative stress associated with the Nrf2 signaling.
8,11,14-Eicosatrienoic Acid
;
analogs & derivatives
;
metabolism
;
pharmacology
;
Adaptor Proteins, Signal Transducing
;
genetics
;
metabolism
;
Apoptosis
;
drug effects
;
Aryl Hydrocarbon Hydroxylases
;
genetics
;
metabolism
;
Atherosclerosis
;
genetics
;
metabolism
;
pathology
;
Catalase
;
genetics
;
metabolism
;
Cytochrome P-450 CYP2C8
;
genetics
;
metabolism
;
Gene Expression Regulation
;
Heme Oxygenase-1
;
genetics
;
metabolism
;
Human Umbilical Vein Endothelial Cells
;
cytology
;
drug effects
;
metabolism
;
Humans
;
Membrane Glycoproteins
;
genetics
;
metabolism
;
Models, Biological
;
NADPH Oxidase 2
;
NADPH Oxidases
;
genetics
;
metabolism
;
NF-E2-Related Factor 2
;
antagonists & inhibitors
;
genetics
;
metabolism
;
Nitric Oxide Synthase Type III
;
genetics
;
metabolism
;
RNA, Small Interfering
;
genetics
;
metabolism
;
Reactive Oxygen Species
;
antagonists & inhibitors
;
metabolism
;
Signal Transduction
;
Tumor Necrosis Factor-alpha
;
metabolism
;
pharmacology
9.A experiment research of beryllium oxide induced oxidative lung injury and the protective effects of LBP in rats.
Zhihong LIU ; Qingfeng ZHANG ; Yao WANG ; Conghui WEI ; Qing YAN ; Aihong GONG ; Xiong GUO ; E-mail: GUO@MAILI.XJTU.EDU.CN.
Chinese Journal of Industrial Hygiene and Occupational Diseases 2015;33(7):512-516
OBJECTIVETo explore beryllium oxide induced oxidative lung injury and the protective effects of LBP.
METHODSIntoxication of animals were induced by once intratracheal injection and LBP intervention by intragastric administration. The content of HIF-1, VEGF and HO-1 of lung tissues were measured by kits. The pathological changes of lung tissue were showed by pathological section. The changes of lung ultrastructure were observed by electron microscope.
RESULTSPathological changes of the lung tissue in beryllium oxide exposure group rats were in line with the characteristics of beryllium disease in human. Compared with the control group, HO-1 was increased in beryllium oxide exposure 40 d group and low doses of LBP group, compared with the control group, HO-1 was increased in beryllium oxide exposure 80d group and LBP treatment groups (P < 0.05 or P < 0.01). Compared with the control group, HIF-1 was increased in beryllium oxide exposure 40 d group, LBP treatment groups, beryllium oxide exposure 60 d and 80 d groups (P < 0.05 or P < 0.01). Compared with the control group, VEGF was increased of all phases, especially in beryllium oxide exposure 40d and 80 groups, LBP treatment groups and beryllium oxide exposure 60 d (P < 0.05 or P < 0.01). The content of HO-1 of beryllium oxide exposure group was higher than the LBP treatment for 40d group but below LBP treatment for 80 d group (P < 0.05). The content of HIF1 of beryllium oxide exposure group was higher than high dose of LBP treatment for 60d group and LBP treatment for 80 d group (P < 0.01). The content of VEGF of beryllium oxide exposure group was higher than LBP treatment for 40 d group and high dose of LBP treatment for 60 d (P < 0.05 or P < 0.01).
CONCLUSIONSBeO can cause abnormal expression of related genes of lung tissue in rats, LBP has protective effects on BeO caused lung injury.
Acute Lung Injury ; chemically induced ; physiopathology ; Acute-Phase Proteins ; pharmacology ; Animals ; Beryllium ; toxicity ; Carrier Proteins ; pharmacology ; Heme Oxygenase (Decyclizing) ; metabolism ; Hypoxia-Inducible Factor 1, alpha Subunit ; metabolism ; Lung ; drug effects ; pathology ; Membrane Glycoproteins ; pharmacology ; Oxidative Stress ; Protective Agents ; pharmacology ; Rats ; Vascular Endothelial Growth Factor A ; metabolism
10.Effects of stanniocalcin-1 and hypoxia-inducible factor-1α on mitochondrial membrane potential stability in renal carcinoma cells.
Qing-tao YANG ; Jiang GU ; Yong-chun ZHANG ; Zhi-hui ZHU ; Yong-an YANG ; Nan WANG ; Qing-liang ZHU
Acta Academiae Medicinae Sinicae 2014;36(1):12-19
OBJECTIVETo explore the effects of stanniocalcin-1 (STC-1) and hypoxia-inducible factor-1α (HIF-1α) on the calcium and thus on the mitochondrial membrane potential (Δψm) in renal carcinoma cells.
METHODSWe successfully established the renal carcinoma cell models with high HIF-1α gene expression. After various concentrations of STC-1 solutions were added to the culture medium, the proliferation of cells, expressions of HIF-1α and STC-1, levels of Ca(2+), Δψm, and mPTP were detected by MTT, RT-PCR, ELISA, fluorescence spectrophotometry, and ultraviolet spectrophotometry, respectively.
RESULTSThe proliferation of renal carcinoma cells and Δψm were improved after HIF-1α gene transfection, STC-1 protein intervention, and STC-1 protein intervention after gene transfection. While the intracellular Ca(2+) level and mPTP were decreased significantly (P<0.05), all the changes were intensified with the gradual increase of STC-1. However, the increasing trend of cell proliferation gradually declined.
CONCLUSIONHIF-1α may participate in malignant proliferation of renal carcinoma cells by promoting STC-1 proliferation or down-regulating Ca(2+); however, such an effect may be gradually attenuated due to the inhibitory effect of STC-1 on HIF-1α.
Calcium ; metabolism ; Carcinoma, Renal Cell ; pathology ; Glycoproteins ; pharmacology ; Humans ; Hypoxia-Inducible Factor 1, alpha Subunit ; metabolism ; Membrane Potential, Mitochondrial ; drug effects ; Tumor Cells, Cultured

Result Analysis
Print
Save
E-mail