1.Chemerin promotes proliferation and migration of ovarian cancer cells by upregulating expression of PD-L1.
Chenxi GAO ; Jinming SHI ; Jingxin ZHANG ; Yin LI ; Yi ZHANG
Journal of Zhejiang University. Science. B 2022;23(2):164-170
Ovarian cancer is the third-most-common malignant reproductive tumor in women. According to the American Cancer Society, it has the highest mortality rate of gynecological tumors. The five-year survival rate was only 29% during the period from 1975 to 2008 (Reid et al., 2017). In recent decades, the five-year survival rate of ovarian cancer has remained around 30% despite continuous improvements in surgery, chemotherapy, radiotherapy, and other therapeutic methods. However, because of the particularity of the volume and location of ovarian tissue, the early symptoms of ovarian cancer are hidden, and there is a lack of highly sensitive and specific screening methods. Most patients have advanced metastasis, including abdominal metastasis, when they are diagnosed (Reid et al., 2017). Therefore, exploring the mechanism of ovarian cancer metastasis and finding early preventive measures are key to improving the survival rate and reducing mortality caused by ovarian cancer.
B7-H1 Antigen/biosynthesis*
;
Cell Proliferation/drug effects*
;
Chemokines/biosynthesis*
;
Female
;
Humans
;
Ovarian Neoplasms/pathology*
;
Survival Rate
;
Up-Regulation
2.Estradiol inhibits differentiation of mouse macrophage into a pro-inflammatory phenotype by upregulating the IRE1α-XBP1 signaling axis.
Ling Jian ZHUO ; Shuo Chen WANG ; Xing LIU ; Bao An CHEN ; Xiang LI
Journal of Southern Medical University 2022;42(3):432-437
OBJECTIVE:
To explore the mechanism by which estradiol modulates the immunophenotype of macrophages through the endoplasmic reticulum stress pathway.
METHODS:
Peritoneal macrophages isolated from C57 mice were cultured in the presence of 60 ng/mL interferon-γ (IFN-γ) followed by treatment with estradiol (1.0 nmol/L) alone, estradiol with estrogen receptor antagonist (Acolbifene, 4 nmol/L), estradiol with IRE1α inhibitor (4 μ 8 C), or estradiol with IRE1α agonist. After the treatments, the expression levels of MHC-Ⅱ, iNOS and endoplasmic reticulum stress marker proteins IRE1α, eIF2α and ATF6 in the macrophages were detected with Western blotting, and the mRNA levels of TGF-β, IL-6, IL-10 and TNF-α were detected with RT-PCR.
RESULTS:
Estrogen treatment of the macrophages significantly decreased the expressions of M1-related proteins MHC-Ⅱ (P=0.021) and iNOS (P < 0.001) and the mRNA expressions of TNF-α (P=0.003) and IL-6 (P=0.004), increased the mRNA expression of TGF-β (P=0.002) and IL-10 (P=0.008), and up-regulated the protein expressions of IRE1α (P < 0.001) and its downstream transcription factor XBP-1 (P < 0.001). Addition of the estrogen inhibitor obviously blocked the effect of estrogen. Compared with estrogen treatment alone, combined treatment of the macrophages with estrogen and the IRE1α inhibitor 4 μ 8 C significantly up-regulated the protein expressions of MHC-Ⅱ (P=0.002) and iNOS (P=0.003) and the mRNA expressions of TNF-α (P=0.003) and IL-6 (P=0.024), and obviously down-regulated the mRNA expression of TGF-β (P < 0.001) and IL-10 (P < 0.001); these changes were not observed in cells treated with estrogen and the IRE1α agonist.
CONCLUSION
Estrogen can inhibit the differentiation of murine macrophages into a pro-inflammatory phenotype by up-regulating the IRE1α-XBP-1 signaling axis, thereby producing an inhibitory effect on inflammatory response.
Animals
;
Cell Differentiation/drug effects*
;
Endoribonucleases/metabolism*
;
Estradiol/pharmacology*
;
Estrogens/metabolism*
;
Interleukin-10
;
Interleukin-6/metabolism*
;
Macrophages, Peritoneal/metabolism*
;
Mice
;
Phenotype
;
Protein Serine-Threonine Kinases/metabolism*
;
RNA, Messenger/metabolism*
;
Signal Transduction/drug effects*
;
Transforming Growth Factor beta/metabolism*
;
Tumor Necrosis Factor-alpha/metabolism*
;
Up-Regulation/drug effects*
;
X-Box Binding Protein 1/metabolism*
3.Protective effect of edaravone on balance of mitochondrial fusion and fission in MPP-treated PC12 cells.
Yang JIAO ; Yue ZHENG ; Cheng-Jie SONG
Acta Physiologica Sinica 2020;72(2):249-254
The aim of this study was to investigate the effect of edaravone (Eda) on the balance of mitochondrial fusion and fission in Parkinson's disease (PD) cell model. A cell model of PD was established by treating PC12 cells with 500 μmol/L 1-methyl-4-phenylpyridinium (MPP). Thiazole blue colorimetry (MTT) was used to detect the effect of different concentrations of Eda on the survival rate of PC12 cells exposed to MPP. The mitochondrial morphology was determined by laser confocal microscope. Western blot was used to measure the protein expression levels of mitochondrial fusion- and fission-related proteins, including OPA1, MFN2, DRP1 and Fis1. The results showed that pretreatment with different concentrations of Eda antagonized MPP-induced PC12 cell damage in a dose-dependent manner. The PC12 cells treated with MPP showed mitochondrial fragmentation, up-regulated DRP1 and Fis1 protein expression levels, and down-regulated MFN2 and OPA1 protein expression levels. Eda could reverse the above changes in the MPP-treated PC12 cells, but did not affect Fis1 protein expression. These results suggest that Eda has a protective effect on the mitochondrial fusion disruption induced by MPP in PC12 cells. The mechanism may be related to the up-regulation of OPA1/MFN2 and down-regulation of DRP1.
1-Methyl-4-phenylpyridinium
;
Animals
;
Dynamins
;
Edaravone
;
pharmacology
;
GTP Phosphohydrolases
;
Mitochondria
;
drug effects
;
Mitochondrial Dynamics
;
Mitochondrial Proteins
;
PC12 Cells
;
Parkinson Disease
;
Rats
;
Up-Regulation
4.Chinese medicine Buyang Huanwu decoction promotes neurogenesis and angiogenesis in ischemic stroke rats by upregulating miR-199a-5p expression.
Lujie ZHUGE ; Yan FANG ; Huaqian JIN ; Lin LI ; Yan YANG ; Xiaowei HU ; Lisheng CHU
Journal of Zhejiang University. Medical sciences 2020;49(6):687-696
OBJECTIVE:
To investigate the mechanism of Chinese medicine Buyang Huanwu decoction (BYHWD) promoting neurogenesis and angiogenesis in ischemic stroke rats.
METHODS:
Male SD rats were randomly divided into sham operation group, model group, BYHWD group, antagonist group and antagonist control group with 14 rats in each. Focal cerebral ischemia was induced by occlusion of the right middle cerebral artery for 90 min with intraluminal filament and reperfusion for 14 d in all groups except sham operation group. BYHWD (13 g/kg) was administrated by gastrogavage in BYHWD group, antagonist group and antagonist control group at 24 h after modeling respectively, and BrdU (50 mg/kg) was injected intraperitoneally in all groups once a day for 14 consecutive days. miR-199a-5p antagomir or NC (10 nmol) was injected into the lateral ventricle at d5 after ischemia in antagonist and antagonist control groups, respectively. The neurological deficits were evaluated by the modified neurological severity score (mNSS) and the corner test, and the infract volume was measured by toluidine blue staining. Neurogenesis and angiogenesis were detected by immunofluorescence double labeling method. The expression level of miR-199a-5p was tested by real-time RT-PCR, and the protein expressions of vascular endothelial growth factor (VEGF) and brain-derived neurotrophic factor (BDNF) were determined by Western blotting.
RESULTS:
BYHWD treatment significantly promoted the recovery of neurological function (
CONCLUSIONS
Buyang Huanwu decoction promotes neurogenesis and angiogenesis in rats with cerebral ischemia, which may be related to increased protein expression of VEGF and BDNF through upregulating miR-199a-5p.
Animals
;
Brain Ischemia/drug therapy*
;
Drugs, Chinese Herbal/therapeutic use*
;
Ischemic Stroke/drug therapy*
;
Male
;
MicroRNAs/genetics*
;
Neurogenesis/drug effects*
;
Rats
;
Rats, Sprague-Dawley
;
Up-Regulation/drug effects*
;
Vascular Endothelial Growth Factor A/genetics*
5.Carfilzomib inhibits the growth of lung adenocarcinoma via upregulation of Gadd45a expression.
Fang YANG ; Wang-Wang LIU ; Hui CHEN ; Jia ZHU ; Ai-Hua HUANG ; Fei ZHOU ; Yi GAN ; Yan-Hua ZHANG ; Li MA
Journal of Zhejiang University. Science. B 2020;21(1):64-76
Proteasome inhibitors have shown remarkable success in the treatment of hematologic neoplasm. There has been a lot of attention to applying these drugs for solid tumor treatment. Recent preclinical study has signified the effectiveness on cell proliferation inhibition in lung adenocarcinoma treated by carfilzomib (CFZ), a second generation proteasome inhibitor. However, no insight has been gained regarding the mechanism. In this study, we have systematically investigated the CFZ functions in cell proliferation and growth, cell cycle arrest, and apoptosis in lung adenocarcinoma cells. Flow cytometry experiments showed that CFZ significantly induced G2/M cell cycle arrest and apoptosis in lung adenocarcinoma. MTS and colony formation assays revealed that CFZ substantially inhibited survival of lung adenocarcinoma cells. All results were consistently correlated to the upregulation expression of Gadd45a, which is an important gene in modulating cell cycle arrest and apoptosis in response to physiologic and environmental stresses. Here, upregulation of Gadd45a expression was observed after CFZ treatment. Knocking down Gadd45a expression suppressed G2/M arrest and apoptosis in CFZ-treated cells, and reduced cytotoxicity of this drug. The protein expression analysis has further identified that the AKT/FOXO3a pathway is involved in Gadd45a upregulation after CFZ treatment. These findings unveil a novel mechanism of proteasome inhibitor in anti-solid tumor activity, and shed light on novel preferable therapeutic strategy for lung adenocarcinoma. We believe that Gadd45a expression can be a highly promising candidate predictor in evaluating the efficacy of proteasome inhibitors in solid tumor therapy.
Adenocarcinoma of Lung/pathology*
;
Apoptosis/drug effects*
;
Cell Cycle Checkpoints/drug effects*
;
Cell Cycle Proteins/genetics*
;
Cell Line, Tumor
;
Forkhead Box Protein O3/physiology*
;
Gene Expression Regulation, Neoplastic/drug effects*
;
Humans
;
Lung Neoplasms/pathology*
;
Oligopeptides/pharmacology*
;
Proto-Oncogene Proteins c-akt/physiology*
;
Up-Regulation
6.Anti-tumor and immune-modulating effect of decoction in mice bearing hepatoma H22 tumor.
Limei CHEN ; Tong JIN ; Chuntao NING ; Suli WANG ; Lijie WANG ; Jingming LIN
Journal of Southern Medical University 2019;39(2):241-248
OBJECTIVE:
To investigate the antitumor activity of decoction and study its liver and kidney toxicity and its effect on the immune system in a tumor-bearing mouse model.
METHODS:
Hepatoma H22 tumor-bearing mouse models were randomized into model group, cyclophosphamide (CTX) group, and low-, moderate-, and high-dose decoction groups (JW-L, JW-M, and JW-H groups, respectively). The antitumor activity of decoction was assessed by calculating the tumor inhibition rate and pathological observation of the tumor tissues. Immunohistochemistry was used to detect the expressions of Bax, Bcl-2, Bax/Bcl-2 and caspase-3 in the tumors. The liver and kidney toxicity of decoction was analyzed by evaluating the biochemical indicators of liver and kidney functions. The immune function of the tumor-bearing mice were assessed by calculating the immune organ index, testing peripheral blood routines, and detection of serum IL-2 and TNF-α levels using enzyme-linked immunosorbent assay.
RESULTS:
Compared with that in the model group, the tumor mass in CTX, JW-M and JW-H groups were all significantly reduced ( < 0.05) with cell rupture and necrosis in the tumors. Immunohistochemistry revealed obviously up-regulated expressions of Bax and caspase-3 and down- regulated expression of Bcl-2 protein with an increased Bax/Bcl-2 ratio in CTX, JW-M and JW-H groups. Treatment with decoction significantly reduced Cr, BUN, AST and ALT levels, improved the immune organ index, increased peripheral blood leukocytes, erythrocytes and hemoglobin levels, and up-regulated the levels of TNF-α and IL-2 in the tumor-bearing mice. These changes were especially significant in JW-H group when compared with the parameters in the model group ( < 0.01).
CONCLUSIONS
decoction has a strong anti-tumor activity and can improve the liver and kidney functions of tumor-bearing mice. Its anti-tumor effect may be attributed to the up-regulation of Bax, caspase-3, TNF-α and IL-2 levels and the down-regulation of Bcl-2 expression as well as the enhancement of the non-specific immune function.
Animals
;
Antineoplastic Agents, Phytogenic
;
pharmacology
;
Carcinoma, Hepatocellular
;
drug therapy
;
immunology
;
metabolism
;
pathology
;
Drugs, Chinese Herbal
;
pharmacology
;
Kidney
;
drug effects
;
Liver
;
drug effects
;
pathology
;
Liver Neoplasms
;
drug therapy
;
immunology
;
metabolism
;
pathology
;
Mice
;
Necrosis
;
Neoplasm Proteins
;
metabolism
;
Random Allocation
;
Up-Regulation
7.Rapamycin alleviates inflammation by up-regulating TGF-β/Smad signaling in a mouse model of autoimmune encephalomyelitis.
Zhenfei LI ; Lingling NIE ; Liping CHEN ; Yafei SUN ; Li GUO
Journal of Southern Medical University 2019;39(1):35-42
OBJECTIVE:
To evaluate the efficacy of rapmycin for treatment of experimental autoimmune encephalomyelitis (EAE) in mice and explore the underlying mechanism.
METHODS:
An EAE model was established in C57BL/6 mice. After immunization, the mice were divided into model group and rapamycin groups treated daily with low-dose (0.3 mg/kg) or high-dose (1 mg/kg) rapamycin. The clinical scores of the mice were observed using Knoz score, the infiltration of IL-17 cells in the central nervous system (CNS) was determined using immunohistochemistry; the differentiation of peripheral Treg cells was analyzed using flow cytometry, and the changes in the levels of cytokines were detected with ELISA; the changes in the expressions of p-Smad2 and p- smad3 were investigated using Western blotting.
RESULTS:
High-dose rapamycin significantly improved the neurological deficits scores of EAE mice. In high-dose rapamycin group, the scores in the onset stage, peak stage and remission stage were 0.14±0.38, 0.43±1.13 and 0.14±0.37, respectively, as compared with 1.14±0.69, 2.14±1.06 and 2.2±0.75 in the model group. The infiltration of inflammatory IL-17 cells was significantly lower in high-dose rapamycin group than in the model group (43±1.83 153.5±7.02). High-dose rapamycin obviously inhibited the production of IL-12, IFN-γ, IL-17 and IL-23 and induced the anti-inflammatory cytokines IL-10 and TGF-β. The percentage of Treg in CD4+ T cells was significantly higher in high- dose rapamycin group than in the model group (10.17 ± 0.68 3.52 ± 0.32). In the experiment, combined treatments of the lymphocytes isolated from the mice with rapamycin and TGF-β induced a significant increase in the number of Treg cells (13.66±1.89) compared with the treatment with rapamycin (6.23±0.80) or TGF-β (4.87±0.85) alone. Rapamycin also obviously up-regulated the expression of p-Smad2 and p-Smad3 in the lymphocytes.
CONCLUSIONS
Rapamycin can promote the differentiation of Treg cells by up-regulating the expression of p-Smad2 and p-smad3 to improve neurological deficits in mice with EAE.
Animals
;
Anti-Inflammatory Agents
;
administration & dosage
;
therapeutic use
;
Cell Differentiation
;
drug effects
;
Encephalomyelitis, Autoimmune, Experimental
;
drug therapy
;
metabolism
;
Interferon-gamma
;
metabolism
;
Interleukins
;
metabolism
;
Lymphocytes
;
cytology
;
Mice
;
Mice, Inbred C57BL
;
Sirolimus
;
administration & dosage
;
therapeutic use
;
Smad Proteins
;
metabolism
;
T-Lymphocytes, Regulatory
;
cytology
;
drug effects
;
Transforming Growth Factor beta
;
metabolism
;
Up-Regulation
8.Overexpression of Dlx2 enhances osteogenic differentiation of BMSCs and MC3T3-E1 cells via direct upregulation of Osteocalcin and Alp.
Jianfei ZHANG ; Wenbin ZHANG ; Jiewen DAI ; Xudong WANG ; Steve Guofang SHEN
International Journal of Oral Science 2019;11(2):12-12
Genetic studies have revealed a critical role of Distal-homeobox (Dlx) genes in bone formation, and our previous study showed that Dlx2 overexpressing in neural crest cells leads to profound abnormalities of the craniofacial tissues. The aim of this study was to investigate the role and the underlying molecular mechanisms of Dlx2 in osteogenic differentiation of mouse bone marrow stromal cells (BMSCs) and pre-osteoblast MC3T3-E1 cells. Initially, we observed upregulation of Dlx2 during the early osteogenesis in BMSCs and MC3T3-E1 cells. Moreover, Dlx2 overexpression enhanced alkaline phosphatase (ALP) activity and extracellular matrix mineralization in BMSCs and MC3T3-E1 cell line. In addition, micro-CT of implanted tissues in nude mice confirmed that Dlx2 overexpression in BMSCs promoted bone formation in vivo. Unexpectedly, Dlx2 overexpression had little impact on the expression level of the pivotal osteogenic transcription factors Runx2, Dlx5, Msx2, and Osterix, but led to upregulation of Alp and Osteocalcin (OCN), both of which play critical roles in promoting osteoblast maturation. Importantly, luciferase analysis showed that Dlx2 overexpression stimulated both OCN and Alp promoter activity. Through chromatin-immunoprecipitation assay and site-directed mutagenesis analysis, we provide molecular evidence that Dlx2 transactivates OCN and Alp expression by directly binding to the Dlx2-response cis-acting elements in the promoter of the two genes. Based on these findings, we demonstrate that Dlx2 overexpression enhances osteogenic differentiation in vitro and accelerates bone formation in vivo via direct upregulation of the OCN and Alp gene, suggesting that Dlx2 plays a crucial role in osteogenic differentiation and bone formation.
Animals
;
Cell Differentiation
;
physiology
;
Core Binding Factor Alpha 1 Subunit
;
Homeodomain Proteins
;
metabolism
;
Mesenchymal Stem Cells
;
metabolism
;
Mice
;
Mice, Nude
;
Osteoblasts
;
metabolism
;
Osteocalcin
;
drug effects
;
Osteogenesis
;
physiology
;
Transcription Factors
;
metabolism
;
Up-Regulation
9.Single-cell Analysis of CAR-T Cell Activation Reveals A Mixed T1/T2 Response Independent of Differentiation.
Iva XHANGOLLI ; Burak DURA ; GeeHee LEE ; Dongjoo KIM ; Yang XIAO ; Rong FAN
Genomics, Proteomics & Bioinformatics 2019;17(2):129-139
The activation mechanism of chimeric antigen receptor (CAR)-engineered T cells may differ substantially from T cells carrying native T cell receptor, but this difference remains poorly understood. We present the first comprehensive portrait of single-cell level transcriptional and cytokine signatures of anti-CD19/4-1BB/CD28/CD3ζ CAR-T cells upon antigen-specific stimulation. Both CD4 helper T (T) cells and CD8 cytotoxic CAR-T cells are equally effective in directly killing target tumor cells and their cytotoxic activity is associated with the elevation of a range of T1 and T2 signature cytokines, e.g., interferon γ, tumor necrotic factor α, interleukin 5 (IL5), and IL13, as confirmed by the expression of master transcription factor genes TBX21 and GATA3. However, rather than conforming to stringent T1 or T2 subtypes, single-cell analysis reveals that the predominant response is a highly mixed T1/T2 function in the same cell. The regulatory T cell activity, although observed in a small fraction of activated cells, emerges from this hybrid T1/T2 population. Granulocyte-macrophage colony stimulating factor (GM-CSF) is produced from the majority of cells regardless of the polarization states, further contrasting CAR-T to classic T cells. Surprisingly, the cytokine response is minimally associated with differentiation status, although all major differentiation subsets such as naïve, central memory, effector memory, and effector are detected. All these suggest that the activation of CAR-engineered T cells is a canonical process that leads to a highly mixed response combining both type 1 and type 2 cytokines together with GM-CSF, supporting the notion that polyfunctional CAR-T cells correlate with objective response of patients in clinical trials. This work provides new insights into the mechanism of CAR activation and implies the necessity for cellular function assays to characterize the quality of CAR-T infusion products and monitor therapeutic responses in patients.
Antigens
;
metabolism
;
CTLA-4 Antigen
;
metabolism
;
Cell Differentiation
;
drug effects
;
Cell Line
;
Cytokines
;
metabolism
;
Cytotoxicity, Immunologic
;
drug effects
;
Granulocyte-Macrophage Colony-Stimulating Factor
;
pharmacology
;
Humans
;
Lymphocyte Activation
;
drug effects
;
immunology
;
Lymphocyte Subsets
;
drug effects
;
metabolism
;
Phenotype
;
Proteomics
;
Receptors, Chimeric Antigen
;
metabolism
;
Single-Cell Analysis
;
methods
;
T-Lymphocytes, Regulatory
;
drug effects
;
metabolism
;
Th1 Cells
;
cytology
;
drug effects
;
Th2 Cells
;
cytology
;
drug effects
;
Transcription, Genetic
;
drug effects
;
Up-Regulation
;
drug effects
10.Anti-Hypertensive Action of Fenofibrate via UCP2 Upregulation Mediated by PPAR Activation in Baroreflex Afferent Pathway.
Jian GUAN ; Miao ZHAO ; Chao HE ; Xue LI ; Ying LI ; Jie SUN ; Wei WANG ; Ya-Li CUI ; Qing ZHANG ; Bai-Yan LI ; Guo-Fen QIAO
Neuroscience Bulletin 2019;35(1):15-24
Fenofibrate, an agonist for peroxisome proliferator-activated receptor alpha (PPAR-α), lowers blood pressure, but whether this action is mediated via baroreflex afferents has not been elucidated. In this study, the distribution of PPAR-α and PPAR-γ was assessed in the nodose ganglion (NG) and the nucleus of the solitary tract (NTS). Hypertension induced by drinking high fructose (HFD) was reduced, along with complete restoration of impaired baroreceptor sensitivity, by chronic treatment with fenofibrate. The molecular data also showed that both PPAR-α and PPAR-γ were dramatically up-regulated in the NG and NTS of the HFD group. Expression of the downstream signaling molecule of PPAR-α, the mitochondrial uncoupling protein 2 (UCP2), was up-regulated in the baroreflex afferent pathway under similar experimental conditions, along with amelioration of reduced superoxide dismutase activity and increased superoxide in HFD rats. These results suggest that chronic treatment with fenofibrate plays a crucial role in the neural control of blood pressure by improving baroreflex afferent function due at least partially to PPAR-mediated up-regulation of UCP2 expression and reduction of oxidative stress.
Afferent Pathways
;
drug effects
;
Animals
;
Antihypertensive Agents
;
pharmacology
;
Baroreflex
;
drug effects
;
Blood Pressure
;
drug effects
;
Fenofibrate
;
pharmacology
;
Male
;
Oxidative Stress
;
drug effects
;
PPAR gamma
;
drug effects
;
metabolism
;
Rats, Sprague-Dawley
;
Signal Transduction
;
drug effects
;
Transcriptional Activation
;
drug effects
;
Uncoupling Protein 2
;
drug effects
;
metabolism
;
Up-Regulation

Result Analysis
Print
Save
E-mail