1.Observation of the effect of Yanghe Pingchuan granules on the homing of BMSCs in asthma based on FTO regulation of Notch1 pathway.
Kun WANG ; Haoxiang FANG ; Xiaomei CAO
Chinese Journal of Cellular and Molecular Immunology 2025;41(7):585-592
Objective To observe the effect of m6A methylation regulation on Notch1 pathway on the homing of BMSCs in asthma, and the intervention study of traditional Chinese medicine compound Yanghe Pingchuan Granules. Methods Rat bone mesenchymal stem cells(BMSC)and bronchial epithelial cells were cocultured. The extracted cells were divided into: bronchial epithelial cell group, asthma bronchial epithelial cell+mesenchymal stem cell co-culture group (co-culture group), co-culture cell+normal serum group, coculture cell+serum containing optimal drug group, siRNA FTO+normal serum group, siRNA FTO-NC+normal serum group, and siRNA FTO+serum containing optimal drug group. The vitality and cell cycle changes of co-cultured cells were detected. The level and markers of homing BMSC were detected by immunofluorescence staining. The expression of Notch1 pathway related genes were detected by qRT-PCR. The expression of Notch1 pathway related proteins were detected by Western blot. Results Compared with bronchial epithelial cell group, the co-cultured cell group showed an increase in the homing level of BMSCs and the expression of C-X-C motif chemokine receptor 4 (CXCR4), stromal cell-derived factor 1 (SDF-1), Notch1, transcription factor recombination signal binding protein-J (RBP-J), and hairy enhancer of split 1 (Hes1) proteins. Compared with the co-cultured cell group and co-cultured cell+normal serum group, the co-cultured cell+serum containing optimal drug group showed an increase in the homing level of BMSCs and the expressions of CXCR4 and SDF-1, while the protein and mRNA levels of Notch1 and Hes1 decreased. Compared with the siRNA FTO-NC+normal serum group, the siRNA FTO+normal serum group showed an increase in the levels of Notch1, activated Notch1, RBP-J, Hes1 protein, and cell viability, while the level of homing BMSC decreased. Compared with siRNA FTO+normal serum group, the levels of Notch1, RBP-J mRNA, activated Notch1, and Hes1 protein decreased, while the level of homing BMSCs increased in siRNA FTO+serum containing optimal drug group. The levels of Notch1, RBP-J, and Hes1 mRNA were reduced in the co-cultured cells+serum containing optimal drug group. Compared with siRNA FTO+serum containing optimal drug group, the expressions of Notch1, activated Notch1, RBP-J, Hes1 protein and cell viability decreased, while the level of homing BMSCs increased in the co-cultured cells+serum containing optimal drug group. Conclusion Yanghe Pingchuan Granules may promote the homing of BMSCs in asthma and alleviate asthma inflammation by upregulating the expression of FTO and inhibiting the expression of downstream genes in the Notch1 signaling pathway.
Animals
;
Receptor, Notch1/genetics*
;
Mesenchymal Stem Cells/cytology*
;
Asthma/genetics*
;
Drugs, Chinese Herbal/pharmacology*
;
Signal Transduction/drug effects*
;
Rats
;
Coculture Techniques
;
Alpha-Ketoglutarate-Dependent Dioxygenase FTO/genetics*
;
Epithelial Cells/metabolism*
;
Rats, Sprague-Dawley
;
Cells, Cultured
;
Male
2.ANXA2 and NF-κB positive feedback loop promotes high glucose-induced pyroptosis in renal tubular epithelial cells.
Jiayi YANG ; Yang LUO ; Zixuan ZHU ; Wenbin TANG
Journal of Central South University(Medical Sciences) 2025;50(6):940-954
OBJECTIVES:
Pyroptosis plays a critical role in tubulointerstitial lesions of diabetic kidney disease (DKD). Annexin A2 (ANXA2) is involved in cell proliferation, apoptosis, and adhesion and may be closely related to DKD, but its specific mechanism remains unclear. This study aims to investigate the role and molecular mechanism of ANXA2 in high glucose-induced pyroptosis of renal tubular epithelial cells, providing new targets for DKD prevention and treatment.
METHODS:
Human renal tubular epithelial HK-2 cells were divided into a normal glucose group (5.5 mmol/L), a high glucose group (30.0 mmol/L), and a osmotic control group (24.5 mmol/L mannitol+5.5 mmol/L glucose). ANXA2 expression was modulated by overexpression of plasmids and small interfering RNA (siRNA). Cell proliferation was measured by 5-ethynyl-2'-deoxyuridine (EdU) assay, apoptosis by flow cytometry, and ANXA2, p50, and p65 subcellular localization by immunofluorescence. Western blotting was employed to detect α-smooth muscle actin (α-SMA), fibronectin (FN), and collagen type IV (Col-IV). Real-time fluorescence quantitative PCR (RT-qPCR) and Western blotting were used to analyze nuclear factor-κB (NF-κB) subunits p50/p65 and the pyroptosis pathway factors NLR family Pyrin domain containing 3 (NLRP3), caspase-1, inferleukin (IL)-1β, and IL-18. Protein interactions between ANXA2 and p50/p65 were examined by co-immunoprecipitation, while chromatin immunoprecipitation (ChIP) and dual-luciferase reporter assays were used to examine NF-κB binding to the ANXA2 promoter.
RESULTS:
High glucose upregulated ANXA2 expression and promoted its nuclear translocation (P<0.01). High glucose reduced cell proliferation, increased apoptosis, and elevated α-SMA, FN, and Col-IV expression (all P<0.05); ANXA2 overexpression aggravated these effects (all P<0.05), while ANXA2 knockdown reversed them (all P<0.05). High glucose activated NF-κB and increased NLRP3, caspase-1, L-1β, and IL-18 mRNA and protein expression (all P<0.05); ANXA2 overexpression further enhanced this, whereas knockdown suppressed NF-κB activation and downstream factors (all P<0.05). Co-immunoprecipitation confirmed ANXA2 directly binds the NF-κB subunit p65. ChIP assays revealed p65 binds specifically to ANXA2 promoter regions (ChIP-2, ChIP-4, and ChIP-6), and luciferase activity in corresponding mutant constructs (M2, M4, and M6) was significantly increased versus controls (all P<0.05), confirming positive transcriptional regulation of ANXA2 by p65.
CONCLUSIONS
ANXA2 and NF-κB form a positive feedback loop that sustains NLRP3 inflammasome activation, promotes pyroptosis pathway activation, and aggravates high glucose-induced renal tubular epithelial cell injury. Targeting ANXA2 or blocking its interaction with p65 may be a novel strategy to slow DKD progression.
Humans
;
Pyroptosis/drug effects*
;
Annexin A2/physiology*
;
Epithelial Cells/cytology*
;
Kidney Tubules/cytology*
;
Glucose/pharmacology*
;
Diabetic Nephropathies/metabolism*
;
NF-kappa B/metabolism*
;
Cell Line
;
Cell Proliferation
;
Transcription Factor RelA/metabolism*
;
Feedback, Physiological
3.Nodakenin ameliorates TNBS-induced experimental colitis in mice by inhibiting pyroptosis of intestinal epithelial cells.
Ju HUANG ; Lixia YIN ; Minzhu NIU ; Zhijun GENG ; Lugen ZUO ; Jing LI ; Jianguo HU
Journal of Southern Medical University 2025;45(2):261-268
OBJECTIVES:
To investigate the therapeutic mechanism of nodakenin for Crohn's disease (CD)-like colitis in mice.
METHODS:
Using a colonic organoid model with lipopolysaccharide (LPS)- and ATP-induced pyroptosis, we investigated the effects of nodakenin on pyroptosis, intestinal barrier function and inflammatory response by detecting key pyroptosis-regulating factors and assessing changes in permeability and pro-inflammatory factors. In a mouse model of 2,4,6-trinitrobenzenesulfonic acid (TNBS)-induced CD-like colitis, the therapeutic effect of nodakenin was evaluated by measuring changes in body weight, DAI score, colonic histopathologies, inflammation score, intestinal barrier function and intestinal epithelial cell pyroptosis. The mechanism of nodakenin protection against pyroptosis of intestinal epithelial cells was explored using network pharmacology analysis and in vivo and in vitro experiments.
RESULTS:
In LPS- and ATP-induced colonic organoids, treatment with nodakenin significantly inhibited the expressions of NLRP3, GSDMD-N, cleaved caspase-1 and caspase-11, improved intestinal FITC-dextran (FD4, 4000) permeability, and decreased the levels of IL-1β and IL-18. In the mouse model of TNBS-induced colitis, nodakenin treatment significantly alleviated weight loss, reduced DAI score, inflammatory cell infiltration and inflammation score, and decreased serum FD4 and I-FABP levels and bacteria translocation to the mesenteric lymph nodes, spleen and liver. The mice with nodakenin treatment had also lowered expressions of NLRP3, GSDMD-N, cleaved caspase-1 and caspase-11 in the intestinal mucosa. Network pharmacology analysis suggested that the inhibitory effect of nodakenin on colitis was associated with the PI3K/Akt pathway. In both the colonic organoid model and mouse models of colitis, nodakenin effectively inhibited the activation of the PI3K/Akt pathway, and the application of IGF-1, a PI3K/Akt pathway activator, strongly attenuated the protective effect of nodakenin against intestinal epithelial cell pyroptosis and intestinal barrier dysfunction.
CONCLUSIONS
Nodakenin protects intestinal barrier function and alleviates CD-like colitis in mice at least partly by inhibiting PI3K/Akt signaling to reduce intestinal epithelial cell pyroptosis.
Animals
;
Pyroptosis/drug effects*
;
Mice
;
Trinitrobenzenesulfonic Acid
;
Colitis/drug therapy*
;
Epithelial Cells/drug effects*
;
Intestinal Mucosa/cytology*
;
Disease Models, Animal
;
Coumarins/pharmacology*
;
NLR Family, Pyrin Domain-Containing 3 Protein/metabolism*
;
Crohn Disease/drug therapy*
4.C6TSEDRVAJZ, a combination of small-molecule compounds, induces differentiation of human placental fibroblasts into epithelioid cells in vitro.
Zhenjia DAI ; Qunwei GAO ; Mengjiao YING ; Ao WANG ; Juan HONG ; Chunjing WANG ; Yu GUO ; Changqing LIU ; Gaofeng LIU
Journal of Southern Medical University 2025;45(2):322-330
OBJECTIVES:
To reprogram human placental fibroblasts (HPFs) into chemically induced epithelioid-like cells (ciEP-Ls) using a combination of small-molecule compounds.
METHODS:
HPFs cultured under normoxic conditions were identified using immunofluorescence assay, PCR and chromosomal karyotyping. Under hypoxic conditions (37 ℃, 5% O2), HPFs were cultured in a medium containing small-molecule compounds C6TSEDRVAJZ (CHIR99021, 616452, TTNPB, SAG, EPZ5676, DZNep, Ruxolitinib, VTP50469, Afuresertib, JNK-IN-8, and EZM0414), and the cell morphology was observed daily. The expression levels of epithelial cell markers in the induced cells were detected by immunofluorescence, Western blotting and PCR. Chromosomal karyotyping of the induced cells was performed and the induction efficiency was calculated.
RESULTS:
Before induction, HPFs showed positive expressions of fibroblast surface markers CD34 and vimentin and were negative for epithelial surface markers. PCR results showed high expressions of fibroblast-specific genes S100A4 and COL1A1 in HPFs with a normal human diploid karyotype. After one day of induction, the HPFs underwent morphological changes from a multinodular spindle shape to a round or polygonal shape, which was morphologically characteristic of ciEP-Ls. On day 4 of induction, the cells exhibited high expressions of the epithelial cell markers E-cadherin and Lin28A. RT-qPCR results also showed that the cells expressed the epithelial markers Smad3, GLi3, PAX8, WT1, KRT19, and KRT18 with significantly down-regulated expressions of all the fibroblast surface markers and a normal human diploid karyotype. The reprogramming efficiency of HPFs into ciEP-Ls ranged from (64.53±2.8)% to (68.10±3.6)%.
CONCLUSIONS
The small-molecule compound combination C6TSEDRVAJZ is capable of inducing HPFs into ciEP-Ls under hypoxic conditions with a high induction efficiency.
Humans
;
Fibroblasts/drug effects*
;
Pregnancy
;
Female
;
Cell Differentiation/drug effects*
;
Pyrimidines/pharmacology*
;
Placenta/cytology*
;
Cells, Cultured
;
Pyridines/pharmacology*
;
Pyrazoles/pharmacology*
;
Epithelial Cells/cytology*
5.Protective effect of Bufei Yishen Formula against cigarette smoke extract-induced human bronchial epithelial cell damage and its mechanism.
Zhengyuan FAN ; Zihan SHEN ; Ya LI ; Tingting SHEN ; Gaofeng LI ; Suyun LI
Journal of Southern Medical University 2025;45(7):1372-1379
OBJECTIVES:
To evaluate the protective effect of Bufei Yishen Formula (BYF) against cigarette smoke extract (CSE)-induced injuries in human bronchial epithelial BEAS-2B cells and explore the underlying mechanism.
METHODS:
BEAS-2B cells exposed to CSE were treated with normal rat serum, BYF-medicated rat serum at low or high doses, pyrrolidine dithiocarbamate (PDTC, a NF-κB inhibitor), PDTC combined with high-dose BYF-medicated serum, or S-carbomethyloysteine (S-CMC, as the positive control). CCK-8 assay was used to determine the optimal concentration and treatment time of CSE, BYF-medicated serum and S-CMC. The treated cells were examined for inflammatory factor levels in the supernatant and cellular expressions of MUC5AC and MUC5B using ELISA, cell ultrastructural changes with transmission electron microscopy, and cell apoptosis rate using flow cytometry. The expression levels of TLR4/NF‑κB pathway-associated mRNAs and proteins were determined by qRT-PCR and Western blotting.
RESULTS:
CSE exposure significantly increased secretions of IL-1β, IL-6 and TNF-α, mRNA and protein expressions of MUC5AC and MUC5B, and early and total apoptosis rates in BEAS-2B cells, where the presence of apoptotic bodies was detected. CSE also significantly enhanced the mRNA and protein expressions of TLR4, I-κB, and NF-κB and reduced mRNA and protein expressions of AQP5. Treatments of the CSE-exposed cells with BYF-medicated serum, PDTC and S-CMC all significantly lowered inflammatory factor levels, MUC5AC and MUC5B expressions, and early and total cell apoptosis rates, and partly reversed the changes in cellular ultrastructure and mRNA and protein expressions of the TLR4/NF-κB pathway, and the effects were the most conspicuous following the combined treatment with high-dose BYF-medicated serum and PDTC.
CONCLUSIONS
BYF can inhibit cell apoptosis, inflammation and mucus hypersecretion in CSE-induced BEAS-2B cells by inhibiting the TLR4/NF-κB signaling pathway.
Humans
;
Epithelial Cells/cytology*
;
Drugs, Chinese Herbal/pharmacology*
;
NF-kappa B/metabolism*
;
Bronchi/cytology*
;
Smoke/adverse effects*
;
Apoptosis/drug effects*
;
Mucin 5AC/metabolism*
;
Cell Line
;
Toll-Like Receptor 4/metabolism*
;
Mucin-5B/metabolism*
;
Signal Transduction/drug effects*
;
Nicotiana
;
Rats
;
Thiocarbamates/pharmacology*
;
Animals
6.Effects of Rg_1 on LPS-induced apoptosis and autophagy of lung epithelial cells.
Qi-Jian JI ; Zhao-Rui SUN ; Zhi-Zhou YANG ; Wei ZHANG ; Yi REN ; Li-Ping CAO ; Liang LI ; Shi-Nan NIE
China Journal of Chinese Materia Medica 2019;44(8):1648-1653
This paper aimed to study the protective effect of ginsenoside Rg_1 on endotoxin(LPS)-induced apoptosis of lung epithelial cells and its mechanism of action. Mouse lung epithelial cells(MLE-12) were first treated with LPS. The autophagy changes and apoptosis and the relationship with concentration and time of LPS were observed. Then,the level of autophagy in MLE-12 was regulated at a specific concentration and action time of LPS,and the changes of apoptosis were observed. Secondly,ginsenoside Rg_1 and autophagy inhibitor 3-MA were added respectively at the same concentration and action time of LPS. The lung epithelial cells were grouped to observe the effect of ginsenoside Rg_1 on LPS-induced apoptosis of lung epithelial cells and its mechanism. In the animal experiment,the mice were grouped and tested by apoptosis protein,lung injury score and HE staining section to verify whether ginsenoside Rg_1 has a protective effect on LPS-induced lung injury. The results showed that apoptosis and autophagy increased as the rise of concentration after treatment with LPS for 12 h. The apoptosis increased gradually,and the autophagy increased first and then decreased over time at the LPS concentration of 25 g·L-1. The apoptosis of LPS group was higher than that of control group,and LPS+3-MA group increased further,while apoptosis decreased significantly in LPS+RAM(rapamycin,autophagy promoter) group. The autophagy increased in LPS group,decreased in LPS+3-MA group and increased in LPS+RAM group. The apoptosis of LPS group was higher than that of control group,and the apoptosis of LPS+Rg_1 group decreased. The apoptosis of LPS+Rg_1+3-MA group increased again. The autophagy of LPS group further increased after administration of ginsenoside Rg_1,but decreased after administration of 3-MA. In the in vivo experiments in mice,the apoptosis of LPS group increased significantly compared with the control group,while LPS + ginsenoside Rg_1 group decreased. Lung injury score and HE staining also conformed to the above trend. LPS can induce the apoptosis of lung epithelial cells in a time-dependent and concentration-dependent manner. The autophagy of lung epithelial cells increases with the rise of LPS concentration. At the specific concentration of LPS,autophagy increases first and then decreases after 12-16 hours. Proper increase of autophagy in lung epithelial cells within a certain period of time can reduce the apoptosis induced by LPS,while inhibition of autophagy can increase apoptosis. Ginsenoside Rg_1 has a protective effect on lung cancer epithelial cell apoptosis induced by autophagy.
Animals
;
Apoptosis
;
Autophagy
;
Cells, Cultured
;
Epithelial Cells
;
drug effects
;
Ginsenosides
;
pharmacology
;
Lipopolysaccharides
;
Lung
;
cytology
;
Mice
7.Polysaccharide extracts of Astragalus membranaceus and Atractylodes macrocephala promote intestinal epithelial cell migration by activating the polyamine-mediated K channel.
Dan ZENG ; Can HU ; Ru-Liu LI ; Chuan-Quan LIN ; Jia-Zhong CAI ; Ting-Ting WU ; Jing-Jing SUI ; Wen-Biao LU ; Wei-Wen CHEN
Chinese Journal of Natural Medicines (English Ed.) 2018;16(9):674-682
Astragalus membranaceus (Radix Astragali, RA) and Atractylodes macrocephala (Rhizoma Atractylodis Macrocephalae, RAM) are often used to treat gastrointestinal diseases. In the present study, we determined the effects of polysaccharides extracts from these two herbs on IEC-6 cell migration and explored the potential underlying mechanisms. A migration model with IEC-6 cells was induced using a single-edged razor blade along the diameter of cell layers in six-well polystyrene plates. The cells were grown in control media or media containing spermidine (5 μmol·L, SPD), alpha-difluoromethylornithine (2.5 mmol·L, DFMO), 4-Aminopyridine (40 μmol·L, 4-AP), the polysaccharide extracts of RA or RAM (50, 100, or 200 mg·L), DFMO plus SPD, or DFMO plus polysaccharide extracts of RA or RAM for 12 or 24 h. Next, cytosolic free Ca ([Ca]) was measured using laser confocal microscopy, and cellular polyamine content was quantified with HPLC. Kv1.1 mRNA expression was assessed using RT-qPCR and Kv1.1 and RhoA protein expressions were measured with Western blotting analysis. A cell migration assay was carried out using Image-Pro Plus software. In addition, GC-MS was introduced to analyze the monosaccharide composition of both polysaccharide extracts. The resutls showed that treatment with polysaccharide extracts of RA or RAM significantly increased cellular polyamine content, elevated [Ca] and accelerated migration of IEC-6 cells, compared with the controls (P < 0.01). Polysaccharide extracts not only reversed the inhibitory effects of DFMO on cellular polyamine content and [Ca], but also restored IEC-6 cell migration to control level (P < 0.01 or < 0.05). Kv1.1 mRNA and protein expressions were increased (P < 0.05) after polysaccharide extract treatment in polyamine-deficient IEC-6 cells and RhoA protein expression was increased. Molar ratios of D-ribose, D-arabinose, L-rhamnose, D-mannose, D-glucose, and D-galactose was 1.0 : 14.1 : 0.3 : 19.9 : 181.3 : 6.3 in RA and 1.0 : 4.3 : 0.1 : 5.7 : 2.8 : 2.2 in RAM. In conclusion, treatment with RA and RAM polysaccharide extracts stimulated migration of intestinal epithelial cells via a polyamine-Kv1.1 channel activated signaling pathway, which facilitated intestinal injury healing.
Animals
;
Astragalus propinquus
;
chemistry
;
Atractylodes
;
chemistry
;
Cell Line
;
Cell Movement
;
drug effects
;
Drugs, Chinese Herbal
;
chemistry
;
isolation & purification
;
pharmacology
;
Epithelial Cells
;
cytology
;
drug effects
;
metabolism
;
Intestines
;
cytology
;
drug effects
;
Kv1.1 Potassium Channel
;
genetics
;
metabolism
;
Polyamines
;
metabolism
;
Polysaccharides
;
chemistry
;
isolation & purification
;
pharmacology
;
Rats
;
Rhizome
;
chemistry
;
Signal Transduction
;
drug effects
;
rhoA GTP-Binding Protein
;
metabolism
8.Epithelial cell adhesion efficacy of a novel peptide identified by panning on a smooth titanium surface.
Hidemichi KIHARA ; David M KIM ; Masazumi NAGAI ; Toshiki NOJIRI ; Shigemi NAGAI ; Chia-Yu CHEN ; Cliff LEE ; Wataru HATAKEYAMA ; Hisatomo KONDO ; John DA SILVA
International Journal of Oral Science 2018;10(3):21-21
Epithelial attachment via the basal lamina on the tooth surface provides an important structural defence mechanism against bacterial invasion in combating periodontal disease. However, when considering dental implants, strong epithelial attachment does not exist throughout the titanium-soft tissue interface, making soft tissues more susceptible to peri-implant disease. This study introduced a novel synthetic peptide (A10) to enhance epithelial attachment. A10 was identified from a bacterial peptide display library and synthesized. A10 and protease-activated receptor 4-activating peptide (PAR4-AP, positive control) were immobilized on commercially pure titanium. The peptide-treated titanium showed high epithelial cell migration ability during incubation in platelet-rich plasma. We confirmed the development of dense and expanded BL (stained by Ln5) with pericellular junctions (stained by ZO1) on the peptide-treated titanium surface. In an adhesion assay of epithelial cells on A10-treated titanium, PAR4-AP-treated titanium, bovine root and non-treated titanium, A10-treated titanium and PAR4-AP-treated titanium showed significantly stronger adhesion than non-treated titanium. PAR4-AP-treated titanium showed significantly higher inflammatory cytokine release than non-treated titanium. There was no significant difference in inflammatory cytokine release between A10-treated and non-treated titanium. These results indicated that A10 could induce the adhesion and migration of epithelial cells with low inflammatory cytokine release. This novel peptide has a potentially useful application that could improve clinical outcomes with titanium implants and abutments by reducing or preventing peri-implant disease.
Amino Acid Sequence
;
Animals
;
Benzeneacetamides
;
chemical synthesis
;
pharmacology
;
Cattle
;
Cell Adhesion
;
drug effects
;
Cell Movement
;
drug effects
;
Cells, Cultured
;
Cytokines
;
metabolism
;
Dental Implants
;
Enzyme-Linked Immunosorbent Assay
;
Epithelial Attachment
;
drug effects
;
Epithelial Cells
;
cytology
;
metabolism
;
Microscopy, Confocal
;
Microscopy, Electron, Scanning
;
Piperidones
;
chemical synthesis
;
pharmacology
;
Platelet-Rich Plasma
;
Receptors, Thrombin
;
Surface Properties
;
Titanium
;
chemistry
9.Combination of musk and olibanum promotes proliferation of prostate stem cells in mice: An experimental study.
Qun-Fang LIN ; Pei HUANG ; Xue-Jun SHANG ; Xue-Fei TIAN ; Guo-Min ZHANG ; Rui-Song GAO ; Xing ZHOU ; Hui-Ying LIU ; Qing ZHOU
National Journal of Andrology 2017;23(2):157-163
Objective:
To study the effects of muskolibanum combination on the proliferation and differentiation of prostate stem cells.
METHODS:
We cultured prostate epithelial cells and urogenital sinus mesenchymal (UGSM) cells from 7-10 d old C57BL/6 mice and 16-18 d old pregnant C57BL/6 mice, transplanted the mixed suspension of the two types of cells under the kidney envelope of SCIDCB.17 male mice, and harvested the transplants 30 days later. We randomly divided the SCIDCB.17 mice into four groups to be treated intragastrically with musk (n = 8), olibanum (n = 8), musk+olibanum (n = 7), and normal saline (blank control, n = 8)) respectively, all for 14 days. Then we collected the kidney tissue for observation of the morphology of the glandular tubes and differentiation of different subsets of stem cells by HE staining and determination of the expressions and distribution of P63, CD133, CD117 and Sca1 by immunohistochemistry and Western blot.
RESULTS:
A system was successfully established for the isolation and mixed culture of Sca1 Lin+ CD49f+ (LSC) cells of prostate stem cells and UGSM cells of the mouse embryonic prostate. Immunohistochemistry showed positive expressions of P63, CD133, Sca1, and CD117 in the prostatic acinar epithelia and proved the presence of prostatic acinar epithelial structure in the transplants. Compared with the blank control group, the expressions of CD133, Sca1 and CD117 were significantly increased in the musk, olibanum, and musk+olibanum groups (P< 0.05), higher in the musk+olibanum than in the musk or olibanum group (P< 0.05), and their protein expressions were even more elevated in the musk+olibanum group (P< 0.01), with statistically significant difference from the olibanum group (P< 0.05).
CONCLUSIONS
The combination of musk and olibanum can improve the proliferation and differentiation of prostate stem cells.
Animals
;
Cell Differentiation
;
drug effects
;
Cell Proliferation
;
drug effects
;
Drug Therapy, Combination
;
Epithelial Cells
;
cytology
;
drug effects
;
Fatty Acids, Monounsaturated
;
pharmacology
;
Female
;
Frankincense
;
pharmacology
;
Male
;
Mesenchymal Stem Cells
;
cytology
;
drug effects
;
Mice
;
Mice, Inbred C57BL
;
Mice, SCID
;
Pregnancy
;
Prostate
;
cytology
;
Random Allocation
;
Receptor Protein-Tyrosine Kinases
;
Receptors, Cholinergic
;
Stem Cells
;
cytology
;
drug effects
10.Arctiin ameliorates advanced oxidation protein product-induced epithelial-to- mesenchymal transition in HK-2 cells by inhibiting endoplasmic reticulum stress.
Jun ZHANG ; Li-Li HUANG ; Xiu-Jie LIANG ; Yue WANG ; Na DUAN ; Xiao-Hong XIANG ; Shuang-Shuang SHU ; Ting-Ting GUO ; Lei YANG ; Xun TANG
Journal of Southern Medical University 2016;36(6):833-837
OBJECTIVETo investigate the effect of arctiin on advanced oxidation protein product (AOPP)-induced epithelial-to-mesenchymal transition (EMT) in tubular cells and explore the mechanisms underlying this effect.
METHODSHuman proximal tubular cells (HK-2 cells) were treated with bovine serum albumin (BSA) or AOPPs in the presence or absence of arctiin. The expressions of E-cadherin, vimentin, and GRP78 at the protein and mRNA levels in the cells were examined using Western blotting and quantitative real-time PCR. The level of reactive oxygen species (ROS) was measured by flow cytometry with DCFH-DA as the fluorescent probe.
RESULTSCompared with BSA-treated cells, the cells treated with AOPPs showed decreased expression of epithelial cell marker E-cadherin and overexpression of mesenchymal marker vimentin and endoplasmic reticulum stress marker GRP78 with an increased ROS level. These changes induced by AOPPs were partly inhibited by arctiin.
CONCLUSIONArctiin can ameliorate AOPP-induced EMT in tubular cells by inhibiting endoplasmic reticulum stress, and oxidative stress response may participate in this process.
Advanced Oxidation Protein Products ; adverse effects ; Cadherins ; metabolism ; Cell Line ; Endoplasmic Reticulum Stress ; Epithelial Cells ; cytology ; drug effects ; Epithelial-Mesenchymal Transition ; Furans ; pharmacology ; Glucosides ; pharmacology ; Heat-Shock Proteins ; metabolism ; Humans ; Kidney Tubules ; cytology ; drug effects ; Oxidative Stress ; Reactive Oxygen Species ; metabolism ; Vimentin ; metabolism

Result Analysis
Print
Save
E-mail