1.Impact of lithocholic acid on the osteogenic and adipogenic differentiation balance of bone marrow mesenchymal stem cells.
Cui WANG ; Jiao LI ; Lingyun LU ; Lu LIU ; Xijie YU
Chinese Journal of Reparative and Reconstructive Surgery 2024;38(1):82-90
		                        		
		                        			OBJECTIVE:
		                        			To Investigate the effects of lithocholic acid (LCA) on the balance between osteogenic and adipogenic differentiation of bone marrow mesenchymal stem cells (BMSCs).
		                        		
		                        			METHODS:
		                        			Twelve 10-week-old SPF C57BL/6J female mice were randomly divided into an experimental group (undergoing bilateral ovariectomy) and a control group (only removing the same volume of adipose tissue around the ovaries), with 6 mice in each group. The body mass was measured every week after operation. After 4 weeks post-surgery, the weight of mouse uterus was measured, femur specimens of the mice were taken for micro-CT scanning and three-dimensional reconstruction to analyze changes in bone mass. Tibia specimens were taken for HE staining to calculate the number and area of bone marrow adipocytes in the marrow cavity area. ELISA was used to detect the expression of bone turnover markers in the serum. Liver samples were subjected to real-time fluorescence quantitative PCR (RT-qPCR) to detect the expression of key genes related to bile acid metabolism, including cyp7a1, cyp7b1, cyp8b1, and cyp27a1. BMSCs were isolated by centrifugation from 2 C57BL/6J female mice (10-week-old). The third-generation cells were exposed to 0, 1, 10, and 100 μmol/L LCA, following which cell viability was evaluated using the cell counting kit 8 assay. Subsequently, alkaline phosphatase (ALP) staining and oil red O staining were conducted after 7 days of osteogenic and adipogenic induction. RT-qPCR was employed to analyze the expressions of osteogenic-related genes, namely ALP, Runt-related transcription factor 2 (Runx2), and osteocalcin (OCN), as well as adipogenic-related genes including Adiponectin (Adipoq), fatty acid binding protein 4 (FABP4), and peroxisome proliferator-activated receptor γ (PPARγ).
		                        		
		                        			RESULTS:
		                        			Compared with the control group, the body mass of the mice in the experimental group increased, the uterus atrophied, the bone mass decreased, the bone marrow fat expanded, and the bone metabolism showed a high bone turnover state. RT-qPCR showed that the expressions of cyp7a1, cyp8b1, and cyp27a1, which were related to the key enzymes of bile acid metabolism in the liver, decreased significantly ( P<0.05), while the expression of cyp7b1 had no significant difference ( P>0.05). Intervention with LCA at concentrations of 1, 10, and 100 μmol/L did not demonstrate any apparent toxic effects on BMSCs. Furthermore, LCA inhibited the expressions of osteogenic-related genes (ALP, Runx2, and OCN) in a dose-dependent manner, resulting in a reduction in ALP staining positive area. Concurrently, LCA promoted the expressions of adipogenic-related genes (Adipoq, FABP4, and PPARγ), and an increase in oil red O staining positive area.
		                        		
		                        			CONCLUSION
		                        			After menopause, the metabolism of bile acids is altered, and secondary bile acid LCA interferes with the balance of osteogenic and adipogenic differentiation of BMSCs, thereby affecting bone remodelling.
		                        		
		                        		
		                        		
		                        			Female
		                        			;
		                        		
		                        			Mice
		                        			;
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Core Binding Factor Alpha 1 Subunit/pharmacology*
		                        			;
		                        		
		                        			PPAR gamma/metabolism*
		                        			;
		                        		
		                        			Steroid 12-alpha-Hydroxylase/metabolism*
		                        			;
		                        		
		                        			Mice, Inbred C57BL
		                        			;
		                        		
		                        			Cell Differentiation
		                        			;
		                        		
		                        			Osteogenesis
		                        			;
		                        		
		                        			Mesenchymal Stem Cells
		                        			;
		                        		
		                        			Bile Acids and Salts/pharmacology*
		                        			;
		                        		
		                        			Bone Marrow Cells
		                        			;
		                        		
		                        			Cells, Cultured
		                        			;
		                        		
		                        			Azo Compounds
		                        			
		                        		
		                        	
2.MOR106 alleviates inflammation in mice with atopic dermatitis by blocking the JAK2/STAT3 signaling pathway and inhibiting IL-17C-mediated Tfh cell differentiation.
Limin TIAN ; Xiaohui HUYAN ; Sen YANG ; Mengjie WANG ; Yuenan YANG
Chinese Journal of Cellular and Molecular Immunology 2024;40(1):26-32
		                        		
		                        			
		                        			Objective To explore the significance of interleukin-17C(IL-17C)-mediated follicular helper T cell (Tfh) differentiation in atopic dermatitis (AD) model. Methods BALB/c mice were divided into control group, AD model group, low-dose MOR106 (anti-IL-17C huIgG1)(MDR106-L)treatment group and high-dose MOR106 (MOR106-H) treatment group, 8 mice in each group. Except for the control group, all the other groups were treated with 2, 4- dinitrochlorobenzene (DNCB) to establish AD models. The low-dose and high-dose MOR106 groups were treated with 5 mg/kg or 10 mg/kg MOR106 respectively. The differentiation of Tfh cell subsets in peripheral blood of mice was analyzed by flow cytometry, and the expression of Janus kinase 2/signal transducer and activator of transcription 3(JAK2/STAT3) signal pathway protein in skin tissue was detected by Western blot analysis. Results Compared with the control group, the dermatitis severity score, mass difference between two ears, spleen mass and spleen index of DNCB group increased significantly, while those of MOR106-L group and MOR106-H group decreased significantly. Compared with the control group, the Tfh subgroup of AD mice showed deregulated differentiation, resulting in a significant increase in the percentage of CD4+CXCR5+IFN-γ+Tfh1 cells, CD4+CXCR5+IL-17A+Tfh17 and CD4+CXCR5+IL-21+Tfh21 cells, and a significant decrease in the percentage of CD4+CXCR5+IL-10+Tfh10 cells and CD4+CXCR5+FOXP3+Tfr cells in peripheral blood. The protein levels of phosphorylated JAK2(p-JAK2) and p-STAT3 were significantly increased. MOR106 effectively reversed these changes of Tfh1, Tfh10, Tfh17, Tfh21 and Tfr cells in peripheral blood of AD mice. Compared with AD group, the levels of p-JAK2 and p-STAT3 protein in low-dose and high-dose MOR106 treatment groups decreased significantly. Conclusion MOR106 can reduce the inflammatory response of AD mice by blocking JAK2/STAT3 signaling pathway and inhibiting the differentiation of Tfh cells mediated by IL-17C.
		                        		
		                        		
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Mice
		                        			;
		                        		
		                        			Dermatitis, Atopic/drug therapy*
		                        			;
		                        		
		                        			Interleukin-17
		                        			;
		                        		
		                        			T Follicular Helper Cells
		                        			;
		                        		
		                        			Janus Kinase 2
		                        			;
		                        		
		                        			Dinitrochlorobenzene
		                        			;
		                        		
		                        			Inflammation
		                        			;
		                        		
		                        			Cell Differentiation
		                        			;
		                        		
		                        			Signal Transduction
		                        			
		                        		
		                        	
3.TREM-2 Drives Development of Multiple Sclerosis by Promoting Pathogenic Th17 Polarization.
Siying QU ; Shengfeng HU ; Huiting XU ; Yongjian WU ; Siqi MING ; Xiaoxia ZHAN ; Cheng WANG ; Xi HUANG
Neuroscience Bulletin 2024;40(1):17-34
		                        		
		                        			
		                        			Multiple sclerosis (MS) is a neuroinflammatory demyelinating disease, mediated by pathogenic T helper 17 (Th17) cells. However, the therapeutic effect is accompanied by the fluctuation of the proportion and function of Th17 cells, which prompted us to find the key regulator of Th17 differentiation in MS. Here, we demonstrated that the triggering receptor expressed on myeloid cells 2 (TREM-2), a modulator of pattern recognition receptors on innate immune cells, was highly expressed on pathogenic CD4-positive T lymphocyte (CD4+ T) cells in both patients with MS and experimental autoimmune encephalomyelitis (EAE) mouse models. Conditional knockout of Trem-2 in CD4+ T cells significantly alleviated the disease activity and reduced Th17 cell infiltration, activation, differentiation, and inflammatory cytokine production and secretion in EAE mice. Furthermore, with Trem-2 knockout in vivo experiments and in vitro inhibitor assays, the TREM-2/zeta-chain associated protein kinase 70 (ZAP70)/signal transducer and activator of transcription 3 (STAT3) signal axis was essential for Th17 activation and differentiation in EAE progression. In conclusion, TREM-2 is a key regulator of pathogenic Th17 in EAE mice, and this sheds new light on the potential of this therapeutic target for MS.
		                        		
		                        		
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Mice
		                        			;
		                        		
		                        			CD4-Positive T-Lymphocytes/pathology*
		                        			;
		                        		
		                        			Cell Differentiation
		                        			;
		                        		
		                        			Encephalomyelitis, Autoimmune, Experimental/metabolism*
		                        			;
		                        		
		                        			Mice, Inbred C57BL
		                        			;
		                        		
		                        			Multiple Sclerosis
		                        			;
		                        		
		                        			Th1 Cells/pathology*
		                        			
		                        		
		                        	
4.Neural Stem Cell Competition.
Neuroscience Bulletin 2024;40(2):277-279
5.Sema3A secreted by sensory nerve induces bone formation under mechanical loads.
Hongxiang MEI ; Zhengzheng LI ; Qinyi LV ; Xingjian LI ; Yumeng WU ; Qingchen FENG ; Zhishen JIANG ; Yimei ZHOU ; Yule ZHENG ; Ziqi GAO ; Jiawei ZHOU ; Chen JIANG ; Shishu HUANG ; Juan LI
International Journal of Oral Science 2024;16(1):5-5
		                        		
		                        			
		                        			Bone formation and deposition are initiated by sensory nerve infiltration in adaptive bone remodeling. Here, we focused on the role of Semaphorin 3A (Sema3A), expressed by sensory nerves, in mechanical loads-induced bone formation and nerve withdrawal using orthodontic tooth movement (OTM) model. Firstly, bone formation was activated after the 3rd day of OTM, coinciding with a decrease in sensory nerves and an increase in pain threshold. Sema3A, rather than nerve growth factor (NGF), highly expressed in both trigeminal ganglion and the axons of periodontal ligament following the 3rd day of OTM. Moreover, in vitro mechanical loads upregulated Sema3A in neurons instead of in human periodontal ligament cells (hPDLCs) within 24 hours. Furthermore, exogenous Sema3A restored the suppressed alveolar bone formation and the osteogenic differentiation of hPDLCs induced by mechanical overload. Mechanistically, Sema3A prevented overstretching of F-actin induced by mechanical overload through ROCK2 pathway, maintaining mitochondrial dynamics as mitochondrial fusion. Therefore, Sema3A exhibits dual therapeutic effects in mechanical loads-induced bone formation, both as a pain-sensitive analgesic and a positive regulator for bone formation.
		                        		
		                        		
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Bone Remodeling
		                        			;
		                        		
		                        			Cell Differentiation
		                        			;
		                        		
		                        			Osteogenesis
		                        			;
		                        		
		                        			Semaphorin-3A/pharmacology*
		                        			;
		                        		
		                        			Trigeminal Ganglion/metabolism*
		                        			
		                        		
		                        	
6.Potassium dehydroandrographolide succinate regulates the MyD88/CDH13 signaling pathway to enhance vascular injury-induced pathological vascular remodeling.
Qiru GUO ; Jiali LI ; Zheng WANG ; Xiao WU ; Zhong JIN ; Song ZHU ; Hongfei LI ; Delai ZHANG ; Wangming HU ; Huan XU ; Lan YANG ; Liangqin SHI ; Yong WANG
Chinese Journal of Natural Medicines (English Ed.) 2024;22(1):62-74
		                        		
		                        			
		                        			Pathological vascular remodeling is a hallmark of various vascular diseases. Previous research has established the significance of andrographolide in maintaining gastric vascular homeostasis and its pivotal role in modulating endothelial barrier dysfunction, which leads to pathological vascular remodeling. Potassium dehydroandrographolide succinate (PDA), a derivative of andrographolide, has been clinically utilized in the treatment of inflammatory diseases precipitated by viral infections. This study investigates the potential of PDA in regulating pathological vascular remodeling. The effect of PDA on vascular remodeling was assessed through the complete ligation of the carotid artery in C57BL/6 mice. Experimental approaches, including rat aortic primary smooth muscle cell culture, flow cytometry, bromodeoxyuridine (BrdU) incorporation assay, Boyden chamber cell migration assay, spheroid sprouting assay, and Matrigel-based tube formation assay, were employed to evaluate the influence of PDA on the proliferation and motility of smooth muscle cells (SMCs). Molecular docking simulations and co-immunoprecipitation assays were conducted to examine protein interactions. The results revealed that PDA exacerbates vascular injury-induced pathological remodeling, as evidenced by enhanced neointima formation. PDA treatment significantly increased the proliferation and migration of SMCs. Further mechanistic studies disclosed that PDA upregulated myeloid differentiation factor 88 (MyD88) expression in SMCs and interacted with T-cadherin (CDH13). This interaction augmented proliferation, migration, and extracellular matrix deposition, culminating in pathological vascular remodeling. Our findings underscore the critical role of PDA in the regulation of pathological vascular remodeling, mediated through the MyD88/CDH13 signaling pathway.
		                        		
		                        		
		                        		
		                        			Mice
		                        			;
		                        		
		                        			Rats
		                        			;
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Myeloid Differentiation Factor 88/metabolism*
		                        			;
		                        		
		                        			Vascular Remodeling
		                        			;
		                        		
		                        			Cell Proliferation
		                        			;
		                        		
		                        			Vascular System Injuries/pathology*
		                        			;
		                        		
		                        			Carotid Artery Injuries/pathology*
		                        			;
		                        		
		                        			Molecular Docking Simulation
		                        			;
		                        		
		                        			Muscle, Smooth, Vascular
		                        			;
		                        		
		                        			Cell Movement
		                        			;
		                        		
		                        			Mice, Inbred C57BL
		                        			;
		                        		
		                        			Signal Transduction
		                        			;
		                        		
		                        			Succinates/pharmacology*
		                        			;
		                        		
		                        			Potassium/pharmacology*
		                        			;
		                        		
		                        			Cells, Cultured
		                        			;
		                        		
		                        			Diterpenes
		                        			;
		                        		
		                        			Cadherins
		                        			
		                        		
		                        	
7.Isolation, culture and validation of CD34+ vascular wall-resident stem cells from mice.
Li-Ju YANG ; Ying MA ; Yuan LI ; Qing-Ya DANG ; Jun CHENG ; Yan YANG ; Peng-Yun LI
Acta Physiologica Sinica 2023;75(2):205-215
		                        		
		                        			
		                        			Vascular wall-resident stem cells (VW-SCs) play a critical role in maintaining normal vascular function and regulating vascular repair. Understanding the basic functional characteristics of the VW-SCs will facilitate the study of their regulation and potential therapeutic applications. The aim of this study was to establish a stable method for the isolation, culture, and validation of the CD34+ VW-SCs from mice, and to provide abundant and reliable cell sources for further study of the mechanisms involved in proliferation, migration and differentiation of the VW-SCs under various physiological and pathological conditions. The vascular wall cells of mouse aortic adventitia and mesenteric artery were obtained by the method of tissue block attachment and purified by magnetic microbead sorting and flow cytometry to obtain the CD34+ VW-SCs. Cell immunofluorescence staining was performed to detect the stem cell markers (CD34, Flk-1, c-kit, Sca-1), smooth muscle markers (SM22, SM MHC), endothelial marker (CD31), and intranuclear division proliferation-related protein (Ki-67). To verify the multipotency of the isolated CD34+ VW-SCs, endothelial differentiation medium EBM-2 and fibroblast differentiation medium FM-2 were used. After culture for 7 days and 3 days respectively, endothelial cell markers and fibroblast markers of the differentiated cells were evaluated by immunofluorescence staining and q-PCR. Furthermore, the intracellular Ca2+ release and extracellular Ca2+ entry signaling were evaluated by TILLvisION system in Fura-2/AM loaded cells. The results showed that: (1) High purity (more than 90%) CD34+ VW-SCs from aortic adventitia and mesenteric artery of mice were harvested by means of tissue block attachment method and magnetic microbead sorting; (2) CD34+ VW-SCs were able to differentiate into endothelial cells and fibroblasts in vitro; (3) Caffeine and ATP significantly activated intracellular Ca2+ release from endoplasmic reticulum of CD34+ VW-SCs. Store-operated Ca2+ entry (SOCE) was activated by using thapsigargin (TG) applied in Ca2+-free/Ca2+ reintroduction protocol. This study successfully established a stable and efficient method for isolation, culture and validation of the CD34+ VW-SCs from mice, which provides an ideal VW-SCs sources for the further study of cardiovascular diseases.
		                        		
		                        		
		                        		
		                        			Mice
		                        			;
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Endothelial Cells
		                        			;
		                        		
		                        			Cell Differentiation/physiology*
		                        			;
		                        		
		                        			Stem Cells
		                        			;
		                        		
		                        			Adventitia
		                        			;
		                        		
		                        			Fibroblasts
		                        			;
		                        		
		                        			Cells, Cultured
		                        			;
		                        		
		                        			Antigens, CD34/metabolism*
		                        			
		                        		
		                        	
8.Mammalian DMRTs: Structure, function and relationship with cancer.
Hai-Long LI ; Zi-Cong ZOU ; Chi FANG ; Yi-Ping ZHENG ; Xiao-Ming GUO ; Wei-Hong YANG
Acta Physiologica Sinica 2023;75(2):269-278
		                        		
		                        			
		                        			DMRT, a gene family related to sexual determination, encodes a large group of transcription factors (DMRTs) with the double-sex and mab-3 (DM) domain (except for DMRT8), which is able to bind to and regulate DNAs. Current studies have shown that the DMRT gene family plays a critical role in the development of sexual organs (such as gender differentiation, gonadal development, germ cell development, etc.) as well as extrasexual organs (such as musculocartilage development, nervous system development, etc.). Additionally, it has been suggested that DMRTs may be involved in the cancer development and progression (such as prostate cancer, breast cancer, lung cancer, etc.). This review summarizes the research progress about the mammalian DMRTs' structure, function and its critical role in cancer development, progression and therapy (mainly in human and mice), which suggests that DMRT gene could be a candidate gene in the study of tumor formation and therapeutic strategy.
		                        		
		                        		
		                        		
		                        			Male
		                        			;
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Mice
		                        			;
		                        		
		                        			Transcription Factors/genetics*
		                        			;
		                        		
		                        			Mammals/metabolism*
		                        			;
		                        		
		                        			Cell Differentiation
		                        			;
		                        		
		                        			Neoplasms/genetics*
		                        			
		                        		
		                        	
9.Exercise regulates bone metabolism via microRNAs.
Yu YUAN ; Lin-Zhen RAO ; Shi-Hua ZHANG ; Yang XU ; Ting-Ting LI ; Jun ZOU ; Xi-Quan WENG
Acta Physiologica Sinica 2023;75(3):429-438
		                        		
		                        			
		                        			It has been well documented that exercise can improve bone metabolism, promote bone growth and development, and alleviate bone loss. MicroRNAs (miRNAs) are widely involved in the proliferation and differentiation of bone marrow mesenchymal stem cells, osteoblasts, osteoclasts and other bone tissue cells, and regulation of balance between bone formation and bone resorption by targeting osteogenic factors or bone resorption factors. Thus miRNAs play an important role in the regulation of bone metabolism. Recently, regulation of miRNAs are shown to be one of the ways by which exercise or mechanical stress promotes the positive balance of bone metabolism. Exercise induces changes of miRNAs expression in bone tissue and regulates the expression of related osteogenic factors or bone resorption factors, to further strengthen the osteogenic effect of exercise. This review summarizes relevant studies on the mechanism whereby exercise regulates bone metabolism via miRNAs, providing a theoretical basis for osteoporosis prevention and treatment with exercise.
		                        		
		                        		
		                        		
		                        			Humans
		                        			;
		                        		
		                        			MicroRNAs/metabolism*
		                        			;
		                        		
		                        			Osteogenesis/genetics*
		                        			;
		                        		
		                        			Cell Differentiation
		                        			;
		                        		
		                        			Osteoblasts
		                        			;
		                        		
		                        			Bone Resorption/metabolism*
		                        			
		                        		
		                        	
10.Fine Particulate Matter Exposure Induces Toxicity by Regulating Nuclear Factor-κB/toll-like Receptor 4/myeloid Differentiation Primary Response Signaling Pathways in RAW264.7 Cells.
Mei Zhu ZHENG ; Yao LU ; Ting Ting LU ; Peng QIN ; Yu Qiu LI ; Dong Fang SHI
Biomedical and Environmental Sciences 2023;36(5):458-462
            
Result Analysis
Print
Save
E-mail