1.Advances in mechanism of traditional Chinese medicine in inhibiting angiogenesis in ovarian cancer.
Mao-Yan TANG ; Dan-Ni DING ; Ya-Ya XIE ; Fang SHEN ; Jia LI ; Fang-Yuan LIU ; Feng-Juan HAN
China Journal of Chinese Materia Medica 2023;48(24):6572-6581
		                        		
		                        			
		                        			Ovarian cancer is one of the three major cancers in gynecology. Ovarian cancer has insidious symptoms in its early stages and mostly has progressed to advanced stages when detected. Surgical treatment combined with chemotherapy is currently the main treatment, but the 5-year survival rate is still less than 45%. Angiogenesis is a key step in the growth and metastasis of ovarian cancer. The inhibition of ovarian cancer angiogenesis has become a new hotspot in anti-tumor targeted therapy, which has many advantages such as less drug resistance, high specificity, few side effects, and broad anti-tumor spectrum. Modern research has confirmed that traditional Chinese medicine(TCM) can inhibit tumor angiogenesis by inhibiting the expression of pro-angiogenic factors, up-regulating the expression of anti-angiogenic factors, inhibiting the proliferation of vascular endothelial cells, reducing the density of tumor microvessels, and regulating related signaling pathways, with unique advantages in the treatment of ovarian cancer. This paper presented a review of the role of TCM in inhibiting ovarian cancer angiogenesis in order to provide references for the optimization of clinical ovarian cancer treatment strategies.
		                        		
		                        		
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Female
		                        			;
		                        		
		                        			Medicine, Chinese Traditional
		                        			;
		                        		
		                        			Vascular Endothelial Growth Factor A/metabolism*
		                        			;
		                        		
		                        			Endothelial Cells/metabolism*
		                        			;
		                        		
		                        			Angiogenesis
		                        			;
		                        		
		                        			Angiogenesis Inhibitors/therapeutic use*
		                        			;
		                        		
		                        			Ovarian Neoplasms/genetics*
		                        			;
		                        		
		                        			Neovascularization, Pathologic/genetics*
		                        			
		                        		
		                        	
2.Exosomal FZD10 derived from non-small cell lung cancer cells promotes angiogenesis of human umbilical venous endothelial cells in vitro.
Xiao Feng WU ; Ri Ming ZHAN ; Da Zhao CHENG ; Li CHEN ; Tian Yu WANG ; Xu Dong TANG
Journal of Southern Medical University 2022;42(9):1351-1358
		                        		
		                        			OBJECTIVE:
		                        			To investigate the effect of exosomal FZD10 derived from non-small cell lung cancer (NSCLC) cells on angiogenesis of human umbilical venous endothelial cells (HUVECs) and explore the possible mechanism.
		                        		
		                        			METHODS:
		                        			We analyzed the expression of FZD10 in two NSCLC cell lines (95D and H1299 cells), normal human bronchial epithelial cells (BEAS-2B cells) and their exosomes isolated by ultracentrifugation. Cultured HUVECs were treated with the exosomes derived from NSCLC cells or NSCLC cells transfected with FZD10-siRNA, and the changes in tube formation ability of the cells were analyzed using an in vitro angiogenesis assay. ELISA was performed to determine the concentration of VEGFA and Ang-1 in the conditioned media of HUVECs, and RT-qPCR was used to analyze the mRNA levels of VEGFA and Ang-1 in the HUVECs. The effects of exosomal FZD10 on the activation of PI3K, Erk1/2 and YAP/TAZ signaling pathways were evaluated using Western blotting.
		                        		
		                        			RESULTS:
		                        			Compared with BEAS-2B cells and their exosomes, 95D and H1299 cells and their exosomes all expressed high levels of FZD10 (P < 0.01). The exosomes derived from 95D and H1299 cells significantly enhanced tube formation ability and increased the expressions of VEGFA and Ang-1 protein and mRNA in HUVECs (P < 0.01), but FZD10 knockdown in 95D and H1299 cells obviously inhibited these effects of the exosomes. Exosomal FZD10 knockdown suppressed the activation of PI3K and Erk1/2 signaling pathways, but had no obvious effect on the activation of YAP/TAZ signaling pathway.
		                        		
		                        			CONCLUSION
		                        			Exosomal FZD10 derived from NSCLC cells promotes HUVEC angiogenesis in vitro, the mechanism of which may involve the activation of PI3K and Erk1/2 signaling pathways.
		                        		
		                        		
		                        		
		                        			Carcinoma, Non-Small-Cell Lung/metabolism*
		                        			;
		                        		
		                        			Cell Proliferation
		                        			;
		                        		
		                        			Culture Media, Conditioned
		                        			;
		                        		
		                        			Exosomes
		                        			;
		                        		
		                        			Frizzled Receptors/metabolism*
		                        			;
		                        		
		                        			Human Umbilical Vein Endothelial Cells/metabolism*
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Lung Neoplasms/metabolism*
		                        			;
		                        		
		                        			MicroRNAs/genetics*
		                        			;
		                        		
		                        			Neovascularization, Pathologic/metabolism*
		                        			;
		                        		
		                        			Phosphatidylinositol 3-Kinases/metabolism*
		                        			;
		                        		
		                        			RNA, Messenger/metabolism*
		                        			;
		                        		
		                        			RNA, Small Interfering/metabolism*
		                        			
		                        		
		                        	
3.Study on the Therapeutic Effect of Lenalidomide on Hemophilic Arthropathy.
Ye-Fan WANG ; Zhen-Yang LIN ; Fei-Xu ZHANG ; Xin-Yue ZHOU ; Xia WU ; Xiao XIAO ; Jun-Jiang SUN ; Bao-Lai HUA
Journal of Experimental Hematology 2022;30(5):1549-1556
		                        		
		                        			OBJECTIVE:
		                        			To explore the effect of lenalidomide on human fibroblast-like synovial cells (HFLS) and the therapeutic efficacy on hemophilic arthropathy in hemophilia A mice model.
		                        		
		                        			METHODS:
		                        			In vitro, to remodel the inflammatory environment of synovial tissue after hemorrhage, ferric citrate and recombinant TNF-α were added into the cell culture medium of HFLS. Cell Counting Kit-8 (CCK-8), Enzyme-linked immunosorbent assay (ELISA), Quantitative Real-time PCR (RT-qPCR) and flow cytometry were employed for detection of the effects of lenalidomide on the proliferation ability, pro-inflammatory cytokines release and apoptosis of HFLS cells. In vivo, hemophilia arthropathy was remodeled in hemophilia A mice by induction of hemarthrosis. A series of doses of lenalidomide (0.1, 0.3 and 1.0 g/kg) was administrated intra-articularly. Tissues of knee joints were collected on the 14th day after administration, and the protective effect of lenalidomide on arthritis in hemophilia A mice were evaluated by RT-qPCR and histological grading.
		                        		
		                        			RESULTS:
		                        			In vitro, compared with the untreated control group, lenalidomide could significantly inhibit the proliferation of HFLS cells (P<0.05), and the effect was the most significant when the concentration was 0.01 μmol/L (P<0.001). Compared with the control group, lenalidomide could significantly inhibit the expression levels of TNF-α, IL-1β, IL-6 and IFN-γ in HFLS cells (P<0.05). The flow cytometry results showed that lenalidomide could enhance the apoptotis of HFLS cells (P<0.05). The results of RT-qPCR showed that lenalidomide could significantly reduce the mRNA expression levels of TNF-α, IL-1β, IL-6,MCP-1 and VEGF in the joint tissues (P<0.05). Histological results showed that compared with the injured group, lenalidomide could significantly reduce the pathological sequela after hemarthrosis induction, e.g. synovial thickening and neo-angiogenesis in the synovium. The protection displayed a dose-response pattern roughly.
		                        		
		                        			CONCLUSION
		                        			In vitro, lenalidomide can inhibit the proliferation of HFLS cells, promote their apoptosis, and inhibit the expression of pro-inflammatory cytokines. In vivo, lenalidomide can significantly decrease the expression of pro-inflammatory cytokines in the joints of mice, and prevent the development of inflammation and neo-angiogenesis. The results provide a theoretical and experimental basis for the clinical application of lenalidomide in the treatment of hemophilic arthropathy.
		                        		
		                        		
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Arthritis
		                        			;
		                        		
		                        			Cytokines/metabolism*
		                        			;
		                        		
		                        			Hemarthrosis/pathology*
		                        			;
		                        		
		                        			Hemophilia A/genetics*
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Interleukin-6
		                        			;
		                        		
		                        			Lenalidomide
		                        			;
		                        		
		                        			Mice
		                        			;
		                        		
		                        			Neovascularization, Pathologic
		                        			;
		                        		
		                        			RNA, Messenger
		                        			;
		                        		
		                        			Sincalide
		                        			;
		                        		
		                        			Tumor Necrosis Factor-alpha
		                        			;
		                        		
		                        			Vascular Endothelial Growth Factor A
		                        			
		                        		
		                        	
4.Roles of integrin in tumor development and the target inhibitors.
Zhao-He LI ; You ZHOU ; You-Xiang DING ; Qing-Long GUO ; Li ZHAO
Chinese Journal of Natural Medicines (English Ed.) 2019;17(4):241-251
		                        		
		                        			
		                        			Integrin is a large family of cell adhesion molecules (CAMs) which involves in the interaction of cells/cells and cells/ extracellular matrix (ECM) to mediate cell proliferation, differentiation, adhesion, migration, etc. In recent years, aberrant expression of integrin has been clearly found in many tumor studies, indicating that integrin is closely related to tumor formation and development. Meanwhile, it has effects on tumor cell differentiation, cell migration, proliferation and tumor neovascularization. The study of drugs targeting integrins is of great significance for the clinical treatment of tumors. Because of its important role in tumorigenesis and development, integrin has become a promising target for the treatment of cancer. This review summarizes the role of integrin in tumor development and the current state of integrin inhibitors to provide a valuable reference for subsequent research.
		                        		
		                        		
		                        		
		                        			Antineoplastic Agents
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			therapeutic use
		                        			;
		                        		
		                        			Biological Products
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			therapeutic use
		                        			;
		                        		
		                        			Cell Movement
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Cell Proliferation
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Extracellular Matrix
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Integrins
		                        			;
		                        		
		                        			antagonists & inhibitors
		                        			;
		                        		
		                        			classification
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Neoplasms
		                        			;
		                        		
		                        			drug therapy
		                        			;
		                        		
		                        			pathology
		                        			;
		                        		
		                        			Neovascularization, Pathologic
		                        			;
		                        		
		                        			drug therapy
		                        			;
		                        		
		                        			pathology
		                        			;
		                        		
		                        			Signal Transduction
		                        			;
		                        		
		                        			drug effects
		                        			
		                        		
		                        	
5.Beta-adrenergic signaling on neuroendocrine differentiation, angiogenesis, and metastasis in prostate cancer progression.
Asian Journal of Andrology 2019;21(3):253-259
		                        		
		                        			
		                        			Prostate cancer is a complex, heterogeneous disease that mainly affects the older male population with a high-mortality rate. The mechanisms underlying prostate cancer progression are still incompletely understood. Beta-adrenergic signaling has been shown to regulate multiple cellular processes as a mediator of chronic stress. Recently, beta-adrenergic signaling has been reported to affect the development of aggressive prostate cancer by regulating neuroendocrine differentiation, angiogenesis, and metastasis. Here, we briefly summarize and discuss recent advances in these areas and their implications in prostate cancer therapeutics. We aim to provide a better understanding of the contribution of beta-adrenergic signaling to the progression of aggressive prostate cancer.
		                        		
		                        		
		                        		
		                        			Cell Differentiation/genetics*
		                        			;
		                        		
		                        			Disease Progression
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Male
		                        			;
		                        		
		                        			Neoplasm Metastasis
		                        			;
		                        		
		                        			Neovascularization, Pathologic/pathology*
		                        			;
		                        		
		                        			Neuroendocrine Cells/pathology*
		                        			;
		                        		
		                        			Prostatic Neoplasms/pathology*
		                        			;
		                        		
		                        			Receptors, Adrenergic, beta
		                        			;
		                        		
		                        			Signal Transduction
		                        			
		                        		
		                        	
6.Mechanism of gambogenic acid in resisting angiogenesis of lung cancer in vitro.
Hui CHENG ; Yun-Long WANG ; Jing-Jing SU ; Rong-Feng HU ; Qing-Lin LI
China Journal of Chinese Materia Medica 2018;43(21):4311-4316
		                        		
		                        			
		                        			The aim of this paper was to observe the effect of gambogenic acid on angiogenesis of lung cancer and its preliminary mechanism. After culturing lung adenocarcinoma A549 cells, the conditioned medium was treated with gambogenic acid and then used to culture human umbilical vein endothelial cells (HUVECs) to establish the indirect contact cell co-culture system. A two-dimensional culture model of HUVEC was established with matrigel to observe the effect of gambogenic acid on angiogenesis. DAPI staining was used to observe the morphological changes in HUVEC cells after treatment with gambogenic acid under the fluorescence microscope. Annexin V-FITC/PI staining and flow cytometry analysis were used to determine gambogenic acid's effect on HUVEC cell apoptosis rate. The protein expressions of PI3K, p-PI3K, Akt, p-Akt were measured by Western blot. PTEN-siRNA was transfected into cells, and RT-PCR was used to detect the expression levels of PI3K and Akt genes. Gambogenic acid can significantly inhibit angiogenesis, and its inhibitory effect was dose-dependent. DAPI staining showed apoptotic morphological features of HUVEC cells under fluorescence microscope. Annexin V-FITC/PI staining showed that gambogenic acid induced apoptosis in HUVECs. The results of Western blot showed that the expressions of p-PI3K and p-Akt protein were down-regulated with gambogenic acid, while the expressions of PI3K and Akt protein was insignificant. The results of RT-PCR indicated that the expressions of PI3K and Akt protein were up-regulated by PTEN siRNA. Gambogenic acid can inhibit angiogenesis in lung cancer in vitro, and the mechanism of inhibiting angiogenesis may be related to the PI3K/Akt signaling pathway.
		                        		
		                        		
		                        		
		                        			A549 Cells
		                        			;
		                        		
		                        			Apoptosis
		                        			;
		                        		
		                        			Coculture Techniques
		                        			;
		                        		
		                        			Human Umbilical Vein Endothelial Cells
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Lung Neoplasms
		                        			;
		                        		
		                        			drug therapy
		                        			;
		                        		
		                        			pathology
		                        			;
		                        		
		                        			Neovascularization, Pathologic
		                        			;
		                        		
		                        			pathology
		                        			;
		                        		
		                        			PTEN Phosphohydrolase
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			Phosphatidylinositol 3-Kinases
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Proto-Oncogene Proteins c-akt
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Transfection
		                        			;
		                        		
		                        			Xanthenes
		                        			;
		                        		
		                        			pharmacology
		                        			
		                        		
		                        	
7.Novel Molecular Mechanisms in the Development of Non-Alcoholic Steatohepatitis.
Davide POVERO ; Ariel E FELDSTEIN
Diabetes & Metabolism Journal 2016;40(1):1-11
		                        		
		                        			
		                        			Non-alcoholic fatty liver disease (NAFLD) is one of the most common causes of chronic liver disease in adults and children worldwide. NAFLD has become a severe health issue and it can progress towards a more severe form of the disease, the non-alcoholic steatohepatitis (NASH). A combination of environmental factors, host genetics, and gut microbiota leads to excessive accumulation of lipids in the liver (steatosis), which may result in lipotoxicity and trigger hepatocyte cell death, liver inflammation, fibrosis, and pathological angiogenesis. NASH can further progress towards liver cirrhosis and cancer. Over the last few years, cell-derived extracellular vesicles (EVs) have been identified as effective cell-to-cell messengers that transfer several bioactive molecules in target cells, modulating the pathogenesis and progression of NASH. In this review, we focused on recently highlighted aspects of molecular pathogenesis of NASH, mediated by EVs via their bioactive components. The studies included in this review summarize the state of art regarding the role of EVs during the progression of NASH and bring novel insight about the potential use of EVs for diagnosis and therapeutic strategies for patients with this disease.
		                        		
		                        		
		                        		
		                        			Adult
		                        			;
		                        		
		                        			Cell Death
		                        			;
		                        		
		                        			Child
		                        			;
		                        		
		                        			Diagnosis
		                        			;
		                        		
		                        			Fatty Liver*
		                        			;
		                        		
		                        			Fibrosis
		                        			;
		                        		
		                        			Genetics
		                        			;
		                        		
		                        			Hepatocytes
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Inflammation
		                        			;
		                        		
		                        			Liver
		                        			;
		                        		
		                        			Liver Cirrhosis
		                        			;
		                        		
		                        			Liver Diseases
		                        			;
		                        		
		                        			Microbiota
		                        			;
		                        		
		                        			Neovascularization, Pathologic
		                        			
		                        		
		                        	
8.MicroRNA-210 Plays a Critical Role in the Angiogenic Effect of Isoprenaline on Human Umbilical Vein Endothelial Cells via Regulation of Noncoding RNAs.
You-You YAN ; Zhi-Hui WANG ; Lei ZHAO ; Dan-Dan SONG ; Chao QI ; Lu-Lu LIU ; Jun-Nan WANG
Chinese Medical Journal 2016;129(22):2676-2682
BACKGROUNDβ-adrenoceptors play a crucial regulatory role in blood vessel endothelial cells. Isoprenaline (ISO, a β-adrenergic agonist) has been reported to promote angiogenesis through upregulation of vascular endothelial growth factor (VEGF) expression; however, the underlying mechanism remains to be investigated. It is widely accepted that certain noncoding RNAs, including microRNAs (miRNAs) and long noncoding RNAs (lncRNAs), can regulate endothelial cell behavior, including their involvement in angiogenesis. Therefore, we aimed to investigate whether noncoding RNAs participate in ISO-mediated angiogenesis using human umbilical vein endothelial cells (HUVECs).
METHODSWe evaluated VEGF-A messenger RNA (mRNA) and protein levels in ISO-treated HUVECs by quantitative real-time polymerase chain reaction and enzyme-linked immunosorbent assay, respectively. To establish whether noncoding RNAs are associated with ISO-mediated angiogenesis, we measured expression of the miRNAs miR-210, miR-21, and miR-1, as well as that of the lncRNAs growth arrest-specific transcript 5 (GAS5), maternally expressed 3 (MEG3), and metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) in HUVECs exposed to ISO. Furthermore, to ascertain its importance in ISO-mediated angiogenesis, we constructed the HUVECs with overexpressing miR-210 and detected the subsequent expression of VEGF-A and noncoding RNAs. All statistical analyses were performed using SPSS 16.0 software. Intergroup comparisons were carried out by one-way analysis of variance.
RESULTSVEGF-A mRNA levels were elevated in the ISO group (1.57 ± 0.09) compared to those in the control group (P < 0.01). Moreover, concentrations of VEGF-A in culture supernatants significantly differed between the control (113.00 ± 19.21 pg/ml) and ISO groups (287.00 ± 20.27 pg/ml; P< 0.01). Expression of miR-1, miR-21, and miR-210 was higher (3.89 ± 0.44, 2.87 ± 087, and 3.33 ± 1.31, respectively) in ISO-treated cells than that in controls (P < 0.01), whereas that of GAS5 and MEG3 (0.22 ± 0.10 and 0.58 ± 0.16, respectively) was lower as a result of ISO administration (P < 0.05). There was no significant difference in the expression of MALAT1 between the groups. Interestingly, miR-210 overexpression heightened the levels of VEGF-A and miR-21 (5.87 ± 1.24 and 2.74 ± 1.15, respectively; P< 0.01) and reduced those of GAS5 and MEG3 (0.19 ± 0.01 and 0.09 ± 0.05, respectively; P< 0.01).
CONCLUSIONSISO-mediated angiogenesis was associated with altered expression of miR-210, miR-21, and the lncRNAs GAS5 and MEG3. The effects of miR-210 on the expression of VEGF-A and noncoding RNAs were similar to those of ISO, indicating that it might play an important role in ISO-mediated angiogenesis.
Cell Line ; Cell Survival ; drug effects ; Human Umbilical Vein Endothelial Cells ; drug effects ; metabolism ; Humans ; Isoproterenol ; pharmacology ; MicroRNAs ; genetics ; physiology ; Neovascularization, Pathologic ; genetics ; RNA, Long Noncoding ; genetics ; Real-Time Polymerase Chain Reaction ; Vascular Endothelial Growth Factor A ; metabolism
9.Effect of jianpi-jiedu formula on tumor angiogenesis-relevant genes expression in colorectal cancer.
Dan MAO ; Sanlin LEI ; Jin'an MA ; Li SHI ; Shaofan ZHANG ; Jianhua HUANG ; Xinyi LIU ; Dengfeng DING ; Yingjin ZHANG ; Lei FENG ; Sifang ZHANG
Journal of Central South University(Medical Sciences) 2016;41(12):1297-1304
		                        		
		                        			
		                        			To investigate the effect of the jianpi-jiedu formula (JPJD) on the expression of angiogenesis-relevant genes in colon cancer.
 Methods: Crude extract was obtained from JPJD by water extract method. The effect of JPJD crude extract on colon cancer cell proliferation capacity was determined by MTT assays. The IC50 value was calculated by GraphPad Prism5 software. Affymetrix gene expression profiling chip was used to detect significant differences in expressions of genes after JPJD intervention, and pathway enrichment analysis was performed to analyze the differentially expressed genes. Ingenuity Pathway Analysis software was applied to analyze differentially expressed genes relevant to tumor angiogenesis based on mammalian target of rapamycin (mTOR) signaling pathway and then the network diagram was built. Western blot was used to verify the protein levels of key genes related to tumor angiogenesis.
 Results: JPJD crud extract inhibited the proliferation capacity in colon cancer cells. The IC50 values in 24, 48, and 72 hours after treatment were 13.060, 9.646 and 8.448 mg/mL, respectively. The results of chip showed that 218 genes significantly upgraded, and 252 genes significantly downgraded after JPJD treatment. Most of the genes were related to the function of biosynthesis, metabolism, cell apoptosis, antigen extraction, angiogenesis and so on. There were 12 differentially expressed angiogenesis genes. IPA software analysis showed that the JPJD downregulated expression of sphingomyelin phosphodiesterase 3 (SMPD3), VEGF, vascular endothelial growth factor A (VEGFA), integrin subunit alpha 1 (ITGA1), cathepsin B (CTSB), and cathepsin S (CTSS) genes, while upregulated expressions of GAB2 and plasminogen activator, urokinase receptor (PLAUR) genes in the colorectal cancer cell. Western blot results demonstrated that JPJD obviously downregulated expressions of phospho-mTOR (P-mTOR), signal transducer and activator of transcription 3 (STAT3), hypoxia inducible factor-1α (HIF-1α), and VEGF proteins, while obviously upregulated the level of phospho-P53 (P-P53) protein.
 Conclusion: JPJD may inhibit colorectal tumor angiogenesis through regulation of the mTOR-HIF-1α-VEGF signal pathway.
		                        		
		                        		
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Blotting, Western
		                        			;
		                        		
		                        			Cathepsin B
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Cathepsins
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Cell Line, Tumor
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Colorectal Neoplasms
		                        			;
		                        		
		                        			blood supply
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			Down-Regulation
		                        			;
		                        		
		                        			Drugs, Chinese Herbal
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			Gene Expression Profiling
		                        			;
		                        		
		                        			methods
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Hypoxia-Inducible Factor 1, alpha Subunit
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Integrin alpha Chains
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Neovascularization, Pathologic
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			Receptors, Urokinase Plasminogen Activator
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			STAT3 Transcription Factor
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Signal Transduction
		                        			;
		                        		
		                        			Sphingomyelin Phosphodiesterase
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			TOR Serine-Threonine Kinases
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Tumor Suppressor Protein p53
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Up-Regulation
		                        			;
		                        		
		                        			Vascular Endothelial Growth Factor A
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			metabolism
		                        			
		                        		
		                        	
10.Tie-1: A potential target for anti-angiogenesis therapy.
Ping YANG ; Na CHEN ; Jing-hui JIA ; Xue-jiao GAO ; Shi-han LI ; Jing CAI ; Zehua WANG
Journal of Huazhong University of Science and Technology (Medical Sciences) 2015;35(5):615-622
		                        		
		                        			
		                        			The tyrosine kinase system angiopoietin (Ang)/Tie interacts with vascular endothelial growth factor pathway and regulates vessel quiescence in adults as well as later steps of the angiogenic cascade related to vessel maturation. Since all Angs are able to bind to Tie-2 but none binds to Tie-1, the function of Tie-2 and its ligands have captured attention. However, emerging evidence indicates unique roles of the orphan receptor Tie-1 in angiogenesis under physiological and pathological conditions. It is required for maintaining vascular endothelial cell integrity and survival during murine embryo development and in adult and may be involved in modulating differentiation of hematopoietic cells in adult. Tie-1 exhibits poor tyrosine kinase activity and signals via forming heterodimers with Tie-2, inhibiting Tie-2 signaling mediated by Angs. This inhibition can be relieved by Tie-1 ectodomain cleavage mediated by tumor- and inflammatory-related factors, which causes destabilization of vessels and initiates vessel remodeling. Up-regulated Tie-1 expression has been found not only in some leukemia cells and tumor related endothelial cells but also in cytoplasm of carcinoma cells of a variety of human solid tumors, which is associated with tumor progression. In addition, it has pro-inflammatory functions in endothelial cells and is involved in some inflammatory diseases associated with angiogenesis. Recent research indicated that Tie-1 gene ablation exhibited significant effects on tumor blood- and lymph-angiogenesis and improved anti-Ang therapy, suggesting Tie-1 may be a potential target for tumor anti-angiogenesis treatment.
		                        		
		                        		
		                        		
		                        			Angiogenesis Inhibitors
		                        			;
		                        		
		                        			therapeutic use
		                        			;
		                        		
		                        			Angiopoietins
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Embryo, Mammalian
		                        			;
		                        		
		                        			Embryonic Development
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			Endothelial Cells
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			pathology
		                        			;
		                        		
		                        			Gene Expression Regulation, Developmental
		                        			;
		                        		
		                        			Gene Expression Regulation, Neoplastic
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Mice
		                        			;
		                        		
		                        			Neoplasms
		                        			;
		                        		
		                        			drug therapy
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			pathology
		                        			;
		                        		
		                        			Neovascularization, Pathologic
		                        			;
		                        		
		                        			drug therapy
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			pathology
		                        			;
		                        		
		                        			Protein Binding
		                        			;
		                        		
		                        			Receptor, TIE-1
		                        			;
		                        		
		                        			antagonists & inhibitors
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Receptor, TIE-2
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Signal Transduction
		                        			
		                        		
		                        	
            
Result Analysis
Print
Save
E-mail