2.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*
3.Effects of components in stasis-resolving and collateral-dredging Chinese herbal medicines on angiogenesis and inflammatory response of human umbilical vein endothelial cells induced by VEGF.
Bing LUAN ; Rong YUAN ; Qi-Qi XIN ; Wei-Hong CONG ; Ping SONG
China Journal of Chinese Materia Medica 2022;47(3):737-744
The present study investigated the mechanism of components in stasis-resolving and collateral-dredging Chinese herbal medicines, including scutellarin(Scu), paeonol(Pae), and hydroxy safflower yellow A(HSYA), in the treatment of psoriasis by regulating angiogenesis and inflammation. The human umbilical vein endothelial cells(HUVECs) cultured in vitro were divided into a normal group, a model group, a VEGFR tyrosine kinase inhibitor Ⅱ(VRI) group, and Scu, Pae, and HSYA groups with low, me-dium, and high doses. Cell viability was detected by the CCK-8 assay. Cell migration was detected by wound healing assay. Tube formation assay was used to measure the tube formation ability. Western blot was used to detect the protein expression of the VEGFR2/Akt/ERK1/2 signaling pathway. The secretion levels of inflammatory cytokines IFN-γ, IL-1β, IL-6, and TNF-α were detected by ELISA. The results showed that compared with the model group, all the Scu, Pae, and HSYA groups could reduce cell viability, inhibit cell migration and tube formation(P<0.05, P<0.01), and down-regulated the protein expression of VEGFR2, p-VEGFR2, Akt, p-Akt, ERK1/2, and p-ERK1/2. Scu and Pae could down-regulate VEGFR2 expression(P<0.05, P<0.01), while other groups only showed a downward trend. Scu and Pae significantly reduced IFN-γ and IL-6 levels(P<0.01), and HSYA significantly reduced the levels of IFN-γ, IL-1β, and IL-6(P<0.01). Scu, Pae, and HSYA had no significant effect on TNF-α. The results suggested that Scu, Pae, and HSYA may exert a therapeutic role in psoriasis-related angiogenesis and inflammation by inhibiting VEGFR2/Akt/ERK1/2 signaling pathway and inhibiting the secretion of IFN-γ, IL-1β, and IL-6.
Angiogenesis Inhibitors/pharmacology*
;
China
;
Human Umbilical Vein Endothelial Cells
;
Humans
;
Neovascularization, Pathologic/drug therapy*
;
Vascular Endothelial Growth Factor A/metabolism*
4.Effect of Jianpi Huogu Formula on function damage of vascular endothelial cells induced by glucocorticoid.
Teng-Teng XU ; Jin-Xia WANG ; Rui-Rui MING ; Chao YANG ; Luo-Chang-Ting FANG ; Xiao-Xiao WANG ; Zhi-Xing HU ; Wei-Heng CHEN ; Chun-Fang LIU ; Na LIN
China Journal of Chinese Materia Medica 2022;47(6):1625-1631
This study aimed to observe the intervention effect of Jianpi Huogu Formula(JPHGF) on the functional damage of vascular endothelial cells caused by glucocorticoid, and explore its action mechanism from the PI3 K/Akt and mitogen activated protein kinase(MAPK) signaling pathways. The extracted thoracic aorta ring of normal SD rats were intervened first with vascularendothelial growth factor(VEGF, 20 μg·L-1) and/or sodium succinate(MPS, 0. 04 g·L-1) in vitro and then with JPHGF(8, 16, and 32 μg·L-1) for five mcontinuous ethylpdays, rednisolofollowed nebythe statistics of the number, length, and area of microvessels budding fromvascular rings. In addition, the human umbilical vein endothelial cells(HUVECs) induced by VEGF(20 μg·L-1) were added with MPS(0. 04 g·L-1) and then with JPHGF(8, 16, and 32 μg·L-1) for observing the migration, invasion, and luminal formation abilities of HUVECs in the migration, invasion and luminal formation experiments. The protein expression levels of PI3 K, p-Akt, p-JN K, and p-ERK in HUVECs were assayed by Western blot. The results showed that JPHGF dose-dependently improved the num-ber,length, and area of microvessels in MPS-induced rat thoracic aortic ring, reversed the migration, invasion and lumen formation abiliti es of HUVECs reduced by MPS, and up-regulated the protein expression levels of PI3 K, p-Akt, and p-JNK in HUVECs. All thesehave suggested that JPHGF exerts the protective effect against hormone-induced damage to the angiogenesis of vascular endothelial cells by activating the PI3 K/Akt and MAPK signaling pathways, which has provided reference for exploring the mechanism of JPHGF in treating s teroid-induced avascular necrosis of femoral head(SANFH) and also the experimental evidence for enriching the scientific connotationof spleen-invigorating and blood-activating therapy.
Animals
;
Glucocorticoids/pharmacology*
;
Human Umbilical Vein Endothelial Cells
;
Humans
;
Neovascularization, Pathologic/metabolism*
;
Rats
;
Rats, Sprague-Dawley
;
Vascular Endothelial Growth Factor A/metabolism*
5.Role of high-mobility group box 1 in cancer.
Juan XU ; Pengzuo TAO ; Dongjin LÜ ; Yu'e JIANG ; Quansong XIA
Journal of Central South University(Medical Sciences) 2022;47(4):505-511
High-mobility group box 1 (HMGB1) is a non-histone nuclear protein in most eukaryocytes. Inside the nucleus, HMGB1 plays an important role in several DNA events such as DNA repair, transcription, telomere maintenance, and genome stability. While outside the nucleus, it fulfils more complicated functions, including promoting cell proliferation, inflammation, angiogenesis, immune tolerance and immune escape, which may play a pro-tumoral role.Meanwhile, HMGB1 acts as an anti-tumoral protein by regulating immune cell recruitment and inducing immunogenic cell death (ICD) during the carcinogenesis process. Therefore, abnormal expression of HMGB1 is associated with oncogenesis, development, and metastasis of cancer, which may play a dual role of pro-tumor and anti-tumor.
Carcinogenesis
;
Cell Proliferation
;
HMGB1 Protein/metabolism*
;
Humans
;
Neoplasms/pathology*
;
Neovascularization, Pathologic
6.Agkistrodon halys venom antitumor component-I inhibits vasculogenic mimicry in triple-negative breast cancer cells in vitro by down-regulating MMP2.
Yu GE ; Lin Ming LU ; Shu Yu TIAN ; Yu XIAO ; Shang Fu XIE ; Qi WANG ; Hui ZHI
Journal of Southern Medical University 2022;42(3):438-442
OBJECTIVE:
To investigate the inhibitory effect of agkistrodon halys venom antitumor component-I (AHVAC-I) on vasculogenic mimicry (VM) formation in triple-negative breast cancer MDA-MB-231 cells and explore its possible mechanism.
METHODS:
CCK8 assay was used to determine the optimal concentration of AHVAC-I for cell treatment based on its halfinhibitory concentration (IC50). MDA-MB-231 cells were treated with different concentrations of AHVAC-I or 5-Fu, and the changes in vasomimetic capacity of the cells were examined using Matrigel assay. The expression levels of matrix metalloproteinase-2 (MMP2) and MMP9 in the treated cells were detected using quantitative PCR and Western blotting.
RESULTS:
Compared with the control treatment with culture medium, treatment with 5, 10 and 20 μg/mL AHVAC-I significantly reduced vasomimetic ability of MDA-MB-231 cells in a dose-dependent manner (P < 0.01). MMP2 supplementation obviously restored the vasomimetic ability of the cells inhibited by AHVAC-I.
CONCLUSION
AHVAC-I inhibits VM formation in triplenegative breast cancer cells in vitro by down-regulating MMP2 production.
Agkistrodon/metabolism*
;
Animals
;
Cell Line, Tumor
;
Healthy Life Expectancy
;
Humans
;
Matrix Metalloproteinase 2/metabolism*
;
Neovascularization, Pathologic/metabolism*
;
Triple Negative Breast Neoplasms/metabolism*
;
Venoms
7.Wogonoside alleviates high glucose-induced dysfunction of retinal microvascular endothelial cells and diabetic retinopathy in rats by up-regulating SIRT1.
Xiao Li SHAO ; Jiang Yi YU ; Wei Hui NI
Journal of Southern Medical University 2022;42(4):463-472
OBJECTIVE:
To investigate the effects of wogonoside on high glucose-induced dysfunction of human retinal microvascular endothelial cells (hRMECs) and streptozotocin (STZ)-induced diabetic retinopathy in rats and explore the underlying molecular mechanism.
METHODS:
HRMECs in routine culture were treated with 25 mmol/L mannitol or exposed to high glucose (30 mmol/L glucose) and treatment with 10, 20, 30, 40 μmol/L wogonoside. CCK-8 assay and Transwell assay were used to examine cell proliferation and migration, and the changes in tube formation and monolayer cell membrane permeability were tested. ROS, NO and GSH-ST kits were used to evaluate oxidative stress levels in the cells. The expressions of IL-1β and IL-6 in the cells were examined with qRT-PCR and ELISA, and the protein expressions of VEGF, HIF-1α and SIRT1 were detected using Western blotting. We also tested the effect of wogonoside on retinal injury and expressions of HIF-1α, ROS, VEGF, TNF-α, IL-1β, IL-6 and SIRT1 proteins in rat models of STZ-induced diabetic retinopathy.
RESULTS:
High glucose exposure caused abnormal proliferation and migration, promoted angiogenesis, increased membrane permeability (P < 0.05), and induced inflammation and oxidative stress in hRMECs (P < 0.05). Wogonoside treatment concentration-dependently inhibited high glucose-induced changes in hRMECs. High glucose exposure significantly lowered the expression of SIRT1 in hRMECs, which was partially reversed by wogonoside (30 μmol/L) treatment; interference of SIRT1 obviously attenuated the inhibitory effects of wogonoside against high glucose-induced changes in proliferation, migration, angiogenesis, membrane permeability, inflammation and oxidative stress in hRMECs. In rat models of STZ-induced diabetic retinopathy, wogonoside effectively suppressed retinal thickening (P < 0.05), alleviated STZ-induced retinal injury, and increased the expression of SIRT1 in the retinal tissues (P < 0.001).
CONCLUSION
Wogonoside alleviates retinal damage caused by diabetic retinopathy by up-regulating SIRT1 expression.
Animals
;
Diabetes Mellitus/metabolism*
;
Diabetic Retinopathy/metabolism*
;
Endothelial Cells
;
Flavanones
;
Glucose/pharmacology*
;
Glucosides
;
Inflammation/metabolism*
;
Interleukin-6/metabolism*
;
Neovascularization, Pathologic/metabolism*
;
Rats
;
Reactive Oxygen Species/metabolism*
;
Sirtuin 1/metabolism*
;
Streptozocin/pharmacology*
;
Vascular Endothelial Growth Factor A/metabolism*
8.Expert consensus on off-label use of small molecule anti-angiogenic drugs in the treatment of metastatic breast cancer.
Chinese Journal of Oncology 2022;44(6):523-530
Breast cancer is the most common cancer in the world, and 5-year survival rate of metastatic breast cancer is about 20%. The treatment of metastatic breast cancer is mainly chemotherapy, endocrine therapy and targeted therapy. However, after multiline treatment, patients with MBC especially the triple negative breast cancer face the problem of drug resistance. Tumor angiogenesis theory suggests that blocking angiogenesis can inhibit tumor growth and migration. Based on this, angiogenesis treatment strategy is proposed. Antiangiogenic drugs mainly include biological macromolecular drugs targeting vascular endothelial growth factor (VEGF) or vascular endothelial growth factor receptor (VEGFR) and small molecule VEGFR inhibitors. Angiogenesis is known to play a key role in the growth and metastasis of breast cancer. Therefore, anti-angiogenetic therapy has potential in metastatic breast cancer patients. Since the approval of tumor drug indications by NPMA in China is often later than the release of the latest research data, the National Health Commission issued "the guiding principles for the clinical application of new antitumor drugs" in 2020. The principle pointed out that under special circumstances such as the absence of better treatment, medical institutions should manage the usage of drugs that are not clearly defined in the instructions but have evidence-based data. Based on the latest research progress in breast cancer, the consensus writing expert group collated published reports, international academic conferences, conducted analysis, discussion and summary, collected data on the use of small molecule anti-vascular targeting drugs for advanced breast cancer, and formulated "expert consensus on the application of small molecule anti-angiogenic drugs in the treatment of advanced breast cancer" . For clinicians' reference only.
Angiogenesis Inhibitors/therapeutic use*
;
Breast Neoplasms/pathology*
;
Consensus
;
Female
;
Humans
;
Neovascularization, Pathologic/pathology*
;
Off-Label Use
;
Vascular Endothelial Growth Factor A/metabolism*
9.JAG1 promotes migration, invasion, and adhesion of triple-negative breast cancer cells by promoting angiogenesis.
Jun Ping LIU ; Yu Tong SHI ; Min Min WU ; Meng Qi XU ; Feng Mei ZHANG ; Zhi Qiang HE ; Min TANG
Journal of Southern Medical University 2022;42(7):1100-1108
OBJECTIVE:
To investigate the effect of JAG1 on the malignant phenotype of triple-negative breast cancer (TNBC) and its role in angiogenesis in breast cancer microenvironment.
METHODS:
The expressions of Notch molecules were detected in human TNBC 231 and 231B cells using RT-qPCR. Five female nude mice were inoculated with 231 cells and another 5 with 231B cells into the mammary fat pads, and 4-6 weeks later, the tumors were collected for immunohistochemical and immunofluorescence tests. 231 cells and 231B cells were treated with recombinant JAG (rJAG) protein and DAPT, respectively, and changes in their malignant phenotypes were assessed using CCK-8 assay, Hoechst 33258 staining, wound healing assay, Transwell chamber assay and endothelial cell adhesion assay. Western blotting was used to detect the changes in the expressions of proteins related with the malignant phenotypes of 231 and 231B cells. The effects of conditioned medium (CM) derived from untreated 231 and 231 B cells, rJAG1-treated 231 cells and DAPT-treated 231B cells on proliferation and tube formation ability of cultured human umbilical vein endothelial cells (HUVECs) were evaluated using CCK-8 assay and tube-forming assay.
RESULTS:
The expression of JAG1 was higher in 231B cells than in 231 cells (P < 0.05). Tumor 231B showed higher expression of VEGFA and CD31. Compared with 231-Blank group, the migration, invasion and adhesion of 231 cells in 231-rJAG1 were significantly enhanced (P < 0.05). Protein levels of Twist1 and Snail increased (P < 0.01), anti-apoptotic protein Bcl-2 increased (P < 0.05), while DAPT inhibited the related phenomena and indicators of 231B. The 231-rJAG1-CM increased the cell number and tubule number of HUVEC (P < 0.05).
CONCLUSION
JAG1 may affect the malignant phenotype of TNBC and promote angiogenesis in the tumor microenvironment.
Animals
;
Cell Line, Tumor
;
Cell Movement
;
Cell Proliferation
;
Culture Media, Conditioned
;
Female
;
Human Umbilical Vein Endothelial Cells/metabolism*
;
Humans
;
Jagged-1 Protein/metabolism*
;
Mice
;
Mice, Nude
;
Neovascularization, Pathologic/metabolism*
;
Platelet Aggregation Inhibitors
;
Sincalide/metabolism*
;
Triple Negative Breast Neoplasms/metabolism*
;
Tumor Microenvironment
10.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*

Result Analysis
Print
Save
E-mail