10 results on '"Xian-yi Cai"'
Search Results
2. Supplementary Figure 1 from Rho Kinase Inhibitor Fasudil Suppresses the Vasculogenic Mimicry of B16 Mouse Melanoma Cells Both In Vitro and In Vivo
- Author
-
Gang Wu, Fang Zhu, Rui-Guang Zhang, Zhen-Yu Li, Jing Chen, Jing-Hua Ren, Li-Ling Zhang, Ji-Quan Fan, Xian-Yi Cai, and Yun Xia
- Abstract
B16 cells were seeded in 96-well plates and incubated with various concentrations of fasudil for 24h. The cell inhibitory rate was measured using an MTT assay. Fasudil inhibited B16 cell proliferation in a dose-dependent manner, and the IC50 for 24h of fasudil in B16 cells was 71 {plus minus} 5.8μM.
- Published
- 2023
- Full Text
- View/download PDF
3. Supplementary Figure 2 from Rho Kinase Inhibitor Fasudil Suppresses the Vasculogenic Mimicry of B16 Mouse Melanoma Cells Both In Vitro and In Vivo
- Author
-
Gang Wu, Fang Zhu, Rui-Guang Zhang, Zhen-Yu Li, Jing Chen, Jing-Hua Ren, Li-Ling Zhang, Ji-Quan Fan, Xian-Yi Cai, and Yun Xia
- Abstract
(A) HE staining of tissue section. A vessel formed with elongated endothelial cells was showed by a red arrow, moreover, the vessel lack of endothelial cells nearby was pointed out by a yellow arrow. Original magnification, ×400. (B) Plenty of blood sinusoids were observed in the CD31/PAS dual-stained sections. Original magnification, ×100. (C) Endothelial cell lined-blood vessels containing CD31-positive immunoreactivity (dark brown) . (D) A VM structure were strictly defined as PAS-positive (purple magenta) CD31-negative. Detached tumor cells (black arrows) reached the blood stream, and it is suggested that VM networks facilitate tumor metastasis. Original magnification, ×400.
- Published
- 2023
- Full Text
- View/download PDF
4. Digoxin targets low density lipoprotein receptor-related protein 4 and protects against osteoarthritis
- Author
-
Zhe-Sheng Chen, Nan Jiang, Aubryanna Hettinghouse, Zi-Ning Lei, Asya Khleborodova, Guanghua Lei, Jing Wu, Chuan-ju Liu, Xiang Ding, Chao Zeng, Kaidi Wang, and Xian-yi Cai
- Subjects
Drug ,Cartilage, Articular ,Digoxin ,Anabolism ,media_common.quotation_subject ,Immunology ,Osteoarthritis ,Pharmacology ,General Biochemistry, Genetics and Molecular Biology ,Chondrocyte ,Article ,Cohort Studies ,Chondrocytes ,Rheumatology ,medicine ,polycyclic compounds ,Immunology and Allergy ,Humans ,cardiovascular diseases ,Ouabain ,LDL-Receptor Related Proteins ,media_common ,Cartilage homeostasis ,Catabolism ,business.industry ,digestive, oral, and skin physiology ,Drug Repositioning ,medicine.disease ,carbohydrates (lipids) ,medicine.anatomical_structure ,LDL receptor ,business ,medicine.drug - Abstract
ObjectivesDysregulated chondrocyte metabolism is closely associated with the pathogenesis of osteoarthritis (OA). Suppressing chondrocyte catabolism to restore cartilage homeostasis has been extensively explored, whereas far less effort has been invested toward enhancing chondrocyte anabolism. This study aimed to repurpose clinically approved drugs as potential stimulators of chondrocyte anabolism in treating OA.MethodsScreening of a Food and Drug Administration-approved drug library; Assays for examining the chondroprotective effects of digoxin in vitro; Assays for defining the therapeutic effects of digoxin using a surgically-induced OA model; A propensity-score matched cohort study using The Health Improvement Network to examine the relationship between digoxin use and the risk of joint OA-associated replacement among patients with atrial fibrillation; identification and characterisation of the binding of digoxin to low-density lipoprotein receptor-related protein 4 (LRP4); various assays, including use of CRISPR-Cas9 genome editing to delete LRP4 in human chondrocytes, for examining the dependence on LRP4 of digoxin regulation of chondrocytes.ResultsSerial screenings led to the identification of ouabain and digoxin as stimulators of chondrocyte differentiation and anabolism. Ouabain and digoxin protected against OA and relieved OA-associated pain. The cohort study of 56 794 patients revealed that digoxin use was associated with reduced risk of OA-associated joint replacement. LRP4 was isolated as a novel target of digoxin, and deletion of LRP4 abolished digoxin’s regulations of chondrocytes.ConclusionsThese findings not only provide new insights into the understanding of digoxin’s chondroprotective action and underlying mechanisms, but also present new evidence for repurposing digoxin for OA.
- Published
- 2021
5. Catecholamines contribute to the neovascularization of lung cancer via tumor-associated macrophages
- Author
-
Ye Wei, Ruiguang Zhang, Fang Zhu, Liling Zhang, Rui Meng, Zhenyu Li, Xiaorong Dong, Sheng Zhang, Yun Xia, Gang Wu, and Xian-Yi Cai
- Subjects
Male ,0301 basic medicine ,Lung Neoplasms ,Adrenergic receptor ,Angiogenesis ,medicine.medical_treatment ,Immunology ,Macrophage polarization ,Neovascularization ,Mice ,03 medical and health sciences ,Behavioral Neuroscience ,Catecholamines ,0302 clinical medicine ,Cell Movement ,Cell Line, Tumor ,Tumor Microenvironment ,medicine ,Animals ,Humans ,Tube formation ,Mice, Inbred BALB C ,Tumor microenvironment ,Neovascularization, Pathologic ,Endocrine and Autonomic Systems ,Chemistry ,Macrophages ,Macrophage Activation ,M2 Macrophage ,030104 developmental biology ,Cytokine ,Cancer research ,Cytokines ,medicine.symptom ,030217 neurology & neurosurgery ,Signal Transduction - Abstract
Purpose Elevated catecholamines in the tumor microenvironment often correlate with tumor development. However, the mechanisms by which catecholamines modulate lung cancer growth are still poorly understood. This study is aimed at examining the functions and mechanisms of catecholamine-induced macrophage polarization in angiogenesis and tumor development. Experimental design We established in vitro and in vivo models to investigate the relationship between catecholamines and macrophages in lung cancer. Flow cytometry, cytokine detection, tube formation assay, immunofluorescence, and western blot analysis were performed, and animal models were also used to explore the underlying mechanism of catecholamine-induced macrophage polarization and host immunological response. Results Catecholamines were shown to be secreted into tumor under the control of the sympathetic nerve system to maintain the pro-tumoral microenvironment. In vivo, the chemical depletion of the natural catecholamine stock with 6OHDA could reduce the release of catecholamines within tumor tissues, restrain the function of alternatively activated M2 macrophage, attenuate tumor neovascularization, and inhibit tumor growth. In vitro, catecholamine treatment triggered the M2 polarization of macrophages, enhanced the expression of VEGF, promoted tumor angiogenesis, and these catecholamine-stimulated effects could be reversed by the adrenergic receptor antagonist propranolol. In addition to regulating tumor-associated macrophages (TAM) recruitment, decreasing catecholamine levels could also shift the immunosuppressive microenvironment by decreasing myeloid-derived suppressor cells’ (MDSCs) recruitment and facilitating dendritic cells’ (DCs) activation, potentially resulting in a positive antitumor immune response. Conclusion Our study demonstrates the potential of adrenergic stress and catecholamine-driven adrenergic signaling of TAMs to regulate the immune status of a tumor microenvironment and provides promising targets for anticancer therapies.
- Published
- 2019
- Full Text
- View/download PDF
6. The role of sema4D in vasculogenic mimicry formation in non-small cell lung cancer and the underlying mechanisms
- Author
-
Jinghua Ren, Fang Zhu, Gang Wu, Ji-Quan Fan, Xian-Yi Cai, Yun Xia, Ruiguang Zhang, Liling Zhang, and Zhenyu Li
- Subjects
Cancer Research ,RHOA ,biology ,Chemistry ,SEMA4D ,Motility ,Cancer ,medicine.disease ,Metastasis ,03 medical and health sciences ,0302 clinical medicine ,Oncology ,030220 oncology & carcinogenesis ,medicine ,Cancer research ,biology.protein ,Vasculogenic mimicry ,Small GTPase ,Signal transduction - Abstract
Vasculogenic mimicry (VM) is a special vascular pattern in malignant tumors, which is composed of highly aggressive tumor cells. This tumor cell-mediated blood supply pattern is closely associated with a poor prognosis in cancer patients. The interaction of axon guidance factor Sema4D and its high affinity receptor plexinB1 could activate small GTPase RhoA and its downstream ROCKs; this process has an active role in the migration of endothelial cells and tumor angiogenesis. Here, we have begun to uncover the role of this pathway in VM formation in non-small cell lung cancer (NSCLC). First, we confirmed this special form of vasculature in NSCLC tissues and found the existence of VM channels in tumor tissues was correlated with Sema4D expression. Further, we found that inhibition of Sema4D in the human NSCLC cells H1299 and HCC827 reduces VM formation both in vitro and in vivo. Moreover, we demonstrated that downregulating the expression of plexinB1 by siRNA expressing vectors and inhibiting the RhoA/ROCK signaling pathway using fasudil can reduce VM formation of H1299 and HCC827 cells. Finally, we found that suppression of Sema4D leads to less stress fibers and depleted the motility of H1299 and HCC827 cells. Collectively, our study implicates Sema4D plays an important role in the process of VM formation in NSCLC through activating the RhoA/ROCK pathway and regulating tumor cell plasticity and migration. Modulation of the Sema4D/plexinB1 and downstream RhoA/ROCK pathway may prevent the tumor blood supply through the VM pattern, which may eventually halt growth and metastasis of NSCLC.
- Published
- 2018
- Full Text
- View/download PDF
7. Digoxin targets low density lipoprotein receptor-related protein 4 and protects against osteoarthritis.
- Author
-
Kai-di Wang, Xiang Ding, Nan Jiang, Chao Zeng, Jing Wu, Xian-yi Cai, Hettinghouse, Aubryanna, Khleborodova, Asya, Zi-Ning Lei, Zhe-Sheng Chen, Guang-hua Lei, Chuan-ju Liu, Wang, Kai-di, Ding, Xiang, Jiang, Nan, Zeng, Chao, Wu, Jing, Cai, Xian-Yi, Lei, Zi-Ning, and Chen, Zhe-Sheng
- Subjects
DIGOXIN ,CARTILAGE cells ,CARDIAC glycosides ,CELL receptors ,OSTEOARTHRITIS ,RESEARCH funding ,ARTICULAR cartilage ,LONGITUDINAL method ,PHARMACODYNAMICS - Abstract
Objectives: Dysregulated chondrocyte metabolism is closely associated with the pathogenesis of osteoarthritis (OA). Suppressing chondrocyte catabolism to restore cartilage homeostasis has been extensively explored, whereas far less effort has been invested toward enhancing chondrocyte anabolism. This study aimed to repurpose clinically approved drugs as potential stimulators of chondrocyte anabolism in treating OA.Methods: Screening of a Food and Drug Administration-approved drug library; Assays for examining the chondroprotective effects of digoxin in vitro; Assays for defining the therapeutic effects of digoxin using a surgically-induced OA model; A propensity-score matched cohort study using The Health Improvement Network to examine the relationship between digoxin use and the risk of joint OA-associated replacement among patients with atrial fibrillation; identification and characterisation of the binding of digoxin to low-density lipoprotein receptor-related protein 4 (LRP4); various assays, including use of CRISPR-Cas9 genome editing to delete LRP4 in human chondrocytes, for examining the dependence on LRP4 of digoxin regulation of chondrocytes.Results: Serial screenings led to the identification of ouabain and digoxin as stimulators of chondrocyte differentiation and anabolism. Ouabain and digoxin protected against OA and relieved OA-associated pain. The cohort study of 56 794 patients revealed that digoxin use was associated with reduced risk of OA-associated joint replacement. LRP4 was isolated as a novel target of digoxin, and deletion of LRP4 abolished digoxin's regulations of chondrocytes.Conclusions: These findings not only provide new insights into the understanding of digoxin's chondroprotective action and underlying mechanisms, but also present new evidence for repurposing digoxin for OA. [ABSTRACT FROM AUTHOR]- Published
- 2022
- Full Text
- View/download PDF
8. The role of sema4D in vasculogenic mimicry formation in non-small cell lung cancer and the underlying mechanisms
- Author
-
Yun, Xia, Xian-Yi, Cai, Ji-Quan, Fan, Li-Ling, Zhang, Jing-Hua, Ren, Zhen-Yu, Li, Rui-Guang, Zhang, Fang, Zhu, and Gang, Wu
- Subjects
Mice, Inbred BALB C ,rho-Associated Kinases ,Lung Neoplasms ,Neovascularization, Pathologic ,Endothelial Cells ,Mice, Nude ,Nerve Tissue Proteins ,Receptors, Cell Surface ,Semaphorins ,Enzyme Activation ,Mice ,A549 Cells ,Antigens, CD ,Cell Movement ,1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,Animals ,Humans ,RNA Interference ,RNA, Small Interfering ,rhoA GTP-Binding Protein ,Protein Kinase Inhibitors - Abstract
Vasculogenic mimicry (VM) is a special vascular pattern in malignant tumors, which is composed of highly aggressive tumor cells. This tumor cell-mediated blood supply pattern is closely associated with a poor prognosis in cancer patients. The interaction of axon guidance factor Sema4D and its high affinity receptor plexinB1 could activate small GTPase RhoA and its downstream ROCKs; this process has an active role in the migration of endothelial cells and tumor angiogenesis. Here, we have begun to uncover the role of this pathway in VM formation in non-small cell lung cancer (NSCLC). First, we confirmed this special form of vasculature in NSCLC tissues and found the existence of VM channels in tumor tissues was correlated with Sema4D expression. Further, we found that inhibition of Sema4D in the human NSCLC cells H1299 and HCC827 reduces VM formation both in vitro and in vivo. Moreover, we demonstrated that downregulating the expression of plexinB1 by siRNA expressing vectors and inhibiting the RhoA/ROCK signaling pathway using fasudil can reduce VM formation of H1299 and HCC827 cells. Finally, we found that suppression of Sema4D leads to less stress fibers and depleted the motility of H1299 and HCC827 cells. Collectively, our study implicates Sema4D plays an important role in the process of VM formation in NSCLC through activating the RhoA/ROCK pathway and regulating tumor cell plasticity and migration. Modulation of the Sema4D/plexinB1 and downstream RhoA/ROCK pathway may prevent the tumor blood supply through the VM pattern, which may eventually halt growth and metastasis of NSCLC.
- Published
- 2018
9. RhoA/ROCK pathway inhibition by fasudil suppresses the vasculogenic mimicry of U2OS osteosarcoma cells in vitro
- Author
-
Zhenyu Li, Ji-Quan Fan, Gang Wu, Xian-Yi Cai, Fang Zhu, Liling Zhang, Yun Xia, and Jinghua Ren
- Subjects
0301 basic medicine ,Cancer Research ,RHOA ,Angiogenesis ,Cell Plasticity ,Fluorescent Antibody Technique ,Bone Neoplasms ,Biology ,03 medical and health sciences ,0302 clinical medicine ,Cell Movement ,1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine ,Cell Line, Tumor ,Humans ,Pharmacology (medical) ,ROCK1 ,Vasculogenic mimicry ,Protein Kinase Inhibitors ,Cytoskeleton ,Cell Proliferation ,Pharmacology ,Matrigel ,Osteosarcoma ,rho-Associated Kinases ,Neovascularization, Pathologic ,Kinase ,Fasudil ,Cell biology ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,biology.protein ,Signal transduction ,rhoA GTP-Binding Protein ,Signal Transduction - Abstract
GTPase RhoA and its downstream Rho-associated coiled-coil-containing protein kinases (ROCKs) are frequently overexpressed in human cancers. Inhibition of the RhoA/ROCK pathway blocks angiogenesis mediated by the vascular endothelial growth factor, which led us to investigate the role of this pathway in vasculogenic mimicry (VM) - a process by which aggressive cancer cells form vessel-like structures that provide adequate blood supply for tumor growth. We showed that the expression of RhoA and its effector kinases ROCK1/2 was much higher in human osteosarcoma (OS) tissues and the human OS cell line U2OS than in nontumorous tissues and cell line hFOB 1.19 using western blot analysis and real-time PCR. Inhibition of the RhoA/ROCK signaling pathway by the pharmacological inhibitor fasudil reduced vascular-like channels of U2OS cells in Matrigel. Furthermore, we used rhodamine-phalloidin immunofluorescence, wound healing assay, and transwell migration assay to examine the effect of fasudil on tumor cell plasticity and motility, both of which play key roles in VM formation. Finally, we explored the underlying mechanisms of fasudil-induced VM destruction. In this context, we showed that the RhoA/ROCK signaling pathway is a novel regulator in VM of U2OS OS cells and suggest that fasudil in conjunction with established treatments may present a novel therapeutic strategy for OS.
- Published
- 2017
10. Rho Kinase Inhibitor Fasudil Suppresses the Vasculogenic Mimicry of B16 Mouse Melanoma Cells Both In Vitro and In Vivo
- Author
-
Yun Xia, Fang Zhu, Zhenyu Li, Jinghua Ren, Xian-Yi Cai, Jing Chen, Gang Wu, Ruiguang Zhang, Liling Zhang, and Ji-Quan Fan
- Subjects
Male ,Cancer Research ,RHOA ,Angiogenesis ,Blotting, Western ,Melanoma, Experimental ,Pharmacology ,Cell Movement ,1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine ,Cell Line, Tumor ,Animals ,Humans ,Vasculogenic mimicry ,Protein Kinase Inhibitors ,Cell Proliferation ,Matrigel ,rho-Associated Kinases ,Microscopy, Confocal ,biology ,Neovascularization, Pathologic ,Reverse Transcriptase Polymerase Chain Reaction ,Fasudil ,Cell migration ,Tumor Burden ,Endothelial stem cell ,Mice, Inbred C57BL ,Oncology ,Rho kinase inhibitor ,biology.protein ,Cancer research ,Lysophospholipids ,Neoplasm Transplantation - Abstract
The aim of this study was to investigate the biologic role of the Rho kinase inhibitor fasudil in the vasculogenic mimicry (VM) of B16 mouse melanoma cells. It was previously reported that RhoA plays a critical role in angiogenesis by coordinating endothelial cell cytoskeleton remodeling and promoting endothelial cell motility. Although RhoA has been implicated in the regulation of angiogenesis, little has been described regarding its control of these tumor cell–lined channels. In this study, we established an in vitro model of VM using 3-dimensional cell culturing of mouse B16 melanoma cells and studied VM in vivo by transplanting B16 cells into C57/BL mice. Next, we explored the effect of RhoA and Rho-associated, coiled-coil containing protein kinase (ROCK) on VM formation using the Rho kinase inhibitor fasudil. We provide direct evidence that fasudil leads to reduced vascular-like channels in Matrigel. Additional experiments suggested that fasudil prevents both initial cellular architecture changes and cell migration in vitro. Finally, we provide in-depth evidence for the underlying mechanisms of fasudil-induced VM destruction using the Rho-GTPase agonist lysophosphatidic acid. In vivo studies revealed that fasudil reduced B16 melanoma cell xenograft tumor growth without causing significant toxicity in mice. Fasudil-treated tumors also displayed fewer VM channels. These results suggest that fasudil may be an emerging therapeutic option for targeting cancer VM. Mol Cancer Ther; 14(7); 1582–90. ©2015 AACR.
- Published
- 2014
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.