1
|
Chen Y, Wang L, Liu X, Wang F, An Y, Zhao W, Tian J, Kong D, Zhang W, Xu Y, Ba Y, Zhou H. The Genus Broussonetia: An Updated Review of Phytochemistry, Pharmacology and Applications. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27165344. [PMID: 36014582 PMCID: PMC9414938 DOI: 10.3390/molecules27165344] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/15/2022] [Accepted: 08/18/2022] [Indexed: 02/05/2023]
Abstract
The Broussonetia genus (Moraceae), recognized for its value in many Chinese traditional herbs, mainly includes Broussonetia papyrifera (L.) L’Hér. ex Vent. (BP), Broussonetia kazinoki Siebold (BK), and Broussonetia luzonica (Blanco) Bureau (BL). Hitherto, researchers have found 338 compounds isolated from BP, BK, and BL, which included flavonoids, polyphenols, phenylpropanoids, alkaloids, terpenoids, steroids, and others. Moreover, its active compounds and extracts have exhibited a variety of pharmacological effects such as antitumor, antioxidant, anti-inflammatory, antidiabetic, anti-obesity, antibacterial, and antiviral properties, and its use against skin wrinkles. In this review, the phytochemistry and pharmacology of Broussonetia are updated systematically, after its applications are first summarized. In addition, this review also discusses the limitations of investigations and the potential direction of Broussonetia. This review can help to further understand the phytochemistry, pharmacology, and other applications of Broussonetia, which paves the way for future research.
Collapse
|
2
|
Kim JH, Kim S, Han S, Ahn EK, Cho YR, Jeong W, Kim SJ, Bae GU, Oh JS, Seo DW. Broussonin A- and B-mediated inhibition of angiogenesis by blockade of VEGFR-2 signalling pathways and integrin β1 expression. J Cell Mol Med 2022; 26:1194-1205. [PMID: 34994065 PMCID: PMC8831976 DOI: 10.1111/jcmm.17173] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 12/27/2021] [Accepted: 12/29/2021] [Indexed: 11/28/2022] Open
Abstract
In the present study, we demonstrate the regulatory effects and mechanism of broussonin A and B, diphenylpropane derivatives isolated from Broussonetia kazinoki, on vascular endothelial growth factor‐A (VEGF‐A)–stimulated endothelial cell responses in vitro and microvessel sprouting ex vivo. Treatment with broussonin A or B suppressed VEGF‐A‐stimulated endothelial cell proliferation by regulating the expression of cell cycle–related proteins and the phosphorylation status of retinoblastoma protein. In addition, treatment with broussonin A or B abrogated VEGF‐A‐stimulated angiogenic responses including endothelial cell migration, invasion, tube formation and microvessel formation from rat aortic rings. These anti‐angiogenic activities of broussonin A and B were mediated through inactivation of VEGF‐A‐stimulated downstream signalling pathways, localization of vascular endothelial‐cadherin at cell‐cell contacts, and down‐regulation of integrin β1 and integrin‐liked kinase. Furthermore, treatment with broussonin A or B inhibited proliferation and invasion of non–small cell lung cancer and ovarian cancer cells. Taken together, our findings suggest the pharmacological potential of broussonin A and B in the regulation of angiogenesis, cancer cell growth and progression.
Collapse
Affiliation(s)
- Jae Hyeon Kim
- Department of Pharmacy, College of Pharmacy, Dankook University, Cheonan, Republic of Korea
| | - Sunho Kim
- Department of Pharmacy, College of Pharmacy, Dankook University, Cheonan, Republic of Korea
| | - Surim Han
- Department of Pharmacy, College of Pharmacy, Dankook University, Cheonan, Republic of Korea
| | - Eun-Kyung Ahn
- Biocenter, Gyeonggi Business & Science Accelerator, Suwon, Republic of Korea
| | - Young-Rak Cho
- Biocenter, Gyeonggi Business & Science Accelerator, Suwon, Republic of Korea
| | - Wonsik Jeong
- Biocenter, Gyeonggi Business & Science Accelerator, Suwon, Republic of Korea
| | - Sung Joon Kim
- Department of Pharmacy, College of Pharmacy, Dankook University, Cheonan, Republic of Korea
| | - Gyu-Un Bae
- Department of Pharmacy, College of Pharmacy, Sookmyung Women's University, Seoul, Republic of Korea
| | - Joa Sub Oh
- Department of Pharmacy, College of Pharmacy, Dankook University, Cheonan, Republic of Korea
| | - Dong-Wan Seo
- Department of Pharmacy, College of Pharmacy, Dankook University, Cheonan, Republic of Korea
| |
Collapse
|
3
|
Protective Effects of Broussonetia kazinoki Siebold Fruit Extract against Palmitate-Induced Lipotoxicity in Mesangial Cells. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:4509403. [PMID: 30728845 PMCID: PMC6341277 DOI: 10.1155/2019/4509403] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 12/19/2018] [Accepted: 12/25/2018] [Indexed: 01/06/2023]
Abstract
Diabetic nephropathy is one of the most serious complications of diabetes. Lipotoxicity in glomerular mesangial cells is associated with the progression of diabetic nephropathy. Paper mulberry, Broussonetia kazinoki Siebold (BK), has been used in oriental medicine for human health problems. However, to date, the beneficial effect of BK fruit has not been studied. In this study, we investigated the protective effect of an ethanolic extract of BK fruit (BKFE) against palmitate- (PA-) induced toxicity in mesangial cells. BKFE significantly increased the viability of PA-treated SV40 MES13 cells. BKFE significantly inhibited PA-induced apoptosis and decreased the expression of apoptotic genes, cleaved caspase-3, and cleaved PARP. Moreover, BKFE inhibited the expression of endoplasmic reticulum (ER) stress-related genes, such as BiP, phosphorylated eIF2α, cleaved ATF6, and spliced XBP-1, in PA-treated SV40 MES13 cells. BKFE decreased PA-induced ROS production. In addition, BKFE activated the transcription factor Nrf2 and increased the expression of antioxidant enzymes. However, knockdown of Nrf2 using siRNA suppressed this BKFE-induced increase in antioxidant enzyme expression. Furthermore, the protective effect of BKFE on PA-induced apoptosis was significantly reduced by Nrf2 knockdown. In conclusion, BKFE induced the expression of antioxidant enzymes via activation of Nrf2 and protected against PA-induced lipotoxicity in mesangial cells.
Collapse
|
4
|
Cho YR, Park K, Kang JS, Byun HW, Oh JS, Seo DW, Ahn EK. Trigonostemon reidioides modulates endothelial cell proliferation, migration and tube formation via downregulation of the Akt signaling pathway. Oncol Lett 2017; 14:4677-4683. [PMID: 29085467 PMCID: PMC5649608 DOI: 10.3892/ol.2017.6760] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 03/07/2017] [Indexed: 12/15/2022] Open
Abstract
Trigonostemon reidi`oides (TR) is used as a Thai traditional medicine for the treatment of drug addiction, asthma, food poisoning, constipation and snake bites. The present study investigated the effects and molecular mechanisms of the ethanolic extract of TR (ETR) on mitogen-induced human umbilical vein endothelial cells (HUVECs) responses, proliferation, adhesion, migration and tube formation. ETR treatment inhibited mitogen-induced HUVEC proliferation by downregulation of cell cycle-associated proteins, including cyclins and cyclin-dependent kinases, which induced retinoblastoma protein hypophosphorylation. The present study also demonstrated that ETR treatment suppressed mitogen-induced HUVEC adhesion, migration, invasion and tube formation, and that these anti-angiogenic activities were mediated by inactivation of mitogen-induced Akt and matrix metalloproteinase (MMP)-2, but not of extracellular signal-regulated kinase, p70 ribosomal S6 kinase or MMP-9. Collectively, the results of the present study suggested pharmacological functions and molecular mechanisms of ETR in regulating endothelial cell fates, and supported further evaluation and development of ETR as a potential therapeutic agent for the treatment and prevention of angiogenesis-associated diseases, including cancer.
Collapse
Affiliation(s)
- Young-Rak Cho
- Bio-Center, Gyeonggi Institute of Science and Technology Promotion, Suwon, Gyeonggi 16229, Republic of Korea
| | - Kyuhee Park
- Bio-Center, Gyeonggi Institute of Science and Technology Promotion, Suwon, Gyeonggi 16229, Republic of Korea
| | - Jae-Shin Kang
- Biological Genetic Resources Utilization Division, National Institute of Biological Resources, Seo, Incheon 22689, Republic of Korea
| | - Hye-Woo Byun
- Biological Genetic Resources Utilization Division, National Institute of Biological Resources, Seo, Incheon 22689, Republic of Korea
| | - Joa Sub Oh
- Department of Pharmacy, College of Pharmacy, Dankook University, Cheonan, Chungcheong 31116, Republic of Korea
| | - Dong-Wan Seo
- Department of Pharmacy, College of Pharmacy, Dankook University, Cheonan, Chungcheong 31116, Republic of Korea
| | - Eun-Kyung Ahn
- Bio-Center, Gyeonggi Institute of Science and Technology Promotion, Suwon, Gyeonggi 16229, Republic of Korea
| |
Collapse
|
5
|
Wang JQ, Kou JF, Zhao ZZ, Qiu KQ, Chao H. Anthraquinone-bridged diruthenium(ii) complexes inhibit migration and invasion of human hepatocarcinoma MHCC97-H cells. Inorg Chem Front 2017. [DOI: 10.1039/c7qi00149e] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Four diruthenium(ii) complexes exhibited anti-metastatic properties on MHCC97-H cells, which involved in the inhibition of migration and invasion, negative remodulation of the cytoskeleton, blocking cell cycles and regulation of relative signal pathways.
Collapse
Affiliation(s)
- Jin-Quan Wang
- Guangdong Provincial Key Laboratory of Biotechnology Candidate Drug Research
- Guangdong Pharmaceutical University
- Guangzhou 510275
- China
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry
| | - Jun-Feng Kou
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry
- School of Chemistry
- Sun Yat-Sen University
- Guangzhou 510275
- China
| | - Zi-Zhuo Zhao
- Sun Yat-sen Memorial Hospital
- Sun Yat-Sen University
- Guangzhou
- China
| | - Kang-Qiang Qiu
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry
- School of Chemistry
- Sun Yat-Sen University
- Guangzhou 510275
- China
| | - Hui Chao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry
- School of Chemistry
- Sun Yat-Sen University
- Guangzhou 510275
- China
| |
Collapse
|
6
|
Kim JH, Kim MS, Lee BH, Kim JK, Ahn EK, Ko HJ, Cho YR, Lee SJ, Bae GU, Kim YK, Oh JS, Seo DW. Marmesin-mediated suppression of VEGF/VEGFR and integrin β1 expression: Its implication in non-small cell lung cancer cell responses and tumor angiogenesis. Oncol Rep 2016; 37:91-97. [DOI: 10.3892/or.2016.5245] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 06/29/2016] [Indexed: 11/06/2022] Open
|
7
|
Hwang-Bo J, Park JH, Bae MG, Chung IS. Recombinant canstatin inhibits VEGF-A-induced lymphangiogenesis and metastasis in an oral squamous cell carcinoma SCC-VII animal model. Cancer Med 2016; 5:2977-2988. [PMID: 27650585 PMCID: PMC5083751 DOI: 10.1002/cam4.866] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 06/22/2016] [Accepted: 07/24/2016] [Indexed: 01/30/2023] Open
Abstract
We describe the inhibitory effects of recombinant canstatin on tumor growth and lymphangiogenesis induced by an oral squamous cell carcinoma (SCC) using an orthotropic oral SCC animal model. Recombinant canstatin treatment decreased final tumor volumes and weights, as well as densities of blood and lymphatic vessels. Lung metastasis of oral SCC was significantly reduced in recombinant canstatin‐treated animals. Recombinant canstatin reduced vascular endothelial growth factor (VEGF)‐A expression in SCC‐VII cells treated with the hypoxia mimetic agent, CoCl2. VEGF‐A induced in vivo lymphatic vessel formation in a Matrigel plug, but this was remarkably reduced in a recombinant canstatin‐treated Matrigel. Recombinant canstatin suppressed the expression of vascular endothelial growth factor receptors (VEGFR)‐1 and ‐2 stimulated by VEGF‐A. Based on immunohistochemical analysis, recombinant canstatin significantly reduced the expression of VEGF‐A, VEGFR‐1, and ‐2 in SCC‐VII‐induced tumors. Recombinant canstatin did not affect the expression of VEGF‐C or VEGFR‐3. In addition, recombinant canstatin suppressed the VEGF‐A‐induced phosphorylation of VEGFR‐1 and ‐2. Our results indicate that recombinant canstatin exhibits antitumoral and antilymphangiogenic activities against oral SCC cells. Antilymphangiogenic signaling by recombinant canstatin is probably mediated by the suppression of the integrin αvβ3/VEGFR‐1 and/or ‐2 signaling induced by VEGF‐A. Our results also suggest that recombinant canstatin has a high potential to inhibit oral SCC‐induced tumors and lymphatic metastasis.
Collapse
Affiliation(s)
- Jeon Hwang-Bo
- Department of Genetic Engineering and Graduate School of Biotechnology, Kyung Hee University, Yongin, 17104, Korea
| | - Jong-Hwa Park
- Department of Genetic Engineering and Graduate School of Biotechnology, Kyung Hee University, Yongin, 17104, Korea
| | - Mun Gyeong Bae
- Department of Genetic Engineering and Graduate School of Biotechnology, Kyung Hee University, Yongin, 17104, Korea
| | - In Sik Chung
- Department of Genetic Engineering and Graduate School of Biotechnology, Kyung Hee University, Yongin, 17104, Korea.
| |
Collapse
|
8
|
Lee H, Li H, Jeong JH, Noh M, Ryu JH. Kazinol B from Broussonetia kazinoki improves insulin sensitivity via Akt and AMPK activation in 3T3-L1 adipocytes. Fitoterapia 2016; 112:90-6. [DOI: 10.1016/j.fitote.2016.05.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 05/13/2016] [Accepted: 05/16/2016] [Indexed: 11/26/2022]
|
9
|
Gao F, Sun M, Gong Y, Wang H, Wang Y, Hou H. MicroRNA-195a-3p inhibits angiogenesis by targeting Mmp2 in murine mesenchymal stem cells. Mol Reprod Dev 2016; 83:413-23. [PMID: 26989874 DOI: 10.1002/mrd.22638] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 03/10/2016] [Indexed: 12/25/2022]
Abstract
MicroRNAs (miRNAs) modulate complex physiological and pathological processes, including the regulation of angiogenesis. Our previous study reported that bone marrow-derived mesenchymal stem cells (MSCs) are recruited into choroidal neovascularization lesions. miRNA-195 is highly expressed in MSCs, but its function remains unknown. In the present study, miR-195a-3p abundance was significantly decreased in hypoxia-treated murine MSCs; on the other hand, its overexpression reduced MSC proliferation and migration while increasing the activation of anti-angiogenic factor pigment epithelium-derived factor (PEDF). We further discovered that matrix metalloproteinase 2 (Mmp2) transcript is a target of miR-195a-3p, and that silencing Mmp2 phenocopied the reduced proliferation and migration of MSCs. The therapeutic potential of miR-195a-3p as an angiogenesis inhibitor was also demonstrated in a laser-induced choroidal neovascularization mouse model. These findings collectively indicate that miR-195a-3p is a negative modulator of angiogenesis, and could be used as an angiogenesis inhibitor. Mol. Reprod. Dev. 83: 413-423, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Fan Gao
- Department of Ophthalmology, Eye Institute of Chinese PLA, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Meng Sun
- Department of Cardiology, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Yumei Gong
- Department of Cardiovascular Surgery, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Haiyan Wang
- Department of Ophthalmology, Eye Institute of Chinese PLA, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yusheng Wang
- Department of Ophthalmology, Eye Institute of Chinese PLA, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Huiyuan Hou
- Department of Ophthalmology, Eye Institute of Chinese PLA, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| |
Collapse
|
10
|
In JK, Kim JK, Oh JS, Seo DW. 5-Caffeoylquinic acid inhibits invasion of non-small cell lung cancer cells through the inactivation of p70S6K and Akt activity: Involvement of p53 in differential regulation of signaling pathways. Int J Oncol 2016; 48:1907-12. [PMID: 26984670 DOI: 10.3892/ijo.2016.3436] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 02/24/2016] [Indexed: 11/06/2022] Open
Abstract
In the present study, we investigated the effects and molecular mechanism of 5-caffeoylquinic acid (5-CQA), a natural phenolic compound isolated from Ligularia fischeri, on cell invasion, proliferation and adhesion in p53 wild-type A549 and p53-deficient H1299 non-small cell lung cancer (NSCLC) cells. 5-CQA abrogated mitogen-stimulated invasion, but not proliferation, in both A549 and H1299 cells. In addition, 5-CQA inhibited mitogen-stimulated adhesion in A549 cells only. Anti-invasive activity of 5-CQA in A549 cells was mediated by the inactivation of p70(S6K)-dependent signaling pathway. In contrast, in H1299 cells the inactivation of Akt was found to be involved in 5-CQA-mediated inhibition of cell invasion. Collectively, these findings demonstrate the pharmacological roles and molecular targets of 5-CQA in regulating NSCLC cell fate, and suggest further evaluation and development of 5-CQA as a potential therapeutic agent for the treatment and prevention of lung cancer.
Collapse
Affiliation(s)
- Jae-Kyung In
- Department of Pharmacy, College of Pharmacy, Dankook University, Cheonan 31116, Republic of Korea
| | - Jin-Kyu Kim
- Biocenter, Gyeonggi Institute of Science and Technology Promotion, Suwon 16229, Republic of Korea
| | - Joa Sub Oh
- Department of Pharmacy, College of Pharmacy, Dankook University, Cheonan 31116, Republic of Korea
| | - Dong-Wan Seo
- Department of Pharmacy, College of Pharmacy, Dankook University, Cheonan 31116, Republic of Korea
| |
Collapse
|
11
|
Kim JH, Kim JK, Ahn EK, Ko HJ, Cho YR, Lee CH, Kim YK, Bae GU, Oh JS, Seo DW. Marmesin is a novel angiogenesis inhibitor: Regulatory effect and molecular mechanism on endothelial cell fate and angiogenesis. Cancer Lett 2015; 369:323-30. [DOI: 10.1016/j.canlet.2015.09.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 09/16/2015] [Accepted: 09/18/2015] [Indexed: 12/27/2022]
|
12
|
JOO JIHYE, HONG SEONGSU, CHO YOUNGRAK, SEO DONGWAN. 10-Gingerol inhibits proliferation and invasion of MDA-MB-231 breast cancer cells through suppression of Akt and p38MAPK activity. Oncol Rep 2015; 35:779-84. [DOI: 10.3892/or.2015.4405] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 10/05/2015] [Indexed: 11/06/2022] Open
|
13
|
Wang W, Wang HJ, Wang B, Li Y, Qin Y, Zheng LS, Zhou JS, Qu PH, Shi JH, Zhang HS. The Role of the Y Box Binding Protein 1 C-Terminal Domain in Vascular Endothelial Cell Proliferation, Apoptosis, and Angiogenesis. DNA Cell Biol 2015; 35:24-32. [PMID: 26430912 DOI: 10.1089/dna.2015.2908] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Different domains of the multifunctional transcription factor Y-box binding protein 1 (YB1) regulate proliferation, differentiation, and apoptosis by transactivating or repressing the promoters of various genes. Here we report that the C-terminal domain of YB1 (YB1 CTD) is involved in endothelial cell proliferation, apoptosis, and tube formation. The oligo pull-down assays demonstrated that YB1 directly binds double-stranded GC box sequences in endothelial cells through the 125-220 amino acids. Adenovirus expression vectors harboring green fluorescent protein (GFP) or GFP-tagged YB1 CTD were constructed and used to infect EA.hy926 endothelial cells. Overexpression of the YB1 CTD significantly increased p21 expression, decreased cyclin B1 expression, and inhibited the proliferation of EA.hy926 cells. YB1 CTD overexpression also increased Bax and active caspase 3 expression, decreased Bcl-2 expression, and induced apoptosis in EA.hy926 cells. Furthermore, overexpression of the YB1 CTD significantly suppressed migration and tube formation in EA.hy926 cells. Finally, YB1 CTD decreased ERK1/2 phosphorylation in EA.hy926 cells. These findings demonstrated vital roles for YB1 in endothelial cell proliferation, apoptosis, and tube formation through transcriptional regulation of GC box-related genes.
Collapse
Affiliation(s)
- Wei Wang
- 1 Central Laboratory, Affiliated Hospital of Hebei University , Baoding, China .,2 Department of Cardiovascular Internal Medicine, Baoding First Central Hospital , Baoding, China
| | - Hong-jie Wang
- 1 Central Laboratory, Affiliated Hospital of Hebei University , Baoding, China
| | - Bing Wang
- 1 Central Laboratory, Affiliated Hospital of Hebei University , Baoding, China
| | - Ying Li
- 3 Department of Geriatrics, Baoding Second Hospital , Baoding, China
| | - Yan Qin
- 1 Central Laboratory, Affiliated Hospital of Hebei University , Baoding, China
| | - Li-shuang Zheng
- 1 Central Laboratory, Affiliated Hospital of Hebei University , Baoding, China
| | - Jin-sa Zhou
- 4 Department of Preventive Medicine, Hebei University , Baoding, China
| | - Peng-huan Qu
- 4 Department of Preventive Medicine, Hebei University , Baoding, China
| | - Jian-hong Shi
- 1 Central Laboratory, Affiliated Hospital of Hebei University , Baoding, China
| | - Hai-song Zhang
- 5 Department of Kidney Internal Medicine, Affiliated Hospital of Hebei University , Baoding, China
| |
Collapse
|
14
|
Kim JH, Kim HJ, Kim JK, Ahn EK, Ko HJ, Cho YR, Lee SJ, Bae GU, Kim YK, Park JW, Oh JS, Seo DW. Ligularia fischeri inhibits endothelial cell proliferation, invasion and tube formation through the inactivation of mitogenic signaling pathways and regulation of vascular endothelial cadherin distribution and matrix metalloproteinase expression. Oncol Rep 2015; 34:221-6. [PMID: 25998480 DOI: 10.3892/or.2015.4000] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 05/04/2015] [Indexed: 11/05/2022] Open
Abstract
Ligularia fischeri (LF) has been used as an edible herb and traditional medicine for the treatment of inflammatory and infectious diseases. In the present study, we report the effects and molecular mechanism of the ethanolic extract of LF on cell proliferation, invasion and tube formation in human umbilical vein endothelial cells (HUVECs). LF-mediated inhibition of cell proliferation was accompanied by reduced expression of cell cycle-related proteins such as cyclin-dependent kinases (Cdks) and cyclins, leading to pRb hypophosphorylation and G1 phase cell cycle arrest. We also show that LF treatment inhibited cell invasion and tube formation in HUVECs. These anti-angiogenic activities of LF were associated with the inactivation of mitogenic signaling pathways, induction of vascular endothelial (VE)-cadherin distribution at cell-cell contacts and inhibition of matrix metalloproteinase (MMP) expression. Collectively, our findings demonstrate the pharmacological functions and molecular mechanisms of LF in regulating endothelial cell fates, and support further development as a potential therapeutic agent for the treatment and prevention of angiogenesis-related disorders including cancer.
Collapse
Affiliation(s)
- Jae Hyeon Kim
- College of Pharmacy, Dankook University, Cheonan 330-714, Republic of Korea
| | - Hyeon-Ju Kim
- College of Pharmacy, Dankook University, Cheonan 330-714, Republic of Korea
| | - Jin-Kyu Kim
- Natural Products Research Institute, Gyeonggi Institute of Science and Technology Promotion, Suwon 443‑270, Republic of Korea
| | - Eun-Kyung Ahn
- Natural Products Research Institute, Gyeonggi Institute of Science and Technology Promotion, Suwon 443‑270, Republic of Korea
| | - Hye-Jin Ko
- Natural Products Research Institute, Gyeonggi Institute of Science and Technology Promotion, Suwon 443‑270, Republic of Korea
| | - Young-Rak Cho
- Natural Products Research Institute, Gyeonggi Institute of Science and Technology Promotion, Suwon 443‑270, Republic of Korea
| | - Sang-Jin Lee
- Research Center for Cell Fate Control, College of Pharmacy, Sookmyung Women's University, Seoul 140-742, Republic of Korea
| | - Gyu-Un Bae
- Research Center for Cell Fate Control, College of Pharmacy, Sookmyung Women's University, Seoul 140-742, Republic of Korea
| | - Yong Kee Kim
- Research Center for Cell Fate Control, College of Pharmacy, Sookmyung Women's University, Seoul 140-742, Republic of Korea
| | - Jong Woo Park
- School of Pharmacy, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| | - Joa Sub Oh
- College of Pharmacy, Dankook University, Cheonan 330-714, Republic of Korea
| | - Dong-Wan Seo
- College of Pharmacy, Dankook University, Cheonan 330-714, Republic of Korea
| |
Collapse
|
15
|
Zhang L, Li Y, Liu Y, Wang X, Chen M, Xing Y, Zhu D. STAT3-mediated MMP-2 expression is required for 15-HETE-induced vascular adventitial fibroblast migration. J Steroid Biochem Mol Biol 2015; 149:106-17. [PMID: 25623089 DOI: 10.1016/j.jsbmb.2015.01.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 01/19/2015] [Accepted: 01/21/2015] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Vascular adventitial fibroblasts (VAFs) migration was involved in neointima formation, and increased 15-HETE levels contributed to vascular remodeling. However, how 15-HETE-induced VAF migration was not clear. METHODS AND RESULTS 15-HETE-stimulated VAF phenotypic changes and migration as measured by the wound healing assay required STAT3 phosphorylation. JNK1 and CREB inhibition blocked 15-HETE-induced STAT3 activation and VAF changes. 15-HETE-induced MMP-2 expression and secretion were analyzed by Western blot and ELISA, respectively. MMP-2 knockdown blocked VAF migration and phenotypic alterations. JNK1, STAT3 and CREB blockade suppressed 15-HETE-induced MMP-2 expression in VAFs. MMP-2 promoter activity was assessed by chromatin immunoprecipitation using anti-STAT3 antibodies, which demonstrated that STAT3 was essential for 15-HETE-induced MMP-2 expression. Rats that suffered from hypoxia injury with or without treatment were examined. Pulmonary artery remodeling was obviously observed, and even the media was broken. MMP-2-positive staining was observed in the adventitia and intima. MMP-2 Serum secretion was enhanced as detected by ELISA, and MMP-2 and α-SMA protein expressions were increased after inducing hypoxia in the rats, which was restored in rats that had been administrated with NDGA. CONCLUSION These results reveal that STAT3-mediated MMP-2 expression is required for 15-HETE induced-VAF migration.
Collapse
Affiliation(s)
- Li Zhang
- Department of Pharmacology, Harbin Medical University - Daqing, Daqing, Heilongjiang Province 163319, China
| | - Yumei Li
- Department of Pharmacology, Harbin Medical University - Daqing, Daqing, Heilongjiang Province 163319, China; Biopharmaceutical Institute of the Heilongjiang Academy of Medical Sciences, Harbin, Heilongjiang Province 150081, China
| | - Yumei Liu
- Department of Pharmacology, Harbin Medical University - Daqing, Daqing, Heilongjiang Province 163319, China
| | - Xiaoyan Wang
- Department of Pharmacology, Harbin Medical University - Daqing, Daqing, Heilongjiang Province 163319, China
| | - Minggang Chen
- Department of Pharmacology, Harbin Medical University - Daqing, Daqing, Heilongjiang Province 163319, China
| | - Yan Xing
- Department of Pharmacology, Harbin Medical University - Daqing, Daqing, Heilongjiang Province 163319, China
| | - Daling Zhu
- Department of Pharmacology, Harbin Medical University - Daqing, Daqing, Heilongjiang Province 163319, China; Biopharmaceutical Institute of the Heilongjiang Academy of Medical Sciences, Harbin, Heilongjiang Province 150081, China.
| |
Collapse
|
16
|
|