1
|
Chien HW, Chuang CC, Hsieh YH, Lee CY, Yu NY, Yang SF. Tricetin suppresses the cell migration and BMP-6 expression through p38 signaling pathways in human retinal pigment epithelium cells. ENVIRONMENTAL TOXICOLOGY 2024; 39:4148-4155. [PMID: 38654487 DOI: 10.1002/tox.24294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 01/29/2024] [Accepted: 02/10/2024] [Indexed: 04/26/2024]
Abstract
Proliferative vitreoretinopathy (PVR) is a visual-threatening disease, which cause from the migration of retinal pigment epithelium (RPE). Tricetin, a family of flavonoids, can inhibit the metastasis of several cancers. Herein, we aim to evaluate the possible effect of tricetin on inhibiting ARPE-19 cells migration. The Boyden chamber assay, wound healing assay, RNA sequencing, and Western blot analysis were applied in our experiment. The results revealed that tricetin inhibited the cell migration abilities of ARPE-19 cells. Moreover, using RNA sequencing technology, we revealed that tricetin repressed bone morphogenetic protein-6 (BMP-6) gene expressions in ARPE-19 cells. Overexpression of BMP-6 resulted in significant restoration of cell migration capabilities of tricetin-treated ARPE-19 cells. Furthermore, tricetin suppressed the phosphorylation of the p38 signaling pathway. Moreover, blocking the p38 pathway also inhibits BMP-6 expression and migration in the ARPE-19 cells. In conclusion, this study revealed that tricetin inhibits the ARPE-19 cell migration mainly via the suppression of BMP-6 expression and p38 signaling pathway.
Collapse
Affiliation(s)
- Hsiang-Wen Chien
- Department of Ophthalmology, Cathay General Hospital, Taipei, Taiwan
- Department of Ophthalmology, Sijhih Cathay General Hospital, New Taipei City, Taiwan
- School of Medicine, National Tsing Hua University, Hsinchu, Taiwan
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei, Taiwan
| | - Chih-Chun Chuang
- Department of Ophthalmology, Changhua Christian Hospital, Changhua, Taiwan
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Yi-Hsien Hsieh
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chia-Yi Lee
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Nobel Eye Institute, Taipei, Taiwan
| | - Nuo-Yi Yu
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
2
|
Shoda C, Lee D, Miwa Y, Yamagami S, Nakashizuka H, Nimura K, Okamoto K, Kawagishi H, Negishi K, Kurihara T. Inhibition of hypoxia-inducible factors suppresses subretinal fibrosis. FASEB J 2024; 38:e23792. [PMID: 38953555 DOI: 10.1096/fj.202400540rrr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 06/15/2024] [Accepted: 06/24/2024] [Indexed: 07/04/2024]
Abstract
Age-related macular degeneration (AMD) is a common cause of vision loss. The aggressive form of AMD is associated with ocular neovascularization and subretinal fibrosis, representing a responsive outcome against neovascularization mediated by epithelial-mesenchymal transition of retinal pigment epithelium (RPE) cells. A failure of the current treatment (anti-vascular endothelial growth factor therapy) has also been attributed to the progression of subretinal fibrosis. Hypoxia-inducible factors (HIFs) increase gene expressions to promote fibrosis and neovascularization. HIFs act as a central pathway in the pathogenesis of AMD. HIF inhibitors may suppress ocular neovascularization. Nonetheless, further investigation is required to unravel the aspects of subretinal fibrosis. In this study, we used RPE-specific HIFs or von Hippel-Lindau (VHL, a regulator of HIFs) conditional knockout (cKO) mice, along with pharmacological HIF inhibitors, to demonstrate the suppression of subretinal fibrosis. Fibrosis was suppressed by treatments of HIF inhibitors, and similar suppressive effects were detected in RPE-specific Hif1a/Hif2a- and Hif1a-cKO mice. Promotive effects were observed in RPE-specific Vhl-cKO mice, where fibrosis-mediated pathologic processes were evident. Marine products' extracts and their component taurine suppressed fibrosis as HIF inhibitors. Our study shows critical roles of HIFs in the progression of fibrosis, linking them to the potential development of therapeutics for AMD.
Collapse
Affiliation(s)
- Chiho Shoda
- Laboratory of Photobiology, Keio University School of Medicine, Tokyo, Japan
- Ophthalmology, Keio University School of Medicine, Tokyo, Japan
- Ophthalmology, Nihon University School of Medicine, Tokyo, Japan
| | - Deokho Lee
- Laboratory of Photobiology, Keio University School of Medicine, Tokyo, Japan
- Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Yukihiro Miwa
- Laboratory of Photobiology, Keio University School of Medicine, Tokyo, Japan
- Ophthalmology, Keio University School of Medicine, Tokyo, Japan
- Aichi Animal Eye Clinic, Nagoya, Aichi, Japan
| | - Satoru Yamagami
- Ophthalmology, Nihon University School of Medicine, Tokyo, Japan
| | | | - Kazumi Nimura
- Shizuoka Prefectural Research Institute of Fishery and Ocean, Shizuoka, Japan
| | - Kazutoshi Okamoto
- Shizuoka Prefectural Research Institute of Fishery and Ocean, Shizuoka, Japan
- Marine Open Innovation Institute, Shizuoka, Japan
| | - Hirokazu Kawagishi
- Faculty of Agriculture, Shizuoka University, Shizuoka, Japan
- Research Institute for Mushroom Science, Shizuoka University, Shizuoka, Japan
| | - Kazuno Negishi
- Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Toshihide Kurihara
- Laboratory of Photobiology, Keio University School of Medicine, Tokyo, Japan
- Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
3
|
de Morais EF, de Oliveira LQR, de Farias Morais HG, de Souto Medeiros MR, Freitas RDA, Rodini CO, Coletta RD. The Anticancer Potential of Kaempferol: A Systematic Review Based on In Vitro Studies. Cancers (Basel) 2024; 16:585. [PMID: 38339336 PMCID: PMC10854650 DOI: 10.3390/cancers16030585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 01/25/2024] [Accepted: 01/26/2024] [Indexed: 02/12/2024] Open
Abstract
Given the heterogeneity of different malignant processes, planning cancer treatment is challenging. According to recent studies, natural products are likely to be effective in cancer prevention and treatment. Among bioactive flavonoids found in fruits and vegetables, kaempferol (KMP) is known for its anti-inflammatory, antioxidant, and anticancer properties. This systematic review aims to highlight the potential therapeutic effects of KMP on different types of solid malignant tumors. This review was conducted following the Preferred Reporting Items for Systematic Review and Meta-Analyses (PRISMA) guidelines. Searches were performed in EMBASE, Medline/PubMed, Cochrane Collaboration Library, Science Direct, Scopus, and Google Scholar. After the application of study criteria, 64 studies were included. In vitro experiments demonstrated that KMP exerts antitumor effects by controlling tumor cell cycle progression, proliferation, apoptosis, migration, and invasion, as well as by inhibiting angiogenesis. KMP was also able to inhibit important markers that regulate epithelial-mesenchymal transition and enhanced the sensitivity of cancer cells to traditional drugs used in chemotherapy, including cisplatin and 5-fluorouracil. This flavonoid is a promising therapeutic compound and its combination with current anticancer agents, including targeted drugs, may potentially produce more effective and predictable results.
Collapse
Affiliation(s)
- Everton Freitas de Morais
- Graduate Program in Oral Biology, Department of Oral Diagnosis, School of Dentistry, University of Campinas, Piracicaba 13414-018, SP, Brazil; (E.F.d.M.); (L.Q.R.d.O.)
| | - Lilianny Querino Rocha de Oliveira
- Graduate Program in Oral Biology, Department of Oral Diagnosis, School of Dentistry, University of Campinas, Piracicaba 13414-018, SP, Brazil; (E.F.d.M.); (L.Q.R.d.O.)
| | - Hannah Gil de Farias Morais
- Postgraduate Program in Oral Science, Federal University of Rio Grande do Norte, Natal 59000-000, RN, Brazil; (H.G.d.F.M.); (M.R.d.S.M.); (R.d.A.F.)
| | - Maurília Raquel de Souto Medeiros
- Postgraduate Program in Oral Science, Federal University of Rio Grande do Norte, Natal 59000-000, RN, Brazil; (H.G.d.F.M.); (M.R.d.S.M.); (R.d.A.F.)
| | - Roseana de Almeida Freitas
- Postgraduate Program in Oral Science, Federal University of Rio Grande do Norte, Natal 59000-000, RN, Brazil; (H.G.d.F.M.); (M.R.d.S.M.); (R.d.A.F.)
| | - Camila Oliveira Rodini
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Bauru 17012-901, SP, Brazil;
| | - Ricardo D. Coletta
- Graduate Program in Oral Biology, Department of Oral Diagnosis, School of Dentistry, University of Campinas, Piracicaba 13414-018, SP, Brazil; (E.F.d.M.); (L.Q.R.d.O.)
| |
Collapse
|
4
|
|
5
|
Liu D, Zhang C, Zhang J, Xu GT, Zhang J. Molecular pathogenesis of subretinal fibrosis in neovascular AMD focusing on epithelial-mesenchymal transformation of retinal pigment epithelium. Neurobiol Dis 2023; 185:106250. [PMID: 37536385 DOI: 10.1016/j.nbd.2023.106250] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 07/11/2023] [Accepted: 07/31/2023] [Indexed: 08/05/2023] Open
Abstract
Age-related macular degeneration (AMD) is a leading cause of vision loss among elderly people in developed countries. Neovascular AMD (nAMD) accounts for more than 90% of AMD-related vision loss. At present, intravitreal injection of anti-vascular endothelial growth factor (anti-VEGF) is widely used as the first-line therapy to decrease the choroidal and retinal neovascularizations, and thus to improve or maintain the visual acuity of the patients with nAMD. However, about 1/3 patients still progress to irreversible visual impairment due to subretinal fibrosis even with adequate anti-VEGF treatment. Extensive literatures support the critical role of epithelial-mesenchymal transformation (EMT) of retinal pigment epithelium (RPE) in the pathogenesis of subretinal fibrosis in nAMD, but the underlying mechanisms still remain largely unknown. This review summarized the molecular pathogenesis of subretinal fibrosis in nAMD, especially focusing on the transforming growth factor-β (TGF-β)-induced EMT pathways. It was also discussed how these pathways crosstalk and respond to signals from the microenvironment to mediate EMT and contribute to the progression of nAMD-related subretinal fibrosis. Targeting EMT signaling pathways might provide a promising and effective therapeutic strategy to treat subretinal fibrosis secondary to nAMD.
Collapse
Affiliation(s)
- Dandan Liu
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, China
| | - Chaoyang Zhang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University, Shanghai, China; National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Jingting Zhang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University, Shanghai, China; National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Guo-Tong Xu
- Department of Ophthalmology of Tongji Hospital and Laboratory of Clinical and Visual Sciences of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, China.
| | - Jingfa Zhang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University, Shanghai, China; National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China.
| |
Collapse
|
6
|
Eltahir S, Ahmad A. Flavonoids on the Frontline against Cancer Metastasis. Cancers (Basel) 2023; 15:4139. [PMID: 37627166 PMCID: PMC10452402 DOI: 10.3390/cancers15164139] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023] Open
Abstract
Metastasis is the leading cause of death in cancer patients [...].
Collapse
Affiliation(s)
- Sarah Eltahir
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar;
| | - Aamir Ahmad
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar;
- Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar
- Department of Dermatology and Venereology, Rumailah Hospital, Hamad Medical Corporation, Doha 3050, Qatar
- Department of Anesthesiology and Perioperative Medicine, University of Alabama, 901 19th St. South, Birmingham, AL 35294, USA
| |
Collapse
|
7
|
Lee CY, Ho YC, Lin CW, Hsin MC, Wang PH, Tang YC, Yang SF, Hsiao YH. EF-24 inhibits TPA-induced cellular migration and MMP-9 expression through the p38 signaling pathway in cervical cancer cells. ENVIRONMENTAL TOXICOLOGY 2023; 38:451-459. [PMID: 36413041 DOI: 10.1002/tox.23709] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 10/26/2022] [Accepted: 11/05/2022] [Indexed: 06/16/2023]
Abstract
Diphenyl difluoroketone (EF-24), a synthetic curcumin analog, has enhanced bioavailability over curcumin. EF-24 acts more powerful bioactivity for anti-inflammatory and anti-cancer activity. However, the effects and mechanism of EF-24 on cervical cancer has not been fully investigated. Herein, this study evaluated the effects of EF-24 on TPA-induced cellular migration of cervical cancer. The results showed that EF-24 substantially reduced the cellular migration and cellular invasion of the HeLa and SiHa cells. Moreover, gelatin zymography, western blotting analyses and real-time PCR revealed that EF-24 suppressed Matrix metalloproteinase-9 (MMP-9) activity, protein expression and mRNA levels. Mechanistically, EF-24 inhibited the phosphorylation of the p38 signaling pathway. In conclusion, EF-24 inhibited TPA-induced cellular migration and cellular invasion of cervical cancer cell lines through modulating MMP-9 expression via downregulating signaling p38 pathway and EF-24 may have potential to serve as a chemopreventive agent of cervical cancer.
Collapse
Affiliation(s)
- Chung-Yuan Lee
- Department of Obstetrics and Gynecology, Chiayi Chang Gung Memorial Hospital, Chiayi, Taiwan
- Department of Nursing, Chang Gung University of Science and Technology, Chiayi, Taiwan
| | - Yung-Chuan Ho
- Department of Medical Applied Chemistry, Chung Shan Medical University, Taichung, Taiwan
| | - Chiao-Wen Lin
- Institute of Oral Sciences, Chung Shan Medical University, Taichung, Taiwan
| | - Min-Chieh Hsin
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Po-Hui Wang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Obstetrics and Gynecology, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Ya-Cheng Tang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Yi-Hsuan Hsiao
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Obstetrics and Gynecology, Changhua Christian Hospital, Changhua, Taiwan
- Women's Health Research Laboratory, Changhua Christian Hospital, Changhua, Taiwan
| |
Collapse
|
8
|
Guan B, Li H, Yao J, Guo J, Yu F, Li G, Wan B, Ma J, Huang D, Sun L, Chen Y. CCL3-CCR5 axis promotes cell migration and invasion of colon adenocarcinoma via Akt signaling pathway. ENVIRONMENTAL TOXICOLOGY 2023; 38:172-184. [PMID: 36346222 DOI: 10.1002/tox.23675] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 08/19/2022] [Accepted: 09/16/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Infiltration of tumor-associated macrophages (TAMs) can promote tumorigenesis and development. C-C motif chemokine ligand 3 (CCL3) was reported to be derived from TAMs and tumor cells and facilitate the progression of several cancers. Nevertheless, whether CCL3 can be derived from TAMs and tumor cells of colon adenocarcinoma (COAD) is unclarified. METHODS Peripheral blood monocytes-derived macrophages were polarized by the conditioned medium from COAD cells to establish TAM-like macrophages (TAM1/2). RT-qPCR and western blotting were used for detection of expression levels of CCL3 and its receptors C-C motif chemokine receptor 1 (CCR1) and CCR5 in TAM1/2 and COAD cells. Immunofluorescence staining was utilized for evaluating CCL3, CD163 and CCR5 expression. The Akt signaling pathway-associated protein levels were measured by western blotting. Transwell assays were used for assessing cell migration and invasiveness. RESULTS CCL3 displayed a high level in TAMs and cancer cells of COAD. CCL3 activated the Akt signaling pathway by binding to CCR5. CCL3-CCR5 axis facilitated COAD cell migration and invasiveness by activating the Akt signaling. CCL3 derived from both TAMs and cancer cells contributed to the malignant behaviors of COAD cells. High expression of CCL3/CCR5 was closely associated with poor prognoses of COAD patients. CONCLUSION CCL3-CCR5 interaction promotes cell migration and invasiveness, and functions as a prognostic biomarker for COAD.
Collapse
Affiliation(s)
- Bugao Guan
- Department of General Surgery, Jinhu People's Hospital, Huai'an, China
| | - Hongbo Li
- Department of General Surgery, Jinhu People's Hospital, Huai'an, China
| | - Jian Yao
- Department of General Surgery, Jinhu People's Hospital, Huai'an, China
| | - Jinbao Guo
- Department of General Surgery, Jinhu People's Hospital, Huai'an, China
| | - Fei Yu
- Department of General Surgery, Jinhu People's Hospital, Huai'an, China
| | - Guangrun Li
- Department of General Surgery, Jinhu People's Hospital, Huai'an, China
| | - Benhai Wan
- Department of General Surgery, Jinhu People's Hospital, Huai'an, China
| | - Jun Ma
- Department of General Surgery, Jinhu People's Hospital, Huai'an, China
| | - Desong Huang
- Department of General Surgery, Jinhu People's Hospital, Huai'an, China
| | - Lu Sun
- Department of General Surgery, Jinhu People's Hospital, Huai'an, China
| | - Yan Chen
- The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an Second People's Hospital, Huai'an, China
| |
Collapse
|
9
|
Lee H, Han JH, Kang YJ, Hwangbo H, Yoon A, Kim HS, Lee D, Lee SY, Choi BH, Kim JJ, Kim SR, Choi YH, Hur J. CD82 attenuates TGF-β1-mediated epithelial-mesenchymal transition by blocking smad-dependent signaling in ARPE-19 cells. Front Pharmacol 2022; 13:991056. [PMID: 36386228 PMCID: PMC9640495 DOI: 10.3389/fphar.2022.991056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 10/17/2022] [Indexed: 11/27/2022] Open
Abstract
In retinal pigment epithelial (RPE) cells, transforming growth factor-beta (TGF-β) plays a critical role in epithelial-mesenchymal transition (EMT), which contributes to various fibrotic retinal disorders. In the present study, we investigated the effect of recombinant human cluster of differentiation 82 (rhCD82), a tumor metastasis suppressor, on TGF-β-induced EMT in the human RPE cell line APRE-19. The results show that TGF-β1 significantly enhanced cell migration, invasion and the expression of EMT-mediate factors in ARPE-19 cells. However, rhCD82 markedly inhibited cell mobility and the expression of epithelial marker, zonula occludens-1, as well as increased the expression of mesenchymal markers, such as vimentin and α-smooth muscle actin in TGF-β1-treated APRE-19 cells. In addition, TGF-β1 upregulated the phosphorylation of Smad, extracellular signal regulated kinase (ERK) and glycogen synthase kinase-3β (GSK-3β), but only phosphorylation of Smad was suppressed by rhCD82. Noteworthy, rhCD82 greatly suppressed the expression of TGF-β receptor I (TGFRI), TGFRII and integrins in TGF-β1-treated APRE-19 cells. In particular, the result of molecular docking analysis and structural modeling show that rhCD82 partially interacts with the TGF-β1 binding sites of TGFRI, TGFRII, integrin β1 and integrin αv. Taken together, this finding suggested that rhCD82 suppressed TGF-β1-induced EMT of RPE by blocking of Smad-dependent pathway, which is caused by rhCD82 interaction with TGFRs and integrins, suggesting new insight into CD82 as a potential therapeutic strategy in fibrotic retinal disorders.
Collapse
Affiliation(s)
- Hyesook Lee
- Department of Convergence Medicine, Pusan National University School of Medicine, Yangsan, South Korea
| | - Jung-Hwa Han
- Department of Convergence Medicine, Pusan National University School of Medicine, Yangsan, South Korea
- PNU GRAND Convergence Medical Science Education Research Center, Pusan National University School of Medicine, Yangsan, South Korea
| | - Yun Jeong Kang
- Department of Convergence Medicine, Pusan National University School of Medicine, Yangsan, South Korea
| | - Hyun Hwangbo
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan, South Korea
| | - Aeseon Yoon
- Department of Convergence Medicine, Pusan National University School of Medicine, Yangsan, South Korea
| | - Hyung-Sik Kim
- Department of Oral Biochemistry, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan, South Korea
| | - Dongjun Lee
- Department of Convergence Medicine, Pusan National University School of Medicine, Yangsan, South Korea
| | - Soo Yong Lee
- Division of Cardiology, Department of Internal Medicine and Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, South Korea
| | - Byung Hyun Choi
- Division of Hepato-Biliary-Pancreatic Surgery and Transplantation, Department of Surgery, Pusan National University School of Medicine and Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, South Korea
| | - Jae-Joon Kim
- Medical Oncology and Hematology, Department of Internal Medicine, Pusan National University Yangsan Hospital, Yangsan, South Korea
| | - Seo Rin Kim
- Department of Nephrology and Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, South Korea
| | - Yung Hyun Choi
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan, South Korea
- Anti-Aging Research Center and Core-Facility Center for Tissue Regeneration, Dong-eui University, Busan, South Korea
- *Correspondence: Yung Hyun Choi, ; Jin Hur,
| | - Jin Hur
- Department of Convergence Medicine, Pusan National University School of Medicine, Yangsan, South Korea
- PNU GRAND Convergence Medical Science Education Research Center, Pusan National University School of Medicine, Yangsan, South Korea
- *Correspondence: Yung Hyun Choi, ; Jin Hur,
| |
Collapse
|
10
|
Zhao J, Wang J, Liu J, Li S, Liu P, Zhang X. Effect and mechanisms of kaempferol against endometriosis based on network pharmacology and in vitro experiments. BMC Complement Med Ther 2022; 22:254. [PMID: 36184634 PMCID: PMC9528065 DOI: 10.1186/s12906-022-03729-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 09/12/2022] [Indexed: 12/03/2022] Open
Abstract
Endometriosis is a common gynecological disease, and its underlying mechanisms remain elusive. Patients are at a higher risk of recurrence after surgery or drug withdrawal. In this study, to identify a potentially effective and safe therapy for endometriosis, we screened potential target genes of kaempferol on endometriosis using network pharmacology and further validation. Network pharmacology showed kaempferol may suppress migratory and invasive properties by modulating the phosphoinositide 3-kinase (PI3K) pathway and its downstream target matrix metalloproteinase (MMP)9. Furthermore, in vitro experiments showed that kaempferol repressed the migration and invasion of endometrial cells, and this effect may be involved in mediating the PI3K-related genes, phosphatase and tensin homolog (PTEN) and MMP9. Network pharmacology and in vitro experiments showed that kaempferol, repressed the implantation of endometrial cells and formation of ectopic lesions by inhibiting migration and invasion and regulating PTEN and MMP9, which may be associated with the PI3K pathway.
Collapse
Affiliation(s)
- Junde Zhao
- grid.452402.50000 0004 1808 3430Department of Traditional Chinese Medicine, Qilu Hospital of Shandong University, Jinan, 250012 China ,grid.452402.50000 0004 1808 3430Laboratory of Basic Medical Sciences, Qilu Hospital of Shandong University, Jinan, 250012 China ,grid.464402.00000 0000 9459 9325First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250011 China
| | - Juntao Wang
- grid.452402.50000 0004 1808 3430Division of Hand and Foot Surgery, Department of Orthopedic Surgery, Qilu Hospital of Shandong University, Jinan, 250012 China
| | - Jinxing Liu
- grid.464402.00000 0000 9459 9325First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250011 China
| | - Shuling Li
- grid.452402.50000 0004 1808 3430Department of Traditional Chinese Medicine, Qilu Hospital of Shandong University, Jinan, 250012 China
| | - Pengfei Liu
- grid.464402.00000 0000 9459 9325First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250011 China
| | - Xiaodan Zhang
- grid.452402.50000 0004 1808 3430Department of Traditional Chinese Medicine, Qilu Hospital of Shandong University, Jinan, 250012 China
| |
Collapse
|
11
|
Ju PC, Ho YC, Chen PN, Lee HL, Lai SY, Yang SF, Yeh CB. Kaempferol inhibits the cell migration of human hepatocellular carcinoma cells by suppressing MMP-9 and Akt signaling. ENVIRONMENTAL TOXICOLOGY 2021; 36:1981-1989. [PMID: 34156145 DOI: 10.1002/tox.23316] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 04/26/2021] [Accepted: 06/15/2021] [Indexed: 06/13/2023]
Abstract
Metastasis is the most prevalent cause of cancer-related deaths and treatment failure in patients with hepatocellular carcinoma (HCC). Kaempferol is a natural flavonol belonging to the subgroup of flavonoids and exhibits potent anticancer activities. This study provides molecular evidence on the anti-invasive and anti-migratory effects of kaempferol on human HCC cells. The anti-invasive effect was investigated by applying kaempferol on two human HCC cell lines (Huh-7 and SK-Hep-1). Kaempferol reduced the invasion and migration of Huh-7 and SK-Hep-1 cells by Boyden chamber invasion assay and wound healing assay, respectively. A protease array analysis showed that Matrix Metalloproteinase-9 (MMP-9) was dramatically downregulated in HCC cells after kaempferol treatment. Gelatin zymography and Western blot assay showed that kaempferol reduced the activities and protein expression of MMP-9, respectively. Kaempferol also sufficiently suppressed the phosphorylation of the Akt expression. Overall, kaempferol inhibited the invasive properties of human HCC cells by targeting MMP-9 and Akt pathways. Hence, kaempferol could be used as an adjuvant therapeutic agent for the treatment of human HCC cells.
Collapse
Affiliation(s)
- Po-Chung Ju
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Psychiatry, Chung Shan Medical University Hospital, Taichung, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Yung-Chuan Ho
- School of Medical Applied Chemistry, Chung Shan Medical University, Taichung, Taiwan
| | - Pei-Ni Chen
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Hsiang-Lin Lee
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Surgery, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Szu-Yu Lai
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chao-Bin Yeh
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Emergency Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
12
|
Chou CH, Lu KH, Yang JS, Hsieh YH, Lin CW, Yang SF. Dihydromyricetin suppresses cell metastasis in human osteosarcoma through SP-1- and NF-κB-modulated urokinase plasminogen activator inhibition. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 90:153642. [PMID: 34265701 DOI: 10.1016/j.phymed.2021.153642] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 06/18/2021] [Accepted: 06/22/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Metastasis caused a decline in the 5-years survival rate of osteosarcoma. Therefore, developing new targeted therapeutics for osteosarcoma treatment is imperative. Dihydromyricetin (DHM) has several physiological functions: it counteracts inflammation, oxidation, and antitumor properties. However, the effects of DHM on osteosarcoma and its underlying mechanisms are still not well understood. PURPOSE In this study, we investigated the antimetastatic properties of DHM in human osteosarcoma U-2 OS and HOS cells. METHODS The effects of DHM (0, 25, 50, 75, and 100 μM) on cell viability, migration, and invasion were examined. Western blotting, RT-PCR, and quantitative real-time PCR (QPCR) were determined urokinase plasminogen activator (uPA) expression. The expression of transcriptional factor SP-1 and NF-κB was determined by using immunofluorescence assay, chromatin immunoprecipitation assay, and site-directed mutagenesis luciferase reporter. RESULTS We observed that DHM suppresses cell migration and invasion in osteosarcoma cell lines. In addition, DHM inhibits metastasis by downregulating urokinase plasminogen activator (uPA) expression. Moreover, real-time polymerase chain reaction and promoter activity assays revealed that DHM decreased uPA expression at transcription levels. Furthermore, the inhibition of uPA expression was associated with the suppression of SP-1 and NF-κB, which bind to the uPA promoter. Regardless of blocking or inducing the extracellular signal-regulated kinase (ERK) pathway, we verified that the DHM-related suppression of uPA and cell metastasis occurred through the p-ERK pathway. CONCLUSION We are the first study to propose that DHM suppresses osteosarcoma metastasis through the ERK pathway and through the suppression of SP-1 and NF-κB to inhibit downstream uPA expression. DHM is a potential therapeutic agent for antimetastatic therapy against osteosarcoma.
Collapse
Affiliation(s)
- Chia-Hsuan Chou
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan; Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Ko-Hsiu Lu
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan; Department of Orthopedics, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Jia-Sin Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan; Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Yi-Hsien Hsieh
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan
| | - Chiao-Wen Lin
- Institute of Oral Sciences, Chung Shan Medical University, Taichung, Taiwan; Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan.
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan; Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan.
| |
Collapse
|
13
|
Jantan I, Haque MA, Arshad L, Harikrishnan H, Septama AW, Mohamed-Hussein ZA. Dietary polyphenols suppress chronic inflammation by modulation of multiple inflammation-associated cell signaling pathways. J Nutr Biochem 2021; 93:108634. [PMID: 33794330 DOI: 10.1016/j.jnutbio.2021.108634] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 01/20/2021] [Accepted: 03/10/2021] [Indexed: 02/06/2023]
Abstract
The high failure rate of the reductionist approach to discover effective and safe drugs to treat chronic inflammatory diseases has led scientists to seek alternative ways. Recently, targeting cell signaling pathways has been utilized as an innovative approach to discover drug leads from natural products. Cell signaling mechanisms have been identified playing key role in diverse diseases by inducing proliferation, cell survival and apoptosis. Phytochemicals are known to be able to modulate the cellular and molecular networks which are associated to chronic diseases including cancer-associated inflammation. In this review, the roles of dietary polyphenols (apigenin, kaempferol, quercetin, curcumin, genistein, isoliquiritigenin, resveratrol and gallic acid) in modulating multiple inflammation-associated cell signaling networks are deliberated. Scientific databases on suppressive effects of the polyphenols on chronic inflammation via modulation of the pathways especially in the recent five years are gathered and critically analyzed. The polyphenols are able to modulate several inflammation-associated cell signaling pathways, namely nuclear factor-kappa β, mitogen activated protein kinases, Wnt/β-catenin and phosphatidylinositol 3-kinase and protein kinase B via selective actions on various components of the networks. The suppressive effects of the polyphenols on the multiple cell signaling pathways reveal their potential use in prevention and treatment of chronic inflammatory disorders. Understanding the mechanistic effects involved in modulation of the signaling pathways by the polyphenols is necessary for lead identification and development of future functional foods for prevention and treatment of chronic inflammatory diseases.
Collapse
Affiliation(s)
- Ibrahim Jantan
- Institute of Systems Biology (INBIOSIS), Universiti Kebangsaan Malaysia, UKM Bangi, Selangor, Malaysia.
| | - Md Areeful Haque
- Department of Pharmacy, International Islamic University Chittagong, Chittagong, Bangladesh
| | - Laiba Arshad
- Department of Pharmacy, Forman Christian College (A Chartered University), Lahore, Pakistan
| | - Hemavathy Harikrishnan
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Abdi Wira Septama
- Research Center for Chemistry, Indonesian Institute of Sciences, Kawasan PUSPIPTEK Serpong, Tangerang Selatan, Banten, Indonesia
| | - Zeti-Azura Mohamed-Hussein
- Institute of Systems Biology (INBIOSIS), Universiti Kebangsaan Malaysia, UKM Bangi, Selangor, Malaysia; Department of Applied Physics, Faculty of Science & Technology, Universiti Kebangsaan Malaysia, UKM Bangi, Selangor
| |
Collapse
|
14
|
Khater M, Greco F, Osborn HMI. Antiangiogenic Activity of Flavonoids: A Systematic Review and Meta-Analysis. Molecules 2020; 25:E4712. [PMID: 33066630 PMCID: PMC7594036 DOI: 10.3390/molecules25204712] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 10/08/2020] [Accepted: 10/10/2020] [Indexed: 12/16/2022] Open
Abstract
Abstract: An imbalance of angiogenesis contributes to many pathologies such as cancer, arthritis and retinopathy, hence molecules that can modulate angiogenesis are of considerable therapeutic importance. Despite many reports on the promising antiangiogenic properties of naturally occurring flavonoids, no flavonoids have progressed to the clinic for this application. This systematic review and meta-analysis therefore evaluates the antiangiogenic activities of a wide range of flavonoids and is presented in two sections. The first part of the study (Systematic overview) included 402 articles identified by searching articles published before May 2020 using ScienceDirect, PubMed and Web of Science databases. From this initial search, different classes of flavonoids with antiangiogenic activities, related pathologies and use of in vitro and/or in/ex vivo angiogenesis assays were identified. In the second part (Meta-analysis), 25 studies concerning the antiangiogenic evaluation of flavonoids using the in vivo chick chorioallantoic membrane (CAM) assay were included, following a targeted search on articles published prior to June 2020. Meta-analysis of 15 out of the 25 eligible studies showed concentration dependent antiangiogenic activity of six compared subclasses of flavonoids with isoflavones, flavonols and flavones being the most active (64 to 80% reduction of blood vessels at 100 µM). Furthermore, the key structural features required for the antiangiogenic activity of flavonoids were derived from the pooled data in a structure activity relationship (SAR) study. All in all, flavonoids are promising candidates for the development of antiangiogenic agents, however further investigations are needed to determine the key structural features responsible for their activity.
Collapse
Affiliation(s)
- Mai Khater
- School of Pharmacy, University of Reading, Whiteknights, Reading RG6 6AD, UK; (M.K.); (F.G.)
- Therapeutic Chemistry Department, Pharmaceutical & Drug Industries Research Division, National Research Centre, Cairo 12622, Egypt
| | - Francesca Greco
- School of Pharmacy, University of Reading, Whiteknights, Reading RG6 6AD, UK; (M.K.); (F.G.)
| | - Helen M. I. Osborn
- School of Pharmacy, University of Reading, Whiteknights, Reading RG6 6AD, UK; (M.K.); (F.G.)
| |
Collapse
|
15
|
Anti-Cancer Potential of Cannabinoids, Terpenes, and Flavonoids Present in Cannabis. Cancers (Basel) 2020; 12:cancers12071985. [PMID: 32708138 PMCID: PMC7409346 DOI: 10.3390/cancers12071985] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/17/2020] [Accepted: 07/19/2020] [Indexed: 02/06/2023] Open
Abstract
In recent years, and even more since its legalization in several jurisdictions, cannabis and the endocannabinoid system have received an increasing amount of interest related to their potential exploitation in clinical settings. Cannabinoids have been suggested and shown to be effective in the treatment of various conditions. In cancer, the endocannabinoid system is altered in numerous types of tumours and can relate to cancer prognosis and disease outcome. Additionally, cannabinoids display anticancer effects in several models by suppressing the proliferation, migration and/or invasion of cancer cells, as well as tumour angiogenesis. However, the therapeutic use of cannabinoids is currently limited to the treatment of symptoms and pain associated with chemotherapy, while their potential use as cytotoxic drugs in chemotherapy still requires validation in patients. Along with cannabinoids, cannabis contains several other compounds that have also been shown to exert anti-tumorigenic actions. The potential anti-cancer effects of cannabinoids, terpenes and flavonoids, present in cannabis, are explored in this literature review.
Collapse
|
16
|
Flavonoids in Cancer Metastasis. Cancers (Basel) 2020; 12:cancers12061498. [PMID: 32521759 PMCID: PMC7352928 DOI: 10.3390/cancers12061498] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 05/29/2020] [Accepted: 06/04/2020] [Indexed: 12/12/2022] Open
Abstract
Metastasis represents a serious complication in the treatment of cancer. Flavonoids are plant secondary metabolites exerting various health beneficiary effects. The effects of flavonoids against cancer are associated not only with early stages of the cancer process, but also with cancer progression and spread into distant sites. Flavonoids showed potent anti-cancer effects against various cancer models in vitro and in vivo, mediated via regulation of key signaling pathways involved in the migration and invasion of cancer cells and metastatic progression, including key regulators of epithelial-mesenchymal transition or regulatory molecules such as MMPs, uPA/uPAR, TGF-β and other contributors of the complex process of metastatic spread. Moreover, flavonoids modulated also the expression of genes associated with the progression of cancer and improved inflammatory status, a part of the complex process involved in the development of metastasis. Flavonoids also documented clear potential to improve the anti-cancer effectiveness of conventional chemotherapeutic agents. Most importantly, flavonoids represent environmentally-friendly and cost-effective substances; moreover, a wide spectrum of different flavonoids demonstrated safety and minimal side effects during long-termed administration. In addition, the bioavailability of flavonoids can be improved by their conjugation with metal ions or structural modifications by radiation. In conclusion, anti-cancer effects of flavonoids, targeting all phases of carcinogenesis including metastatic progression, should be implemented into clinical cancer research in order to strengthen their potential use in the future targeted prevention and therapy of cancer in high-risk individuals or patients with aggressive cancer disease with metastatic potential.
Collapse
|
17
|
Kumar GB, Nair BG, Perry JJP, Martin DBC. Recent insights into natural product inhibitors of matrix metalloproteinases. MEDCHEMCOMM 2019; 10:2024-2037. [PMID: 32904148 PMCID: PMC7451072 DOI: 10.1039/c9md00165d] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 09/11/2019] [Indexed: 12/19/2022]
Abstract
Members of the matrix metalloproteinase (MMP) family have biological functions that are central to human health and disease, and MMP inhibitors have been investigated for the treatment of cardiovascular disease, cancer and neurodegenerative disorders. The outcomes of initial clinical trials with the first generation of MMP inhibitors proved disappointing. However, our growing understanding of the complexities of the MMP function in disease, and an increased understanding of MMP protein architecture and control of activity now provide new opportunities and avenues to develop MMP-focused therapies. Natural products that affect MMP activities have been of strong interest as templates for drug discovery, and for their use as chemical tools to help delineate the roles of MMPs that still remain to be defined. Herein, we highlight the most recent discoveries of structurally diverse natural product inhibitors to these proteases.
Collapse
Affiliation(s)
- Geetha B Kumar
- School of Biotechnology , Amrita University , Kollam , Kerala , India
| | - Bipin G Nair
- School of Biotechnology , Amrita University , Kollam , Kerala , India
| | - J Jefferson P Perry
- School of Biotechnology , Amrita University , Kollam , Kerala , India
- Department of Biochemistry , University of California , Riverside , CA 92521 , USA .
| | - David B C Martin
- Department of Chemistry , University of California , Riverside , CA 92521 , USA
- Department of Chemistry , University of Iowa , Iowa City , IA 52242 , USA .
| |
Collapse
|