1
|
Lee YH, So BH, Lee KS, Kuk MU, Park JH, Yoon JH, Lee YJ, Kim DY, Kim MS, Kwon HW, Byun Y, Lee KY, Park JT. Identification of Cellular Isoschaftoside-Mediated Anti-Senescence Mechanism in RAC2 and LINC00294. Molecules 2024; 29:4182. [PMID: 39275030 PMCID: PMC11397025 DOI: 10.3390/molecules29174182] [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: 08/06/2024] [Revised: 08/31/2024] [Accepted: 09/02/2024] [Indexed: 09/16/2024] Open
Abstract
As cellular senescence, reactive oxygen species (ROS) accumulate excessively, causing cellular damage. Flavonoids derived from natural products are known for their antioxidant effects and their ability to delay cellular senescence. Previous studies have attempted to mitigate cellular senescence using flavonoids from natural sources. However, the detailed mechanisms and regulatory targets of some flavonoids exhibiting antioxidant effects have not been fully elucidated. Therefore, we screened a library of flavonoids for antioxidant properties. Isoschaftoside, a glycosidic flavonoid, significantly reduced ROS levels in senescent cells. It was found that mitochondrial function was restored, and dependence on glycolysis was reduced in senescent cells treated with isoschaftoside. Additionally, we identified that isoschaftoside suppresses ROS by reducing the expression of RAC2 and LINC00294 in senescent cells. Taken together, this study establishes a novel mechanism for ROS inhibition and the regulation of cellular senescence by isoschaftoside. Our findings contribute important insights to antioxidant and anti-senescence research.
Collapse
Affiliation(s)
- Yun Haeng Lee
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea; (Y.H.L.); (B.H.S.); (M.U.K.); (J.H.P.); (J.H.Y.); (Y.J.L.); (D.Y.K.); (M.S.K.); (H.W.K.)
| | - Byeong Hyeon So
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea; (Y.H.L.); (B.H.S.); (M.U.K.); (J.H.P.); (J.H.Y.); (Y.J.L.); (D.Y.K.); (M.S.K.); (H.W.K.)
| | - Kyeong Seon Lee
- College of Pharmacy, Korea University, Sejong 30019, Republic of Korea; (K.S.L.); (Y.B.)
| | - Myeong Uk Kuk
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea; (Y.H.L.); (B.H.S.); (M.U.K.); (J.H.P.); (J.H.Y.); (Y.J.L.); (D.Y.K.); (M.S.K.); (H.W.K.)
| | - Ji Ho Park
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea; (Y.H.L.); (B.H.S.); (M.U.K.); (J.H.P.); (J.H.Y.); (Y.J.L.); (D.Y.K.); (M.S.K.); (H.W.K.)
| | - Jee Hee Yoon
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea; (Y.H.L.); (B.H.S.); (M.U.K.); (J.H.P.); (J.H.Y.); (Y.J.L.); (D.Y.K.); (M.S.K.); (H.W.K.)
| | - Yoo Jin Lee
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea; (Y.H.L.); (B.H.S.); (M.U.K.); (J.H.P.); (J.H.Y.); (Y.J.L.); (D.Y.K.); (M.S.K.); (H.W.K.)
| | - Du Yeol Kim
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea; (Y.H.L.); (B.H.S.); (M.U.K.); (J.H.P.); (J.H.Y.); (Y.J.L.); (D.Y.K.); (M.S.K.); (H.W.K.)
| | - Min Seon Kim
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea; (Y.H.L.); (B.H.S.); (M.U.K.); (J.H.P.); (J.H.Y.); (Y.J.L.); (D.Y.K.); (M.S.K.); (H.W.K.)
| | - Hyung Wook Kwon
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea; (Y.H.L.); (B.H.S.); (M.U.K.); (J.H.P.); (J.H.Y.); (Y.J.L.); (D.Y.K.); (M.S.K.); (H.W.K.)
- Convergence Research Center for Insect Vectors, Incheon National University, Incheon 22012, Republic of Korea
| | - Youngjoo Byun
- College of Pharmacy, Korea University, Sejong 30019, Republic of Korea; (K.S.L.); (Y.B.)
| | - Ki Yong Lee
- College of Pharmacy, Korea University, Sejong 30019, Republic of Korea; (K.S.L.); (Y.B.)
| | - Joon Tae Park
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea; (Y.H.L.); (B.H.S.); (M.U.K.); (J.H.P.); (J.H.Y.); (Y.J.L.); (D.Y.K.); (M.S.K.); (H.W.K.)
- Convergence Research Center for Insect Vectors, Incheon National University, Incheon 22012, Republic of Korea
| |
Collapse
|
2
|
Ain NU, Khan B, Zhu K, Ji W, Tian H, Yu X, Yi L, Li D, Zhang Z. Fabrication of mesoporous silica nanoparticles for releasable delivery of licorice polysaccharide at the acne site in topical application. Carbohydr Polym 2024; 339:122250. [PMID: 38823917 DOI: 10.1016/j.carbpol.2024.122250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 05/05/2024] [Accepted: 05/08/2024] [Indexed: 06/03/2024]
Abstract
Glycyrrhizae Radix et rhizome/licorice is a precious herb in traditional Chinese medicine (TCM). TCM's polysaccharides are medicinally active. But herbal polysaccharides pose some limitations for topical applications. Therefore, this study aimed to utilize licorice polysaccharide via mesoporous silica nanoparticles (MSN) for anti-acne efficacy in topical delivery. The polysaccharide (GGP) was extracted with a 10 % NaOH solution. Chemical characterization suggested that GGP possesses an Mw of 267.9 kDa, comprised primarily of Glc (54.1 %) and Ara (19.12 %), and probably 1,4-linked Glc as a backbone. Then, MSN and amino-functionalized MSN were synthesized, GGP entrapped, and coated with polydopamine (PDA) to produce nanoparticle cargo. The resulted product exhibited 76 % entrapment efficiency and an in vitro release of 89 % at pH 5, which is usually an acne-prone skin's pH. Moreover, it significantly increased Sebocytes' cellular uptake. GGP effectively acted as an anti-acne agent and preserved its efficacy in synthesized nanoparticles. In vivo, the results showed that a 20 % gel of MSN-NH2-GGP@PDA could mediate an inflammatory response via inhibiting pro-inflammatory cytokines and regulating anti-inflammatory cytokines. The MSN-NH2-GGP@PDA inhibited TLR2-activated-MAPK and NF-κB pathway triggered by heat-killed P. acnes. In conclusion, fabricated MSN entrapped GGP for biomimetic anti-acne efficacy in topical application.
Collapse
Affiliation(s)
- Noor Ul Ain
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215021, PR China
| | - Bibimaryam Khan
- School of Medicine, Jiangsu University, Zhenjiang 212013, PR China
| | - Kehan Zhu
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215021, PR China
| | - Wen Ji
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215021, PR China
| | - He Tian
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215021, PR China
| | - Xiaoxiao Yu
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215021, PR China
| | - Lin Yi
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215021, PR China.
| | - Duxin Li
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215021, PR China.
| | - Zhenqing Zhang
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215021, PR China.
| |
Collapse
|
3
|
Park HN, Song MJ, Choi YE, Lee DH, Chung JH, Lee ST. LRG1 Promotes ECM Integrity by Activating the TGF-β Signaling Pathway in Fibroblasts. Int J Mol Sci 2023; 24:12445. [PMID: 37569820 PMCID: PMC10418909 DOI: 10.3390/ijms241512445] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 08/01/2023] [Accepted: 08/03/2023] [Indexed: 08/13/2023] Open
Abstract
Leucine-rich alpha-2-glycoprotein 1 (LRG1) mediates skin repair and fibrosis by stimulating the transforming growth factor-beta (TGF-β) signaling pathway. In the present study, we investigated the effect of LRG1 on extracellular matrix (ECM) integrity in fibroblasts, as well as on skin aging. The treatment of dermal fibroblasts with purified recombinant human LRG1 increased type I collagen secretion and decreased matrix metalloproteinase-1 secretion. Additionally, LRG1 promoted SMAD2/SMAD3 phosphorylation in a pattern similar to that of TGF-β1 treatment. An inhibitor of TGF-β receptor 1 abolished LRG1-induced SMAD2 phosphorylation. RNA sequencing identified "extracellular region", "extracellular space", and "extracellular matrix" as the main Gene Ontology terms in the differentially expressed genes of fibroblasts treated with or without LRG1. LRG1 increased TGF-β1 mRNA levels, suggesting that LRG1 partially transactivates the expression of TGF-β1. Furthermore, an increased expression of type I collagen was also observed in fibroblasts grown in three-dimensional cultures on a collagen gel mimicking the dermis. LRG1 mRNA and protein levels were significantly reduced in elderly human skin tissues with weakened ECM integrity compared to in young human skin tissues. Taken together, our results suggest that LRG1 could retard skin aging by activating the TGF-β signaling pathway, increasing ECM deposition while decreasing its degradation.
Collapse
Affiliation(s)
- Han Na Park
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea; (H.N.P.); (Y.E.C.)
| | - Min Ji Song
- Department of Dermatology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; (M.J.S.); (D.H.L.); (J.H.C.)
| | - Young Eun Choi
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea; (H.N.P.); (Y.E.C.)
| | - Dong Hun Lee
- Department of Dermatology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; (M.J.S.); (D.H.L.); (J.H.C.)
- Laboratory of Cutaneous Aging Research, Biomedical Research Institute, Seoul National University Hospital, Seoul 03080, Republic of Korea
- Institute of Human-Environment Interface Biology, Seoul National University, Seoul 03080, Republic of Korea
| | - Jin Ho Chung
- Department of Dermatology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; (M.J.S.); (D.H.L.); (J.H.C.)
- Laboratory of Cutaneous Aging Research, Biomedical Research Institute, Seoul National University Hospital, Seoul 03080, Republic of Korea
- Institute of Human-Environment Interface Biology, Seoul National University, Seoul 03080, Republic of Korea
- Institute on Aging, Seoul National University, Seoul 03080, Republic of Korea
| | - Seung-Taek Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea; (H.N.P.); (Y.E.C.)
| |
Collapse
|
4
|
Ostrowska-Podhorodecka Z, Ali A, Norouzi M, Ding I, Abbasi S, Arora PD, Wong THF, Magalhaes M, McCulloch CA. Vimentin-mediated myosin 10 aggregation at tips of cell extensions drives MT1-MMP-dependent collagen degradation in colorectal cancer. FASEB J 2023; 37:e23097. [PMID: 37440280 DOI: 10.1096/fj.202300672r] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 06/09/2023] [Accepted: 07/03/2023] [Indexed: 07/14/2023]
Abstract
Colorectal cancer (CRC) is a high prevalence adenocarcinoma with progressive increases in metastasis-related mortality, but the mechanisms governing the extracellular matrix (ECM) degradation important for metastasis in CRC are not well-defined. We investigated a functional relationship between vimentin (Vim) and myosin 10 (Myo10), and whether this relationship is associated with cancer progression. We tested the hypothesis that Vim regulates the aggregation of Myo10 at the tips of cell extensions, which increases membrane-type 1 matrix metalloproteinase (MT1-MMP)-associated local collagen proteolysis and ECM degradation. Analysis of CRC samples revealed colocalization of Vim with Myo10 and MT1-MMP in cell extensions adjacent to sites of collagen degradation, suggesting an association with local cell invasion. We analyzed cultured CRC cells and fibroblasts and found that Vim accelerates aggregation of Myo10 at cell tips, which increases the cell extension rate. Vim stabilizes the interaction of Myo10 with MT1-MMP, which in turn increases collagenolysis. Vim depletion reduced the aggregation of Myo10 at the cell extension tips and MT1-MMP-dependent collagenolysis. We propose that Vim interacts with Myo10, which in turn associates with MT1-MMP to facilitate the transport of these molecules to the termini of cell extensions and there enhance cancer invasion of soft connective tissues.
Collapse
Affiliation(s)
| | - Aiman Ali
- Oral Pathology and Oral Medicine, Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Masoud Norouzi
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Isabel Ding
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Sevil Abbasi
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Pamma D Arora
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Timothy H F Wong
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Marco Magalhaes
- Oral Pathology and Oral Medicine, Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
- Cancer Invasion and Metastasis Laboratory, Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
- Dental and Maxillofacial Sciences Department, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | | |
Collapse
|
5
|
Ham SM, Song MJ, Yoon HS, Lee DH, Chung JH, Lee ST. SPARC Is Highly Expressed in Young Skin and Promotes Extracellular Matrix Integrity in Fibroblasts via the TGF-β Signaling Pathway. Int J Mol Sci 2023; 24:12179. [PMID: 37569556 PMCID: PMC10419001 DOI: 10.3390/ijms241512179] [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: 06/07/2023] [Revised: 07/18/2023] [Accepted: 07/28/2023] [Indexed: 08/13/2023] Open
Abstract
The matricellular secreted protein acidic and rich in cysteine (SPARC; also known as osteonectin), is involved in the regulation of extracellular matrix (ECM) synthesis, cell-ECM interactions, and bone mineralization. We found decreased SPARC expression in aged skin. Incubating foreskin fibroblasts with recombinant human SPARC led to increased type I collagen production and decreased matrix metalloproteinase-1 (MMP-1) secretion at the protein and mRNA levels. In a three-dimensional culture of foreskin fibroblasts mimicking the dermis, SPARC significantly increased the synthesis of type I collagen and decreased its degradation. In addition, SPARC also induced receptor-regulated SMAD (R-SMAD) phosphorylation. An inhibitor of transforming growth factor-beta (TGF-β) receptor type 1 reversed the SPARC-induced increase in type I collagen and decrease in MMP-1, and decreased SPARC-induced R-SMAD phosphorylation. Transcriptome analysis revealed that SPARC modulated expression of genes involved in ECM synthesis and regulation in fibroblasts. RT-qPCR confirmed that a subset of differentially expressed genes is induced by SPARC. These results indicated that SPARC enhanced ECM integrity by activating the TGF-β signaling pathway in fibroblasts. We inferred that the decline in SPARC expression in aged skin contributes to process of skin aging by negatively affecting ECM integrity in fibroblasts.
Collapse
Affiliation(s)
- Seung Min Ham
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea;
| | - Min Ji Song
- Department of Dermatology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; (M.J.S.); (H.-S.Y.); (D.H.L.); (J.H.C.)
- Laboratory of Cutaneous Aging Research, Biomedical Research Institute, Seoul National University Hospital, Seoul 03080, Republic of Korea
- Institute of Human-Environment Interface Biology, Seoul National University, Seoul 03080, Republic of Korea
| | - Hyun-Sun Yoon
- Department of Dermatology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; (M.J.S.); (H.-S.Y.); (D.H.L.); (J.H.C.)
- Laboratory of Cutaneous Aging Research, Biomedical Research Institute, Seoul National University Hospital, Seoul 03080, Republic of Korea
- Institute of Human-Environment Interface Biology, Seoul National University, Seoul 03080, Republic of Korea
- Department of Dermatology, Seoul National University Boramae Hospital, Seoul 07061, Republic of Korea
| | - Dong Hun Lee
- Department of Dermatology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; (M.J.S.); (H.-S.Y.); (D.H.L.); (J.H.C.)
- Laboratory of Cutaneous Aging Research, Biomedical Research Institute, Seoul National University Hospital, Seoul 03080, Republic of Korea
- Institute of Human-Environment Interface Biology, Seoul National University, Seoul 03080, Republic of Korea
| | - Jin Ho Chung
- Department of Dermatology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; (M.J.S.); (H.-S.Y.); (D.H.L.); (J.H.C.)
- Laboratory of Cutaneous Aging Research, Biomedical Research Institute, Seoul National University Hospital, Seoul 03080, Republic of Korea
- Institute of Human-Environment Interface Biology, Seoul National University, Seoul 03080, Republic of Korea
- Institute on Aging, Seoul National University, Seoul 03080, Republic of Korea
| | - Seung-Taek Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea;
| |
Collapse
|
6
|
Lee S, Choi YJ, Huo C, Alishir A, Kang KS, Park IH, Jang T, Kim KH. Laricitrin 3-Rutinoside from Ginkgo biloba Fruits Prevents Damage in TNF-α-Stimulated Normal Human Dermal Fibroblasts. Antioxidants (Basel) 2023; 12:1432. [PMID: 37507970 PMCID: PMC10376084 DOI: 10.3390/antiox12071432] [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: 06/30/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
Human skin comprises the epidermis and dermis, which perform interactive functional activities with each other in order to maintain the skin's tensile strength. In particular, the dermal layer is crucial for skin protection. However, skin aging destroys collagen and elastin fibers, causing wrinkles, pigments, and sagging. Skin aging-related factors, such as tumor necrosis factor-α (TNF-α), promote the generation of intercellular reactive oxygen species (ROS). These are known to stimulate the hypersecretion of matrix metalloproteinase-1 (MMP-1), which degrades collagen and inhibits collagen synthesis. In this study, as part of our ongoing discovery of natural products, we investigated potential natural products derived from ginkgo fruit (Ginkgo biloba fruit) with protective effects against TNF-α-induced skin aging. Phytochemical investigation of the MeOH extract of G. biloba fruits, aided by liquid chromatography-mass spectrometry, led to the isolation of 14 compounds (1-14) from the n-butanol-soluble fraction. These were structurally determined to be: (E)-coniferin (1), syringin (2), 4-hydroxybenzoic acid 4-O-β-D-glucopyranoside (3), vanillic acid 4-O-β-D-glucopyranoside (4), glucosyringic acid (5), (E)-ferulic acid 4-O-β-D-glucoside (6), (E)-sinapic acid 4-O-β-D-glucopyranoside (7), ginkgotoxin-5-glucoside (8), ginkgopanoside (9), (Z)-4-coumaric acid 4-O-β-D-glucopyranoside (10), (1'R,2'S,5'R,8'S,2'Z,4'E)-dihydrophaseic acid 3'-O-β-D-glucopyranoside (11), eucomic acid (12), rutin (13), and laricitrin 3-rutinoside (L3R) (14). Biological evaluation of the isolated compounds for their effects on intracellular ROS generation showed that, of these 14 compounds, L3R (14) inhibited TNF-α-stimulated ROS generation (p < 0.001 at 100 μM). Inhibition of ROS generation by L3R led to the suppression of MMP-1 secretion and protection against collagen degradation. The inhibitory effect of L3R was mediated by the inhibition of extracellular signal regulated kinase (ERK) phosphorylation. Furthermore, L3R diminished the secretion of pro-inflammatory cytokines interleukin 6 (IL-6) and interleukin 8 (IL-8). Based on these experimental results, L3R is a potential bioactive natural product that can be used to protect against skin damage, including aging, in cosmetics and pharmaceuticals.
Collapse
Affiliation(s)
- Sullim Lee
- Department of Life Science, College of Bio-Nano Technology, Gachon University, Seongnam 13120, Republic of Korea
| | - Yea Jung Choi
- College of Korean Medicine, Gachon University, Seongnam 13120, Republic of Korea
| | - Chen Huo
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Akida Alishir
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Ki Sung Kang
- College of Korean Medicine, Gachon University, Seongnam 13120, Republic of Korea
| | - Il-Ho Park
- College of Pharmacy, Sahmyook University, 815, Hwarang-ro, Nowon-gu, Seoul 01795, Republic of Korea
| | - Taesu Jang
- Health Administration, Dankook University, Cheonan 31116, Republic of Korea
| | - Ki Hyun Kim
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
7
|
Zhen AX, Piao MJ, Kang KA, Fernando PDSM, Herath HMUL, Cho SJ, Hyun JW. 3-Bromo-4,5-dihydroxybenzaldehyde Protects Keratinocytes from Particulate Matter 2.5-Induced Damages. Antioxidants (Basel) 2023; 12:1307. [PMID: 37372037 DOI: 10.3390/antiox12061307] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/13/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
Cellular senescence can be activated by several stimuli, including ultraviolet radiation and air pollutants. This study aimed to evaluate the protective effect of marine algae compound 3-bromo-4,5-dihydroxybenzaldehyde (3-BDB) on particulate matter 2.5 (PM2.5)-induced skin cell damage in vitro and in vivo. The human HaCaT keratinocyte was pre-treated with 3-BDB and then with PM2.5. PM2.5-induced reactive oxygen species (ROS) generation, lipid peroxidation, mitochondrial dysfunction, DNA damage, cell cycle arrest, apoptotic protein expression, and cellular senescence were measured using confocal microscopy, flow cytometry, and Western blot. The present study exhibited PM2.5-generated ROS, DNA damage, inflammation, and senescence. However, 3-BDB ameliorated PM2.5-induced ROS generation, mitochondria dysfunction, and DNA damage. Furthermore, 3-BDB reversed the PM2.5-induced cell cycle arrest and apoptosis, reduced cellular inflammation, and mitigated cellular senescence in vitro and in vivo. Moreover, the mitogen-activated protein kinase signaling pathway and activator protein 1 activated by PM2.5 were inhibited by 3-BDB. Thus, 3-BDB suppressed skin damage induced by PM2.5.
Collapse
Affiliation(s)
- Ao-Xuan Zhen
- Department of Biochemistry, College of Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Mei-Jing Piao
- Jeju Research Center for Natural Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Kyoung-Ah Kang
- Jeju Research Center for Natural Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | | | | | - Suk-Ju Cho
- Department of Anesthesiology, Jeju National University Hospital, College of Medicine, Jeju National University, Jeju 63241, Republic of Korea
| | - Jin-Won Hyun
- Department of Biochemistry, College of Medicine, Jeju National University, Jeju 63243, Republic of Korea
- Jeju Research Center for Natural Medicine, Jeju National University, Jeju 63243, Republic of Korea
| |
Collapse
|
8
|
Wu N, Liu H, Lv X, Sun Y, Jiang H. Neobaicalein prevents isoflurane anesthesia-induced cognitive impairment in neonatal mice via regulating CREB1. Clinics (Sao Paulo) 2023; 78:100201. [PMID: 37120983 PMCID: PMC10173397 DOI: 10.1016/j.clinsp.2023.100201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 03/24/2023] [Accepted: 04/04/2023] [Indexed: 05/02/2023] Open
Abstract
OBJECTIVES Isoflurane (ISO) is widely used in the clinic and research. The authors aimed to explore whether Neobaicalein (Neob) could protect neonatal mice from ISO-induced cognitive damage. METHOD The open field test, Morris water maze test, and tail suspension test was performed to assess the cognitive function in mice. Enzyme-linked immunosorbent assay was used to evaluate inflammatory-related protein concentrations. Immunohistochemistry was used to assess Ionized calcium-Binding Adapter molecule-1 (IBA-1) expression. Hippocampal neuron viability was detected using the Cell Counting Kit-8 assay. Double immunofluorescence staining was employed to confirm the interaction between proteins. Western blotting was used to assess protein expression levels. RESULTS Neob notably improved cognitive function and exhibited anti-inflammatory effects; moreover, under iso-treatment, it exhibited neuroprotective effects. Furthermore, Neob suppressed interleukin-1β, tumor necrosis factor-α, and interleukin-6 levels and upregulated interleukin-10 levels in ISO-treated mice. Neob significantly mitigated iso-induced increases in IBA-1-positive cell numbers of the hippocampus in neonatal mice. Furthermore, it inhibited ISO-induced neuronal apoptosis. Mechanistically, Neob was observed to upregulate cAMP Response Element Binding protein (CREB1) phosphorylation and protected hippocampal neurons from ISO-mediated apoptosis. Moreover, it rescued ISO-induced abnormalities of synaptic protein. CONCLUSIONS Neob prevented ISO anesthesia-induced cognitive impairment by suppressing apoptosis and inflammation through upregulating CREB1.
Collapse
Affiliation(s)
- Niming Wu
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hua Liu
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiang Lv
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Sun
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong Jiang
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
9
|
Wei B, Ji M, Lin Y, Wang S, Liu Y, Geng R, Hu X, Xu L, Li Z, Zhang W, Lu J. Mitochondrial transfer from bone mesenchymal stem cells protects against tendinopathy both in vitro and in vivo. Stem Cell Res Ther 2023; 14:104. [PMID: 37101277 PMCID: PMC10134653 DOI: 10.1186/s13287-023-03329-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 04/05/2023] [Indexed: 04/28/2023] Open
Abstract
BACKGROUND Although mesenchymal stem cells (MSCs) have been effective in tendinopathy, the mechanisms by which MSCs promote tendon healing have not been fully elucidated. In this study, we tested the hypothesis that MSCs transfer mitochondria to injured tenocytes in vitro and in vivo to protect against Achilles tendinopathy (AT). METHODS Bone marrow MSCs and H2O2-injured tenocytes were co-cultured, and mitochondrial transfer was visualized by MitoTracker dye staining. Mitochondrial function, including mitochondrial membrane potential, oxygen consumption rate, and adenosine triphosphate content, was quantified in sorted tenocytes. Tenocyte proliferation, apoptosis, oxidative stress, and inflammation were analyzed. Furthermore, a collagenase type I-induced rat AT model was used to detect mitochondrial transfer in tissues and evaluate Achilles tendon healing. RESULTS MSCs successfully donated healthy mitochondria to in vitro and in vivo damaged tenocytes. Interestingly, mitochondrial transfer was almost completely blocked by co-treatment with cytochalasin B. Transfer of MSC-derived mitochondria decreased apoptosis, promoted proliferation, and restored mitochondrial function in H2O2-induced tenocytes. A decrease in reactive oxygen species and pro-inflammatory cytokine levels (interleukin-6 and -1β) was observed. In vivo, mitochondrial transfer from MSCs improved the expression of tendon-specific markers (scleraxis, tenascin C, and tenomodulin) and decreased the infiltration of inflammatory cells into the tendon. In addition, the fibers of the tendon tissue were neatly arranged and the structure of the tendon was remodeled. Inhibition of mitochondrial transfer by cytochalasin B abrogated the therapeutic efficacy of MSCs in tenocytes and tendon tissues. CONCLUSIONS MSCs rescued distressed tenocytes from apoptosis by transferring mitochondria. This provides evidence that mitochondrial transfer is one mechanism by which MSCs exert their therapeutic effects on damaged tenocytes.
Collapse
Affiliation(s)
- Bing Wei
- School of Medicine, Southeast University, No. 87 Dingjiaqiao Road, Gulou District, Jiangsu Province, 210009, Nanjing, People's Republic of China
- Department of Orthopaedic Surgery/Joint and Sports Medicine Center, Zhongda Hospital, Southeast University, Nanjing, 210009, Jiangsu Province, People's Republic of China
| | - Mingliang Ji
- School of Medicine, Southeast University, No. 87 Dingjiaqiao Road, Gulou District, Jiangsu Province, 210009, Nanjing, People's Republic of China
- Department of Orthopaedic Surgery/Joint and Sports Medicine Center, Zhongda Hospital, Southeast University, Nanjing, 210009, Jiangsu Province, People's Republic of China
| | - Yucheng Lin
- School of Medicine, Southeast University, No. 87 Dingjiaqiao Road, Gulou District, Jiangsu Province, 210009, Nanjing, People's Republic of China
- Department of Orthopaedic Surgery/Joint and Sports Medicine Center, Zhongda Hospital, Southeast University, Nanjing, 210009, Jiangsu Province, People's Republic of China
| | - Shanzheng Wang
- School of Medicine, Southeast University, No. 87 Dingjiaqiao Road, Gulou District, Jiangsu Province, 210009, Nanjing, People's Republic of China
- Department of Orthopaedic Surgery/Joint and Sports Medicine Center, Zhongda Hospital, Southeast University, Nanjing, 210009, Jiangsu Province, People's Republic of China
| | - Yuxi Liu
- School of Medicine, Southeast University, No. 87 Dingjiaqiao Road, Gulou District, Jiangsu Province, 210009, Nanjing, People's Republic of China
- Department of Orthopaedic Surgery/Joint and Sports Medicine Center, Zhongda Hospital, Southeast University, Nanjing, 210009, Jiangsu Province, People's Republic of China
| | - Rui Geng
- School of Medicine, Southeast University, No. 87 Dingjiaqiao Road, Gulou District, Jiangsu Province, 210009, Nanjing, People's Republic of China
- Department of Orthopaedic Surgery/Joint and Sports Medicine Center, Zhongda Hospital, Southeast University, Nanjing, 210009, Jiangsu Province, People's Republic of China
| | - Xinyue Hu
- School of Medicine, Southeast University, No. 87 Dingjiaqiao Road, Gulou District, Jiangsu Province, 210009, Nanjing, People's Republic of China
- Department of Orthopaedic Surgery/Joint and Sports Medicine Center, Zhongda Hospital, Southeast University, Nanjing, 210009, Jiangsu Province, People's Republic of China
| | - Li Xu
- School of Medicine, Southeast University, No. 87 Dingjiaqiao Road, Gulou District, Jiangsu Province, 210009, Nanjing, People's Republic of China
- Department of Orthopaedic Surgery/Joint and Sports Medicine Center, Zhongda Hospital, Southeast University, Nanjing, 210009, Jiangsu Province, People's Republic of China
| | - Zhuang Li
- School of Medicine, Southeast University, No. 87 Dingjiaqiao Road, Gulou District, Jiangsu Province, 210009, Nanjing, People's Republic of China
- Department of Orthopaedic Surgery/Joint and Sports Medicine Center, Zhongda Hospital, Southeast University, Nanjing, 210009, Jiangsu Province, People's Republic of China
| | - Weituo Zhang
- School of Medicine, Southeast University, No. 87 Dingjiaqiao Road, Gulou District, Jiangsu Province, 210009, Nanjing, People's Republic of China
- Department of Orthopaedic Surgery/Joint and Sports Medicine Center, Zhongda Hospital, Southeast University, Nanjing, 210009, Jiangsu Province, People's Republic of China
| | - Jun Lu
- School of Medicine, Southeast University, No. 87 Dingjiaqiao Road, Gulou District, Jiangsu Province, 210009, Nanjing, People's Republic of China.
- Department of Orthopaedic Surgery/Joint and Sports Medicine Center, Zhongda Hospital, Southeast University, Nanjing, 210009, Jiangsu Province, People's Republic of China.
| |
Collapse
|
10
|
Jeong HD, Kim JH, Kwon GE, Lee ST. Expression of Polyamine Oxidase in Fibroblasts Induces MMP-1 and Decreases the Integrity of Extracellular Matrix. Int J Mol Sci 2022; 23:ijms231810487. [PMID: 36142401 PMCID: PMC9504367 DOI: 10.3390/ijms231810487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/02/2022] [Accepted: 09/07/2022] [Indexed: 11/16/2022] Open
Abstract
Polyamine oxidase (PAOX) (N1-acetylpolyamine oxidase) is a major enzyme in the polyamine catabolism pathway that generates hydrogen peroxide. Hydrogen peroxide plays a crucial role in skin aging via extracellular matrix (ECM) degradation by increasing the matrix metalloproteinase-1 (MMP-1) levels. We analyzed the integrity of the ECM in foreskin fibroblasts using PAOX expression. PAOX increased the MMP-1 secretion and type Ι collagen degradation in 2D and 3D cultures of fibroblasts, respectively. Similarly, PAOX overexpression increased the messenger ribonucleic acid (mRNA) level of MMP-1. PAOX expression induced polyamine catabolism, decreased the spermine levels, and increased the putrescine levels. However, the exogenous polyamine treatment did not change the MMP-1 and type I collagen levels as much as PAOX expression. PAOX expression increased the reactive oxygen species (ROS) production in fibroblasts, and exogenous hydrogen peroxide increased both the ROS production and MMP-1 secretion. Furthermore, N-acetylcysteine, an antioxidant, reversed the PAOX-induced ROS production and MMP-1 secretion. PAOX induced the signaling pathways that activate activator protein-1 (AP-1) and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), which are important transcription factors for MMP-1 transactivation. We concluded that PAOX increased the ROS levels in fibroblasts, leading to an increase in MMP-1 expression. Therefore, we propose that PAOX is a potential target molecule in protecting the ECM integrity.
Collapse
Affiliation(s)
- Hae Dong Jeong
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
| | - Jin Hyung Kim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
| | - Go Eun Kwon
- Molecular Recognition Research Center, Korea Institute of Science and Technology, Seoul 02792, Korea
| | - Seung-Taek Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
- Correspondence: ; Tel.: +82-221232703
| |
Collapse
|
11
|
Kim JH, Lee ST. Polyamine Oxidase Expression Is Downregulated by 17β-Estradiol via Estrogen Receptor 2 in Human MCF-7 Breast Cancer Cells. Int J Mol Sci 2022; 23:ijms23147521. [PMID: 35886868 PMCID: PMC9317983 DOI: 10.3390/ijms23147521] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/02/2022] [Accepted: 07/04/2022] [Indexed: 02/05/2023] Open
Abstract
Polyamine levels decrease with menopause; however, little is known about the mechanisms regulated by menopause. In this study, we found that among the genes involved in the polyamine pathway, polyamine oxidase (PAOX) mRNA levels were the most significantly reduced by treatment with 17β-estradiol in estrogen receptor (ESR)-positive MCF-7 breast cancer cells. Treatment with 17β-estradiol also reduced the PAOX protein levels. Treatment with selective ESR antagonists and knockdown of ESR members revealed that estrogen receptor 2 (ESR2; also known as ERβ) was responsible for the repression of PAOX by 17β-estradiol. A luciferase reporter assay showed that 17β-estradiol downregulates PAOX promoter activity and that 17β-estradiol-dependent PAOX repression disappeared after deletions (−3126/−2730 and −1271/−1099 regions) or mutations of activator protein 1 (AP-1) binding sites in the PAOX promoter. Chromatin immunoprecipitation analysis showed that ESR2 interacts with AP-1 bound to each of the two AP-1 binding sites. These results demonstrate that 17β-estradiol represses PAOX transcription by the interaction of ESR2 with AP-1 bound to the PAOX promoter. This suggests that estrogen deficiency may upregulate PAOX expression and decrease polyamine levels.
Collapse
|
12
|
Chen YP, Wang KX, Cai JQ, Li Y, Yu HL, Wu Q, Meng W, Wang H, Yin CH, Wu J, Huang MB, Li R, Guan DG. Detecting Key Functional Components Group and Speculating the Potential Mechanism of Xiao-Xu-Ming Decoction in Treating Stroke. Front Cell Dev Biol 2022; 10:753425. [PMID: 35646921 PMCID: PMC9136080 DOI: 10.3389/fcell.2022.753425] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 02/25/2022] [Indexed: 02/05/2023] Open
Abstract
Stroke is a cerebrovascular event with cerebral blood flow interruption which is caused by occlusion or bursting of cerebral vessels. At present, the main methods in treating stroke are surgical treatment, statins, and recombinant tissue-type plasminogen activator (rt-PA). Relatively, traditional Chinese medicine (TCM) has widely been used at clinical level in China and some countries in Asia. Xiao-Xu-Ming decoction (XXMD) is a classical and widely used prescription in treating stroke in China. However, the material basis of effect and the action principle of XXMD are still not clear. To solve this issue, we designed a new system pharmacology strategy that combined targets of XXMD and the pathogenetic genes of stroke to construct a functional response space (FRS). The effective proteins from this space were determined by using a novel node importance calculation method, and then the key functional components group (KFCG) that could mediate the effective proteins was selected based on the dynamic programming strategy. The results showed that enriched pathways of effective proteins selected from FRS could cover 99.10% of enriched pathways of reference targets, which were defined by overlapping of component targets and pathogenetic genes. Targets of optimized KFCG with 56 components can be enriched into 166 pathways that covered 80.43% of 138 pathways of 1,012 pathogenetic genes. A component potential effect score (PES) calculation model was constructed to calculate the comprehensive effective score of components in the components-targets-pathways (C-T-P) network of KFCGs, and showed that ferulic acid, zingerone, and vanillic acid had the highest PESs. Prediction and docking simulations show that these components can affect stroke synergistically through genes such as MEK, NFκB, and PI3K in PI3K-Akt, cAMP, and MAPK cascade signals. Finally, ferulic acid, zingerone, and vanillic acid were tested to be protective for PC12 cells and HT22 cells in increasing cell viabilities after oxygen and glucose deprivation (OGD). Our proposed strategy could improve the accuracy on decoding KFCGs of XXMD and provide a methodologic reference for the optimization, mechanism analysis, and secondary development of the formula in TCM.
Collapse
Affiliation(s)
- Yu-peng Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China,Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, China
| | - Ke-xin Wang
- Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, National Key Clinical Specialty/Engineering Technology Research Center of Education Ministry of China, Neurosurgery Institute, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jie-qi Cai
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China,Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, China
| | - Yi Li
- Department of Radiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hai-lang Yu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China,Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, China
| | - Qi Wu
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wei Meng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China,Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, China
| | - Handuo Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China,Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, China
| | - Chuan-hui Yin
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China,Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, China
| | - Jie Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China,Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, China
| | - Mian-bo Huang
- Department of Histology and Embryology, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China,*Correspondence: Mian-bo Huang, ; Rong Li, ; Dao-gang Guan,
| | - Rong Li
- Department of Cardiovascular Disease, First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China,*Correspondence: Mian-bo Huang, ; Rong Li, ; Dao-gang Guan,
| | - Dao-gang Guan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China,Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, China,*Correspondence: Mian-bo Huang, ; Rong Li, ; Dao-gang Guan,
| |
Collapse
|
13
|
Physicochemical and Anti-UVB-Induced Skin Inflammatory Properties of Lacticaseibacillus paracasei Subsp. paracasei SS-01 Strain Exopolysaccharide. FERMENTATION 2022. [DOI: 10.3390/fermentation8050198] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The exopolysaccharide secreted by Lacticaseibacillus paracasei subsp. paracasei SS-01 strain (LP-EPS) is isolated and purified from yogurt. It is a polysaccharide with a branched and multi-stranded structure, which exists in a smooth rod-like or cloud-like state, and possesses a good thermal stability and a molecular weight of 49.68 kDa (±4.436%). LP-EPS shows a high antioxidant capacity, anti-inflammatory and anti-sensitizing activity during in vitro experimental studies, with half clearance (IC50) rates of 0.449, 1.314, and 2.369 mg/mL for the ABTS, DPPH, and OH radicals, respectively, and a half inhibition rate (IC50) of hyaluronidase of 1.53 mg/mL. A cell-based assay, enzyme-linked immunosorbent assay (ELISA), and quantitative real-time fluorescence PCR (qRT-PCR) show that LP-EPS effectively treats or ameliorates the skin inflammatory responses triggered by UVB irradiation, as evidenced by a highly significant decrease in the secretion of inflammatory factors by human skin keratinocytes (HaCaT), and a highly significant downregulation of the mRNA expression of MAPK/AP-1 pathway cytokines.
Collapse
|
14
|
Ostrowska-Podhorodecka Z, Ding I, Norouzi M, McCulloch CA. Impact of Vimentin on Regulation of Cell Signaling and Matrix Remodeling. Front Cell Dev Biol 2022; 10:869069. [PMID: 35359446 PMCID: PMC8961691 DOI: 10.3389/fcell.2022.869069] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 02/25/2022] [Indexed: 12/12/2022] Open
Abstract
Vimentin expression contributes to cellular mechanoprotection and is a widely recognized marker of fibroblasts and of epithelial-mesenchymal transition. But it is not understood how vimentin affects signaling that controls cell migration and extracellular matrix (ECM) remodeling. Recent data indicate that vimentin controls collagen deposition and ECM structure by regulating contractile force application to the ECM and through post-transcriptional regulation of ECM related genes. Binding of cells to the ECM promotes the association of vimentin with cytoplasmic domains of adhesion receptors such as integrins. After initial adhesion, cell-generated, myosin-dependent forces and signals that impact vimentin structure can affect cell migration. Post-translational modifications of vimentin determine its adaptor functions, including binding to cell adhesion proteins like paxillin and talin. Accordingly, vimentin regulates the growth, maturation and adhesive strength of integrin-dependent adhesions, which enables cells to tune their attachment to collagen, regulate the formation of cell extensions and control cell migration through connective tissues. Thus, vimentin tunes signaling cascades that regulate cell migration and ECM remodeling. Here we consider how specific properties of vimentin serve to control cell attachment to the underlying ECM and to regulate mesenchymal cell migration and remodeling of the ECM by resident fibroblasts.
Collapse
|
15
|
PTK7, a Catalytically Inactive Receptor Tyrosine Kinase, Increases Oncogenic Phenotypes in Xenograft Tumors of Esophageal Squamous Cell Carcinoma KYSE-30 Cells. Int J Mol Sci 2022; 23:ijms23042391. [PMID: 35216506 PMCID: PMC8876147 DOI: 10.3390/ijms23042391] [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: 01/27/2022] [Revised: 02/16/2022] [Accepted: 02/19/2022] [Indexed: 12/24/2022] Open
Abstract
Protein tyrosine kinase 7 (PTK7), a catalytically defective receptor protein tyrosine kinase, is upregulated in tumor tissues and cell lines of esophageal squamous cell carcinoma (ESCC). We showed that PTK7 plays an oncogenic role in various ESCC cell lines. However, its role as an oncogene has not been demonstrated in vivo. Here, we examined the influence of PTK7 on the tumorigenic potential of ESCC KYSE-30 cells, which are known to establish xenograft tumors. Overexpression of PTK7 enhanced the proliferation, adhesion, wound healing, and migration of KYSE-30 cells, and these effects were reversed by the knockdown of PTK7. PTK7 overexpression and knockdown, respectively, increased and decreased the tyrosine phosphorylation of cellular proteins and the phosphorylation of ERK, AKT, and FAK, which are important for cell proliferation, survival, adhesion, and migration. Additionally, PTK7 overexpression and silencing, respectively, increased and decreased the weight, volume, and number of Ki-67-positive proliferating cells in xenograft tumors of KYSE-30 cells. Therefore, we propose that PTK7 plays an important role in the tumorigenesis of ESCC cells in vivo and is a potential therapeutic target for ESCC.
Collapse
|
16
|
Skullcapflavone II Suppresses TNF-α/IFN-γ-Induced TARC, MDC, and CTSS Production in HaCaT Cells. Int J Mol Sci 2021; 22:ijms22126428. [PMID: 34208434 PMCID: PMC8233710 DOI: 10.3390/ijms22126428] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/07/2021] [Accepted: 06/10/2021] [Indexed: 12/29/2022] Open
Abstract
Skullcapflavone II (SFII), a flavonoid derived from Scutellaria baicalensis, has been reported to have anti-inflammatory properties. However, its therapeutic potential for skin inflammatory diseases and its mechanism are unknown. Therefore, this study aimed to investigate the effect of SFII on TNF-α/IFN-γ-induced atopic dermatitis (AD)-associated cytokines, such as thymus- and activation-regulated chemokine (TARC) and macrophage-derived chemokine (MDC). Co-stimulation with TNF-α/IFN-γ in HaCaT cells is a well-established model for induction of pro-inflammatory cytokines. We treated cells with SFII prior to TNF-α/IFN-γ-stimulation and confirmed that it significantly inhibited TARC and MDC expression at the mRNA and protein levels. Additionally, SFII also inhibited the expression of cathepsin S (CTSS), which is associated with itching in patients with AD. Using specific inhibitors, we demonstrated that STAT1, NF-κB, and p38 MAPK mediate TNF-α/IFN-γ-induced TARC and MDC, as well as CTSS expression. Finally, we confirmed that SFII significantly suppressed TNF-α/IFN-γ-induced phosphorylation of STAT1, NF-κB, and p38 MAPK. Taken together, our study indicates that SFII inhibits TNF-α/IFN-γ-induced TARC, MDC, and CTSS expression by regulating STAT1, NF-κB, and p38 MAPK signaling pathways.
Collapse
|
17
|
A Network Pharmacological Approach to Reveal the Pharmacological Targets and Its Associated Biological Mechanisms of Prunetin-5-O-Glucoside against Gastric Cancer. Cancers (Basel) 2021; 13:cancers13081918. [PMID: 33921173 PMCID: PMC8071515 DOI: 10.3390/cancers13081918] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/12/2021] [Accepted: 04/13/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Identification of pharmacological targets in cancer provides a major walkthrough toward treatment strategies. The present research adopted a network pharmacology approach utilizing a flavonoid glucoside prunetin-5-O-glucoside (PG) compound against gastric cancer. The correlative targets were analyzed using Swiss target prediction and DiGeNET databases. Functional enrichment and significant pathways enriched were predicted for the targets to associate its biological mechanisms with cancer. Protein interaction network and cluster analysis was performed using Search Tool for the Retrieval of Interacting Genes/Proteins (STRING). Our analysis revealed three core targets among the clustered modules that plays a crucial role in relation with cancer. With this information, the core targets were examined for the binding affinity with PG using molecular docking analysis and validations on the protein targets was performed using western blot analysis and Human Protein Atlas. Our analysis through comprehensive network pharmacology resulted in the prediction of three core targets of PG that can be significant biomarkers against gastric cancer. Abstract Gastric cancer (GC) is an aggressive malignancy with increased mortality rate and low treatment options. Increasing evidence suggests that network pharmacology will be a novel method for identifying the systemic mechanism of therapeutic compounds in diseases like cancer. The current study aimed to use a network pharmacology approach to establish the predictive targets of prunetin-5-O-glucoside (PG) against gastric cancer and elucidate its biological mechanisms. Primarily, genes associated with the pathogenesis of GC was identified from the DiGeNET database and targets of PG was obtained from the Swiss target prediction database. In total, 65 correlative hits were identified as anti-gastric cancer targets of PG. Functional enrichment and pathway analysis revealed significant biological mechanisms of the targets. Interaction of protein network and cluster analysis using STRING resulted in three crucial interacting hub targets namely, HSP90AA1, CDK2, and MMP1. Additionally, the in vitro cytotoxic potential of PG was assessed on three gastric cancer cells (AGS, MKN-28, and SNU-484). Furthermore, the crucial targets were validated using molecular docking, followed by their expressions being evaluated by western blot and Human Protein Atlas. The findings indicate that the pharmacological action of PG against GC might be associated with the regulation of three core targets: HSP90AA1, CDK2, and MMP1. Thus, the network pharmacology undertaken in the current study established the core active targets of PG, which may be extensively applied with further validations for treatment in GC.
Collapse
|
18
|
Zglińska K, Niemiec T, Łozicki A, Matusiewicz M, Szczepaniak J, Puppel K, Kutwin M, Jaworski S, Rygało-Galewska A, Koczoń P. Effect of Elaeagnus umbellata (Thunb.) fruit extract on H 2O 2-induced oxidative and inflammatory responses in normal fibroblast cells. PeerJ 2021; 9:e10760. [PMID: 33552740 PMCID: PMC7821754 DOI: 10.7717/peerj.10760] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 12/22/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Elaeagnus umbellata is a plant commonly used in traditional Asian medicine for its many health benefits and strong antioxidative activity. Its therapeutic potential is believed to be connected to its effect on fibroblasts. This study aimed to investigate E. umbellata methanol-acetone extract's (EUE) defense against hydrogen peroxide (H2O2)-induced fibroblast damage. METHODS Because the main biologically active compounds of E. umbellata are water-insoluble, we evaluated the effects of methanol-acetone fruit extracts using liquid chromatography (for ascorbic acid and beta-carotene) and spectrophotometry (for lycopene and total phenolics). The extract's antioxidative activity was measured using DPPH radical inhibition, and EUE's effect on human fibroblasts was also evaluated. We assessed the metabolic activity and apoptosis of HFFF-2 fibroblasts exposed to EUE and/or H2O2using the XTT test and flow cytometry, respectively. Superoxide dismutase activity and reactive oxygen species (ROS) production were evaluated using colorimetric and fluorometric assays, respectively. We measured pro-inflammatory cytokine (MIF, fractalkine, MCP-4, BLC, GCP-2, NAP-2, Eotaxin-2, and Eotaxin-3) expression in HFFF-2 cells using immunocytochemistry. RESULT The extract increased HFFF-2 cell proliferation and reduced cell death caused by H2O2-induced stress. H2O2-treated fibroblasts had greater ROS production than cells treated with both H2O2 and EUE. Additionally, the group treated with H2O2 alone showed higher pro-inflammatory cytokine (MIF, MCP-4, NAP-2, Eotaxin-2, and Eotaxin-3) expression. CONCLUSION EUE protected human fibroblasts from H2O2-induced oxidative stress and reduced the fibroblast-mediated inflammatory response triggered by ROS.
Collapse
Affiliation(s)
- Klara Zglińska
- Institute of Animal Science, Warsaw University of Life Sciences, Warsaw, Poland
| | - Tomasz Niemiec
- Institute of Animal Science, Warsaw University of Life Sciences, Warsaw, Poland
| | - Andrzej Łozicki
- Institute of Animal Science, Warsaw University of Life Sciences, Warsaw, Poland
| | | | | | - Kamila Puppel
- Institute of Animal Science, Warsaw University of Life Sciences, Warsaw, Poland
| | - Marta Kutwin
- Institute of Biology, Warsaw Univeristy of Life Sciences, Warsaw, Poland
| | - Slawomir Jaworski
- Institute of Biology, Warsaw Univeristy of Life Sciences, Warsaw, Poland
| | | | - Piotr Koczoń
- Institute of Food Science, Warsaw University of Life Sciences, Warsaw, Poland
| |
Collapse
|
19
|
Choi YE, Song MJ, Hara M, Imanaka-Yoshida K, Lee DH, Chung JH, Lee ST. Effects of Tenascin C on the Integrity of Extracellular Matrix and Skin Aging. Int J Mol Sci 2020; 21:ijms21228693. [PMID: 33217999 PMCID: PMC7698786 DOI: 10.3390/ijms21228693] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/14/2020] [Accepted: 11/16/2020] [Indexed: 12/17/2022] Open
Abstract
Tenascin C (TNC) is an element of the extracellular matrix (ECM) of various tissues, including the skin, and is involved in modulating ECM integrity and cell physiology. Although skin aging is apparently associated with changes in the ECM, little is known about the role of TNC in skin aging. In this study, we found that the Tnc mRNA level was significantly reduced in the skin tissues of aged mice compared with young mice, consistent with reduced TNC protein expression in aged human skin. TNC-large (TNC-L; 330-kDa) and -small (TNC-S; 240-kDa) polypeptides were observed in conditional media from primary dermal fibroblasts. Both recombinant TNC polypeptides, corresponding to TNC-L and TNC-S, increased the expression of type I collagen and reduced the expression of matrix metalloproteinase-1 in fibroblasts. Treatment of fibroblasts with a recombinant TNC polypeptide, corresponding to TNC-L, induced phosphorylation of SMAD2 and SMAD3. TNC increased the level of transforming growth factor-β1 (TGF-β1) mRNA and upregulated the expression of type I collagen by activating the TGF-β signaling pathway. In addition, TNC also promoted the expression of type I collagen in fibroblasts embedded in a three-dimensional collagen matrix. Our findings suggest that TNC contributes to the integrity of ECM in young skin and to prevention of skin aging.
Collapse
Affiliation(s)
- Young Eun Choi
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea;
| | - Min Ji Song
- Department of Dermatology, Seoul National University College of Medicine, Seoul 03080, Korea; (M.J.S.); (D.H.L.); (J.H.C.)
| | - Mari Hara
- Department of Pathology and Matrix Biology, Mie University Graduate School of Medicine, Tsu 514-8507, Japan; (M.H.); (K.I.-Y.)
| | - Kyoko Imanaka-Yoshida
- Department of Pathology and Matrix Biology, Mie University Graduate School of Medicine, Tsu 514-8507, Japan; (M.H.); (K.I.-Y.)
- Mie University Research Center for Matrix Biology, Tsu 514-8507, Japan
| | - Dong Hun Lee
- Department of Dermatology, Seoul National University College of Medicine, Seoul 03080, Korea; (M.J.S.); (D.H.L.); (J.H.C.)
- Institute of Human-Environment Interface Biology, Medical Research Center, Seoul National University, Seoul 03080, Korea
| | - Jin Ho Chung
- Department of Dermatology, Seoul National University College of Medicine, Seoul 03080, Korea; (M.J.S.); (D.H.L.); (J.H.C.)
- Institute of Human-Environment Interface Biology, Medical Research Center, Seoul National University, Seoul 03080, Korea
| | - Seung-Taek Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea;
- Correspondence: ; Tel.: +82-2-2123-2703
| |
Collapse
|
20
|
Parsafar S, Nayeri Z, Aliakbari F, Shahi F, Mohammadi M, Morshedi D. Multiple neuroprotective features of Scutellaria pinnatifida-derived small molecule. Heliyon 2020; 6:e04737. [PMID: 32913905 PMCID: PMC7472859 DOI: 10.1016/j.heliyon.2020.e04737] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 06/29/2020] [Accepted: 08/12/2020] [Indexed: 12/11/2022] Open
Abstract
Parkinson's disease (PD) is one of the most prevalent neurodegenerative disorders with no precise etiology. Multiple lines of evidence support that environmental factors, either neurotoxins or neuroinflammation, can induce Parkinsonism. In this study, we purified an active compound, neobaicalein (Skullcapflavone II), from the roots of Scutellaria pinnatifida (S. pinnatifida). Neobaicalein not only had protective impacts on rotenone-induced neurotoxicity but in glial cultures, it dampened the inflammatory response when stimulated with lipopolysaccharide (LPS). Neobaicalein had high antioxidant activity without any obvious toxicity. In addition, it could raise the cell viability, decrease early apoptosis, reduce the generation of reactive oxygen species (ROS), and keep the neurite's length normal in the treated SH-SY5Y cells. Pathway enrichment analysis (PEA) and target prediction provided insights into the PD related genes, protein-protein interaction (PPI) network, and the key proteins enriched in the signaling pathways. Furthermore, docking simulation (DS) on the proteins of the PD-PPI network revealed that neobaicalein might interact with the key proteins involved in PD pathology, including MAPK14, MAPK8, and CASP3. It also blocks the destructive processes, such as cell death, inflammation, and oxidative stress pathways. Our results demonstrate that neobaicalein alleviates pathological effects of factors related to PD, and may provide new insight into PD therapy.
Collapse
Affiliation(s)
- Soha Parsafar
- Department of Bioprocess Engineering, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Zahra Nayeri
- Department of Bioprocess Engineering, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Farhang Aliakbari
- Department of Bioprocess Engineering, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Farshad Shahi
- Department of Bioprocess Engineering, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Mehdi Mohammadi
- Department of Bioprocess Engineering, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Dina Morshedi
- Department of Bioprocess Engineering, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| |
Collapse
|
21
|
Zhang L, Bian YF, Bai R, Song XS, Liang B, Xiao CS. Circ_BMP2K enhances the regulatory effects of miR-455-3p on its target gene SUMO1 and thereby inhibits the activation of cardiac fibroblasts. Biochem Cell Biol 2020; 98:583-590. [PMID: 32413267 DOI: 10.1139/bcb-2019-0381] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Research has shown that some circular RNAs (circRNA) are abnormally expressed in the process of myocardial fibrosis, but the mechanism behind this was unknown. In the process of inducing cardiac fibroblast (CF) activation with TGF-β1 or Ang II, the expression of circRNA circ_BMP2K and miR-455-3p were significantly inhibited, whereas the expression of SUMO1 was promoted. The results from our dual luciferase reporter gene assays, RIP assays, and pull-down assays show that miR-455-3p directly binds circ_BMP2K, thereby mutually promoting their expression levels. SUMO1 is a target gene of miR-455-3p, and circ_BMP2K enhances the inhibitory effects of miR-455-3p on the expression of SUMO1. Further study showed that both circ_BMP2K and miR-455-3p inhibited the expression of alpha-SMA as well as type I and type III collagen, whereas SUMO1 promoted their expression, and miR-455-3p inhibitors or overexpression of SUMO1 reversed the effects of circ_BMP2K and miR-455-3p. Circ_BMP2K and miR-455-3p inhibits cell proliferation and migration and promotes the apoptosis of CFs, but SUMO1 has the opposite effects; miR-455-3p inhibitors or overexpression of SUMO1 reverses the effects of circ_BMP2K and miR-455-3p. Thus, circ_BMP2K promotes the expression of miR-455-3p, down-regulates the expression of SUMO1, and finally, inhibits the activation, growth, and migration of CFs. These results could provide important therapeutic targets and a theoretical basis for regulating the process of myocardial fibrosis.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Cardiology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan 030032, P.R. China
| | - Yun-Fei Bian
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan 030001, P.R. China
| | - Rui Bai
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan 030001, P.R. China
| | - Xiao-Sun Song
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan 030001, P.R. China
| | - Bin Liang
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan 030001, P.R. China
| | - Chuan-Shi Xiao
- Department of Cardiology, The First Hospital of Shanxi Medical University, Taiyuan 030001, P.R. China
| |
Collapse
|
22
|
Shin WS, Lee HW, Lee ST. Catalytically inactive receptor tyrosine kinase PTK7 activates FGFR1 independent of FGF. FASEB J 2019; 33:12960-12971. [PMID: 31490704 PMCID: PMC6902674 DOI: 10.1096/fj.201900932r] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Protein tyrosine kinase 7 (PTK7), a catalytically defective receptor protein tyrosine kinase (RPTK), plays an oncogenic role by activating an unidentified TKI-258 (dovitinib)-sensitive RPTK in esophageal squamous cell carcinoma (ESCC) cells. Here, we demonstrate that among TKI-258–sensitive RPTKs, fibroblast growth factor receptor (FGFR) 1 is significantly up-regulated in ESCC tissues and cell lines. We show that PTK7 colocalizes with FGFR1 and binds it via its extracellular domain in human embryonic kidney 293 and ESCC TE-10 cells. PTK7 knockdown not only reduced ligand-free and fibroblast growth factor (FGF)-induced phosphorylation of FGFR1 but also the interaction of signaling adaptor proteins with FGFR1 and activation of downstream signaling proteins in TE-10 cells. In addition, PTK7 knockdown reduced FGF-induced oncogenic phenotypes including proliferation, anchorage-independent colony formation, wound healing, and invasion in ESCC cells. Taken together, our data demonstrate that PTK7 binds and activates FGFR1 independent of FGF and thus increases oncogenicity of PTK7- and FGFR1-positive cancers such as ESCC.—Shin, W.-S., Lee, H. W., Lee, S.-T. Catalytically inactive receptor tyrosine kinase PTK7 activates FGFR1 independent of FGF.
Collapse
Affiliation(s)
- Won-Sik Shin
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Hae Won Lee
- Department of Thoracic Surgery, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| | - Seung-Taek Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| |
Collapse
|