1
|
Li S, Li G, Li X, Wu F, Li L. Etanercept ameliorates psoriasis progression through regulating high mobility group box 1 pathway. Skin Res Technol 2023; 29:e13329. [PMID: 37113086 PMCID: PMC10234177 DOI: 10.1111/srt.13329] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 04/04/2023] [Indexed: 04/29/2023]
Abstract
BACKGROUND As a common skin disease, psoriasis is related to inflammation and immune response. Due to the frequent recurrence of psoriasis, the treatment of psoriasis remains a clinical challenge. As an effective tumor necrosis factor-alpha (TNF-α) inhibitor, etanercept has been used for the treatment of psoriasis. However, some patients with psoriasis have no response to etanercept or discontinue treatment. To improve the therapeutic effect of etanercept, searching the potential biomarkers and investigating the related mechanisms of etanercept in the treatment of psoriasis are vital. MATERIALS AND METHODS We stimulated HaCaT cells with lipopolysaccharide (LPS) to generate cellular psoriatic changes and established an imiquimod (IMQ)-induced psoriasis-like mouse model, and then used an etanercept to treat cell and mouse model. RESULTS Etanercept alleviated IMQ-induced pathological changes and inflammation, and it also decreased the protein expression of high mobility group box 1 (HMGB1), receptor for advanced glycation end-products, and toll-like receptor 4. Moreover, the results of in vitro experiments showed that etanercept inhibited proliferation and inflammation, and promoted cell cycle arrest and apoptosis in LPS-treated HaCaT cells. Knockdown of HMGB1 further enhanced the inhibitory effects of etanercept on LPS-treated HaCaT cell viability and inflammation, while overexpression of HMGB1 notably reversed the inhibitory effects of etanercept on LPS-induced HaCaT cell viability and inflammation. CONCLUSION Etanercept inhibited proliferation and inflammation and promoted cell cycle arrest and apoptosis in LPS-induced HaCaT cells, and etanercept ameliorated inflammation in a psoriasis-like mouse model.
Collapse
Affiliation(s)
- Shu Li
- Department of DermatologyTaizhou People's HospitalTaizhouP. R. China
| | - Guangli Li
- Internal Medicine DepartmentFushun Maternal and Child Health HospitalFushunP. R. China
| | - Xiaoyan Li
- Department of DermatologyLianshui County People's HospitalHuai 'anP. R. China
| | - Fan Wu
- Department of DermatologyLianshui County People's HospitalHuai 'anP. R. China
| | - Ling Li
- Department of DermatologyLianshui County People's HospitalHuai 'anP. R. China
| |
Collapse
|
2
|
Shin KK, Park SH, Lim HY, Lorza LR, Qomaladewia NP, You L, Aziz N, Kim SA, Lee JS, Choung ES, Noh JK, Yie DK, Jeong D, Lee J, Cho JY. In Vitro Anti-Photoaging and Skin Protective Effects of Licania macrocarpa Cuatrec Methanol Extract. PLANTS (BASEL, SWITZERLAND) 2022; 11:1383. [PMID: 35631808 PMCID: PMC9144732 DOI: 10.3390/plants11101383] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/16/2022] [Accepted: 05/18/2022] [Indexed: 12/24/2022]
Abstract
The Licania genus has been used in the treatment of dysentery, diabetes, inflammation, and diarrhea in South America. Of these plants, the strong anti-inflammatory activity of Licania macrocarpa Cuatrec (Chrysobalanaceae) has been reported previously. However, the beneficial activities of this plant on skin health have remained unclear. This study explores the protective activity of a methanol extract (50-100 μg/mL) in the aerial parts of L. macrocarpa Cuatrec (Lm-ME) and its mechanism, in terms of its moisturizing/hydration factors, skin wrinkles, UV radiation-induced cell damage, and radical generation (using RT/real-time PCR, carbazole assays, flowcytometry, DPPH/ABTS, and immunoblotting analysis). The anti-pigmentation role of Lm-ME was also tested by measuring levels of melanin, melanogenesis-related genes, and pigmentation-regulatory proteins. Lm-ME decreased UVB-irradiated death in HaCaT cells by suppressing apoptosis and inhibited matrix metalloproteinases 1/2 (MMP1/2) expression by enhancing the activity of extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK), and p38. It was confirmed that Lm-ME displayed strong antioxidative activity. Lm-ME upregulated the expression of hyaluronan synthases-2/3 (HAS-2/3) and transglutaminase-1 (TGM-1), as well as secreted levels of hyaluronic acid (HA) via p38 and JNK activation. This extract also significantly inhibited the production of hyaluronidase (Hyal)-1, -2, and -4. Lm-ME reduced the melanin expression of microphthalmia-associated transcription factor (MITF), tyrosinase, and tyrosinase-related protein-1/2 (TYRP-1/2) in α-melanocyte-stimulating hormone (α-MSH)-treated B16F10 cells via the reduction of cAMP response element-binding protein (CREB) and p38 activation. These results suggest that Lm-ME plays a role in skin protection through antioxidative, moisturizing, cytoprotective, and skin-lightening properties, and may become a new and promising cosmetic product beneficial for the skin.
Collapse
Affiliation(s)
- Kon Kuk Shin
- Department of Integrative Biotechnology, Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Korea; (K.K.S.); (L.R.L.); (N.P.Q.); (L.Y.); (N.A.); (D.J.)
| | - Sang Hee Park
- Department of Biocosmetics, Sungkyunkwan University, Suwon 16419, Korea; (S.H.P.); (H.Y.L.)
| | - Hye Yeon Lim
- Department of Biocosmetics, Sungkyunkwan University, Suwon 16419, Korea; (S.H.P.); (H.Y.L.)
| | - Laura Rojas Lorza
- Department of Integrative Biotechnology, Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Korea; (K.K.S.); (L.R.L.); (N.P.Q.); (L.Y.); (N.A.); (D.J.)
| | - Nurinanda Prisky Qomaladewia
- Department of Integrative Biotechnology, Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Korea; (K.K.S.); (L.R.L.); (N.P.Q.); (L.Y.); (N.A.); (D.J.)
| | - Long You
- Department of Integrative Biotechnology, Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Korea; (K.K.S.); (L.R.L.); (N.P.Q.); (L.Y.); (N.A.); (D.J.)
| | - Nur Aziz
- Department of Integrative Biotechnology, Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Korea; (K.K.S.); (L.R.L.); (N.P.Q.); (L.Y.); (N.A.); (D.J.)
| | - Soo Ah Kim
- DanjoungBio Co., Ltd., Wonju 26303, Korea; (S.A.K.); (J.S.L.); (E.S.C.)
| | - Jong Sub Lee
- DanjoungBio Co., Ltd., Wonju 26303, Korea; (S.A.K.); (J.S.L.); (E.S.C.)
| | - Eui Su Choung
- DanjoungBio Co., Ltd., Wonju 26303, Korea; (S.A.K.); (J.S.L.); (E.S.C.)
| | - Jin Kyung Noh
- Instituto de BioEconomia, El Batan, Quito 170135, Ecuador;
| | - Dong-Keun Yie
- International Biological Material Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea;
| | - Deok Jeong
- Department of Integrative Biotechnology, Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Korea; (K.K.S.); (L.R.L.); (N.P.Q.); (L.Y.); (N.A.); (D.J.)
- Convergence Research Center for Energy and Environmental Sciences, Sungkyunkwan University, Suwon 16419, Korea
| | - Jongsung Lee
- Department of Integrative Biotechnology, Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Korea; (K.K.S.); (L.R.L.); (N.P.Q.); (L.Y.); (N.A.); (D.J.)
- Department of Biocosmetics, Sungkyunkwan University, Suwon 16419, Korea; (S.H.P.); (H.Y.L.)
| | - Jae Youl Cho
- Department of Integrative Biotechnology, Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Korea; (K.K.S.); (L.R.L.); (N.P.Q.); (L.Y.); (N.A.); (D.J.)
- Department of Biocosmetics, Sungkyunkwan University, Suwon 16419, Korea; (S.H.P.); (H.Y.L.)
| |
Collapse
|
3
|
Mu Z, Guo J, Zhang D, Xu Y, Zhou M, Guo Y, Hou Y, Gao X, Han X, Geng L. Therapeutic Effects of Shikonin on Skin Diseases: A Review. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2022; 49:1871-1895. [PMID: 34961421 DOI: 10.1142/s0192415x21500889] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Shikonin is one of the primary active components extracted from the dried root ofZicao (Lithospermum erythrorhizon, Onosma paniculata, or Arnebia euchroma), a traditional Chinese herbal medicine. Shikonin is known to not only exert anti-proliferative, anti-inflammatory, and anti-angiogenic activities, but also play a crucial role in triggering the production of reactive oxygen species, suppressing the release of exosomes, and inducing apoptosis. Increasing evidence suggests that shikonin has a protective effect against skin diseases, including psoriasis, melanoma, and hypertrophic scars. In order to evaluate the application potential of shikonin in the treatment of skin diseases, this review is the first of its kind to provide comprehensive and up-to-date information regarding the uses of shikonin and its derivatives on skin diseases and its underlying mechanisms. In this review, we have focused on the signaling pathways and cellular targets involved in the anti-dermatosis effects of shikonin to bridge the gaps in the literature, thereby providing scientific support for the research and development of new drugs from a traditional medicinal plant.
Collapse
Affiliation(s)
- Zhenzhen Mu
- China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110000, P. R. China.,Department of Dermatology, No. 1 Hospital of China Medical University, 155N, Nanjing Street, Heping District, Shenyang, Liaoning 110000, P. R. China.,Department of Dermatology, Shengjing Hospital of China Medical University, 36N, Sanhao Street, Heping District, Shenyang, Liaoning 110000, P. R. China
| | - Jinrong Guo
- China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110000, P. R. China.,Department of Dermatology, No. 1 Hospital of China Medical University, 155N, Nanjing Street, Heping District, Shenyang, Liaoning 110000, P. R. China.,Department of Dermatology, Jincheng People's Hospital, 456N, Wenchang East Street, Jincheng, Shanxi 048000, P. R. China
| | - Dongxia Zhang
- Department of Dermatology, Zhongshan Torch Development Zone Hospital, 123N, Yixian Road, Torch Zone, Zhongshan 528400, Guangdong, P. R. China
| | - Yuanyuan Xu
- Department of Dermatology, No. 1 Hospital of China Medical University, 155N, Nanjing Street, Heping District, Shenyang, Liaoning 110000, P. R. China
| | - Mingming Zhou
- China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110000, P. R. China.,Department of Dermatology, No. 1 Hospital of China Medical University, 155N, Nanjing Street, Heping District, Shenyang, Liaoning 110000, P. R. China
| | - Yimeng Guo
- China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110000, P. R. China.,Department of Dermatology, No. 1 Hospital of China Medical University, 155N, Nanjing Street, Heping District, Shenyang, Liaoning 110000, P. R. China
| | - Yuzhu Hou
- China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110000, P. R. China.,Department of Dermatology, No. 1 Hospital of China Medical University, 155N, Nanjing Street, Heping District, Shenyang, Liaoning 110000, P. R. China
| | - Xinghua Gao
- Department of Dermatology, No. 1 Hospital of China Medical University, 155N, Nanjing Street, Heping District, Shenyang, Liaoning 110000, P. R. China
| | - Xiuping Han
- Department of Dermatology, Shengjing Hospital of China Medical University, 36N, Sanhao Street, Heping District, Shenyang, Liaoning 110000, P. R. China
| | - Long Geng
- Department of Dermatology, No. 1 Hospital of China Medical University, 155N, Nanjing Street, Heping District, Shenyang, Liaoning 110000, P. R. China
| |
Collapse
|
4
|
Song M, Zhang H, Chen Z, Yang J, Li J, Shao S, Liu J. Shikonin reduces hepatic fibrosis by inducing apoptosis and inhibiting autophagy via the platelet-activating factor-mitogen-activated protein kinase axis. Exp Ther Med 2020; 21:28. [PMID: 33262814 DOI: 10.3892/etm.2020.9460] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 07/23/2020] [Indexed: 12/19/2022] Open
Abstract
Liver fibrosis is a tissue repair process that occurs following various types of chronic liver injury and can develop into liver cirrhosis, portal hypertension or liver cancer without effective treatment. Shikonin has anti-inflammatory, antiviral and antitumor properties. Furthermore, shikonin has an additional effect of antagonizing tissue and organ fibrosis. The aim of the present study was to evaluate the mechanisms of action underlying shikonin against liver fibrosis. Cell viability was assessed using the Cell Counting Kit-8 and EdU incorporation assays. Protein and mRNA expression levels were measured via western blotting and immunofluorescence assays, respectively. Apoptosis was examined via flow cytometry and autophagy via transmission electron microscopy. Compared with the control group, shikonin did not significantly alter LX-2 cell viability at 0.2 µmol/ml, which was used as the intervention concentration. However, shikonin significantly inhibited fibrosis, as indicated by a decrease in the expression of α-smooth muscle actin and collagen-I in the TGF-β + shikonin group compared with the TGF-β group. The results indicated that shikonin potentially inhibited fibrosis via promoting cell apoptosis and inhibiting autophagy. Additionally, the results of the present study indicated that shikonin downregulated the expression levels of platelet-activating factor (PAF) in TGF-β-treated cells, which subsequently activated the MAPK signaling pathway, leading to enhanced cell apoptosis and reduced autophagy. Collectively, the present study indicated that shikonin promoted cell apoptosis and suppressed autophagy via the PAF-MAPK axis in LX-2 cells, thus blocking the development of fibrosis. The results of the present study may provide a potential therapeutic strategy for liver fibrosis.
Collapse
Affiliation(s)
- Min Song
- Department of Gastroenterology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Heng Zhang
- Department of Gastroenterology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Zhitao Chen
- Department of Gastroenterology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Jing Yang
- Department of Gastroenterology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Jie Li
- Department of Gastroenterology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Sue Shao
- Department of Gastroenterology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Jing Liu
- Department of Gastroenterology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| |
Collapse
|
5
|
Wang Y, Zhu F, Zhang Y, Chen C, Lai Y, Sun J, Chen S, Qiu P, Gao J, Deng G. Shikonin suppresses trophoblast cell growth via regulation of GLI1, and p62 mediated caspase 8 activation. Reprod Toxicol 2020; 95:104-112. [PMID: 32461113 DOI: 10.1016/j.reprotox.2020.05.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 05/10/2020] [Accepted: 05/18/2020] [Indexed: 10/24/2022]
Abstract
Unruptured ectopic pregnancy (UEP) is a common cause of morbidity and, occasionally, of mortality in women of reproductive age. Pharmacological intervention is a common therapeutic approach for early-stage UEP. Herein, we investigated the cytotoxic effect and novel mechanism of shikonin, a natural naphthoquinone pigment purified from Lithospermum erythrorhizon, in human trophoblast cells. These data demonstrated that shikonin suppressed proliferation and induced apoptosis in a time-dependent manner in HTR-8/SVneo cells. Shikonin blocked autophagic flux and promoted p62 interaction with caspase 8, resulting in caspase 8 activation. Moreover, shikonin suppressed GLI1 expression, and GLI1 overexpression attenuated shikonin-induced cell apoptosis. Although silencing GLI1 slightly promoted cell apoptosis, p62 overexpression enhanced GLI1 silencing-induced cell apoptosis by activating caspase 8. Furthermore, rapamycin increased shikonin-induced cell apoptosis in HTR-8/SVneo cells, whereas 3-MA attenuated the cytotoxic effect of shikonin. In conclusion, shikonin suppressed trophoblast cell growth by silencing GLI1 and increasing p62 co-mediated activation of caspase 8, which suggested a potential novel therapeutic target for UEP.
Collapse
Affiliation(s)
- Yanxi Wang
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Fangfang Zhu
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Yingxuan Zhang
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Chunlin Chen
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Yuling Lai
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Jianhua Sun
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Si Chen
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Pin Qiu
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Jie Gao
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| | - Gaopi Deng
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| |
Collapse
|
6
|
Wang F, Yao X, Zhang Y, Tang J. Synthesis, biological function and evaluation of Shikonin in cancer therapy. Fitoterapia 2019; 134:329-339. [DOI: 10.1016/j.fitote.2019.03.005] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 03/04/2019] [Accepted: 03/07/2019] [Indexed: 12/16/2022]
|
7
|
Figat R, Zgadzaj A, Geschke S, Sieczka P, Pietrosiuk A, Sommer S, Skrzypczak A. Cytotoxicity and antigenotoxicity evaluation of acetylshikonin and shikonin. Drug Chem Toxicol 2018; 44:140-147. [PMID: 30574814 DOI: 10.1080/01480545.2018.1536710] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Shikonin (SH) is used as a red pigment for food coloring and cosmetics, and has cytotoxic activity towards cancer cells. However, due to strong toxicity SH has limited potential as an anticancer drug. Acetylshikonin (ASH) is one of the SH derivatives with promising anticancer potential. In present study, we attempted to evaluate and compare the cytotoxicity of SH and ASH towards a normal cell line (V79) and in addition to evaluate their antigenotoxic activity. The evaluation was made with the use of the set of cytotoxicity assays with V79 line and the micronucleus test in vitro performed using clinafloxacin (CLFX), ethyl methanesulfonate (EMS) as direct genotoxins and cyclophosphamide (CPA) as indirect genotoxin. For CPA and EMS the simultaneous protocol was used and for CLFX three different variants were performed: pretreatment, simultaneous, and post-treatment. A higher cytotoxic effect was observed for SH. The EC50 values obtained for SH were approximately twofold lower compared to that of ASH. Moreover, ASH exhibited an antigenotoxic potential against CPA-induced genotoxicity, whereas SH has no activity. However, ASH increased the EMS-induced genotoxicity, when SH exhibited no effect. Both compounds decreased the genotoxicity of CLFX in pretreatment and simultaneous protocol. Based on the results of the present study it can be concluded that ASH is less cytotoxic than SH to normal cells and has comparable antigenotoxic potential.
Collapse
Affiliation(s)
- Ramona Figat
- Department of Environmental Health Sciences, Medical University of Warsaw, Poland
| | - Anna Zgadzaj
- Department of Environmental Health Sciences, Medical University of Warsaw, Poland
| | - Sylwia Geschke
- Department of Environmental Health Sciences, Medical University of Warsaw, Poland
| | - Patrycja Sieczka
- Department of Environmental Health Sciences, Medical University of Warsaw, Poland
| | - Agnieszka Pietrosiuk
- Department of Pharmaceutical Biology and Medicinal Plant Biotechnology, Medical University of Warsaw, Poland
| | - Sylwester Sommer
- Centre for Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, Warsaw, Poland
| | - Agata Skrzypczak
- Department of Environmental Health Sciences, Medical University of Warsaw, Poland
| |
Collapse
|
8
|
Synthesis of Novel Shikonin Derivatives and Pharmacological Effects of Cyclopropylacetylshikonin on Melanoma Cells. Molecules 2018; 23:molecules23112820. [PMID: 30380765 PMCID: PMC6278577 DOI: 10.3390/molecules23112820] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 10/19/2018] [Accepted: 10/20/2018] [Indexed: 12/25/2022] Open
Abstract
Despite much research in the last centuries, treatment of malignant melanoma is still challenging because of its mostly unnoticeable metastatic spreading and aggressive growth rate. Therefore, the discovery of novel drug leads is an important goal. In a previous study, we have isolated several shikonin derivatives from the roots of Onosma paniculata Bureau & Franchet (Boraginaceae) which evolved as promising anticancer candidates. β,β-Dimethylacrylshikonin (1) was the most cytotoxic derivative and exhibited strong tumor growth inhibitory activity, in particular, towards melanoma cells. In this study, we synthesized eighteen novel shikonin derivatives in order to obtain compounds which exhibit a higher cytotoxicity than 1. We investigated their cytotoxic potential against various melanoma cell lines and juvenile skin fibroblasts. The most active compound was (R)-1-(1,4-dihydro-5,8-dihydroxy-1,4-dioxonaphthalen-2-yl)-4-methylpent-3-enyl cyclopropylacetate (cyclopropylacetylshikonin) (6). It revealed significant stronger tumor growth inhibitory activity towards two melanoma cell lines derived from metastatic lesions (WM164 and MUG-Mel2). Further investigations have shown that 6 induced apoptosis caspase-dependently, increased the protein levels of cleaved PARP, and led to double-stranded DNA breaks as shown by phosphorylation of H2AX. Cell membrane damage and cell cycle arrest were not observed.
Collapse
|
9
|
Jia J, Li C, Yang J, Wang X, Li R, Luo S, Li Z, Liu J, Liu Z, Zheng Y. Yes-associated protein promotes the abnormal proliferation of psoriatic keratinocytes via an amphiregulin dependent pathway. Sci Rep 2018; 8:14513. [PMID: 30323299 PMCID: PMC6189173 DOI: 10.1038/s41598-018-32522-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 08/06/2018] [Indexed: 11/08/2022] Open
Abstract
Psoriasis is a chronic inflammatory skin disease with high morbidity, poor treatment methods and high rates of relapse. Keratinocyte hyperproliferation and shortened cell cycles are important pathophysiological features of psoriasis. As a known oncogene, Yes-associated protein (YAP) plays a role in promoting cell proliferation and inhibiting cell apoptosis; however, whether YAP is involved in the pathogenesis of psoriasis remains to be determined. Amphiregulin (AREG), a transcriptional target of YAP, was found to be upregulated in psoriasis, and overexpression of AREG promoted keratinocyte proliferation. In the present study, immunohistochemistry showed that YAP expression was elevated in the skin of psoriasis patients and in the Imiquimod (IMQ) mouse model of psoriasis. Knockdown of YAP in HaCaT cells inhibited cell proliferation, caused cell cycle arrest in G0/G1 phase and promoted apoptosis. These changes in YAP-knockdown HaCaT cells were related to changes in AREG expression. We concluded that YAP may play an important role in the regulation of abnormal keratinocyte proliferation via an AREG-dependent pathway and that YAP could be a new target in the treatment of psoriasis.
Collapse
Affiliation(s)
- Jinjing Jia
- Department of Dermatology, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Changji Li
- Department of Dermatology, the Second Affiliated Hospital of Xi'an Jiaotong University, School of Medicine, Xi'an, China
| | - Jiao Yang
- Department of Dermatology, the Second Affiliated Hospital of Xi'an Jiaotong University, School of Medicine, Xi'an, China
| | - Xin Wang
- Department of Dermatology, the Second Affiliated Hospital of Xi'an Jiaotong University, School of Medicine, Xi'an, China
| | - Ruilian Li
- Department of Dermatology, the Second Affiliated Hospital of Xi'an Jiaotong University, School of Medicine, Xi'an, China
| | - Suju Luo
- Department of Dermatology, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhengxiao Li
- Department of Dermatology, the Second Affiliated Hospital of Xi'an Jiaotong University, School of Medicine, Xi'an, China
| | - Jiankang Liu
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
- Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Zhi Liu
- Department of Dermatology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 26599, USA
| | - Yan Zheng
- Department of Dermatology, the Second Affiliated Hospital of Xi'an Jiaotong University, School of Medicine, Xi'an, China.
| |
Collapse
|
10
|
Shilnikova K, Piao MJ, Kang KA, Ryu YS, Park JE, Hyun YJ, Zhen AX, Jeong YJ, Jung U, Kim IG, Hyun JW. Shikonin induces mitochondria-mediated apoptosis and attenuates epithelial-mesenchymal transition in cisplatin-resistant human ovarian cancer cells. Oncol Lett 2018; 15:5417-5424. [PMID: 29563994 PMCID: PMC5858079 DOI: 10.3892/ol.2018.8065] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 08/08/2017] [Indexed: 12/23/2022] Open
Abstract
Cisplatin-based chemotherapy often results in the development of chemoresistance when used to treat ovarian cancer, which is difficult to overcome. The present study investigated the cytotoxic and anti-migratory effects of shikonin, a naphthoquinone compound, on cisplatin-resistant human ovarian cancer A2780 cells (A2780-CR). Shikonin had a potent dose-dependent cytotoxic effect on A2780-CR cells, with 9 µM shikonin treatment reducing A2780-CR cell viability by 50%, validate using an MTT assay. Shikonin induced apoptosis, as evidenced by the increased number of apoptotic bodies, following staining with Hoechst 33342, and terminal deoxynucleotidyl cell transferase dUTP nick end labeling-positive cells following treatment. Flow cytometry and fluorescent microscope imaging, following JC-1 staining, revealed that shikonin increased mitochondrial membrane depolarization. Also it altered the levels of apoptosis-associated proteins, leading to diminished expression of B cell lymphoma-2 (Bcl-2), enhanced expression of Bcl-associated X, and cleavage of caspase-9 and −3, as revealed using western blot analysis. Shikonin activated mitogen-activated protein kinases; while treatment with specific inhibitors of these kinases attenuated the decline in cell viability induced by shikonin treatment. In addition, the cell migration assay and western blot analysis indicated that shikonin decreased the migratory capacity of A2780-CR cells via the upregulation of epithelial-cadherin and downregulation of neural-cadherin. Taken together, the results of the present study indicated that shikonin induces mitochondria-mediated apoptosis and attenuates the epithelial-mesenchymal transition in A2780-CR cells.
Collapse
Affiliation(s)
- Kristina Shilnikova
- Department of Biochemistry, School of Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Mei Jing Piao
- Department of Biochemistry, School of Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Kyoung Ah Kang
- Department of Biochemistry, School of Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Yea Seong Ryu
- Department of Biochemistry, School of Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Jeong Eon Park
- Department of Biochemistry, School of Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Yu Jae Hyun
- Department of Biochemistry, School of Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Ao Xuan Zhen
- Department of Biochemistry, School of Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Yong Joo Jeong
- Department of Bio and Nanochemistry, Kookmin University, Seoul 02707, Republic of Korea
| | - Uhee Jung
- Radiation Biotechnology Research Division, Korea Atomic Energy Research Institute, Jeongeup, Jeollabuk 56212, Republic of Korea
| | - In Gyu Kim
- Department of Radiation Biology, Environmental Radiation Research Group, Korea Atomic Energy Research Institute, Daejeon 34057, Republic of Korea
| | - Jin Won Hyun
- Department of Biochemistry, School of Medicine, Jeju National University, Jeju 63243, Republic of Korea
| |
Collapse
|
11
|
Spyrelli ED, Kyriazou AV, Virgiliou C, Nakas A, Deda O, Papageorgiou VP, Assimopoulou AN, Gika HG. Metabolic profiling study of shikonin's cytotoxic activity in the Huh7 human hepatoma cell line. MOLECULAR BIOSYSTEMS 2018; 13:841-851. [PMID: 28265634 DOI: 10.1039/c6mb00830e] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Shikonin and its enantiomer alkannin, which are natural products, have been extensively studied in vitro and in vivo for, among others, their antitumor activity. The investigation of the molecular pathways involved in their action is of interest, since they are not yet clearly defined. Metabolic profiling in cells can provide a picture of a cell's phenotype upon intervention, assisting in the elucidation of the mechanism of action. In this study, the cytotoxic effect of shikonin on a human hepatocarcinoma cell line was studied. Huh7 cells were treated with shikonin at 5 μM, and it was found that shikonin markedly inhibited cell growth. Metabolic profiling indicated alterations in the metabolic content of the cells and the culture media upon treatment, detecting the metabolic response of the cells. This study demonstrates the potential of metabolomics to improve knowledge on the mechanisms involved in shikonin's antitumor action.
Collapse
Affiliation(s)
- E D Spyrelli
- Department of Chemical Engineering, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Zhang LL, Zhan L, Jin YD, Min ZL, Wei C, Wang Q, Chen YJ, Wu QM, Hu XM, Yuan Q. SIRT2 mediated antitumor effects of shikonin on metastatic colorectal cancer. Eur J Pharmacol 2017; 797:1-8. [PMID: 28088387 DOI: 10.1016/j.ejphar.2017.01.008] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 01/10/2017] [Accepted: 01/11/2017] [Indexed: 01/07/2023]
Abstract
SIRT2 is involved in the development of a variety of cancers. Shikonin is a natural compound that is known to have antitumor effects. This study aims to assess the effects of shikonin on the development and metastatic progression of colorectal cancer (CRC) through regulation of SIRT2 expression and whether this effect is related to the phosphorylation of extracellular signal-regulated kinases (ERKs). The results demonstrated that SIRT2 is downregulated in CRC biopsy samples (n=31) compared with the adjacent non-cancerous tissues (ANCT, n=26). Furthermore, CRC metastases were positive for SIRT2 despite a lack of expression in the primary tumor. In addition, data from an in vitro assay revealed that overexpression of SIRT2 inhibited the proliferation and metastatic progression of SW480 cells while blocking of SIRT2 expression induced the proliferation and metastatic progression of HT29 cells. Shikonin inhibited the viability, migration and invasion of SW480 cells and it also inhibited the tumor growth in the nude mice model; while AGK2 (a specific inhibitor of SIRT2) reversed these effects. Epidermal growth factor (EGF, an activator of ERK) and ERK-overexpression inhibited the effects of shikonin on SIRT2 expression, proliferation and metastasis in SW480 cells. However, this proliferative effect of EGF was reversed by SIRT2 overexpression. In conclusion, these results suggest that SIRT2 is a new therapeutic target for the treatment of CRC. The antitumor effects of shikonin on CRC seem to be mediated by SIRT2 upregulation via phospho-ERK inhibition.
Collapse
Affiliation(s)
- Li-Li Zhang
- Department of Pharmacology, Medical College, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Lin Zhan
- Department of Pharmacology, Medical College, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Yong-Dong Jin
- Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Zhen-Li Min
- Department of Pharmacology, Medical College, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Can Wei
- Department of Pharmacology, Medical College, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Qiang Wang
- Department of Immunology, Medical College, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Ya-Jun Chen
- Department of Pharmacology, Medical College, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Qing-Ming Wu
- Department of Pharmacology, Medical College, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Xia-Min Hu
- Department of Pharmacology, Medical College, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Qiong Yuan
- Department of Pharmacology, Medical College, Wuhan University of Science and Technology, Wuhan 430065, China.
| |
Collapse
|
13
|
Shikonin regulates C-MYC and GLUT1 expression through the MST1-YAP1-TEAD1 axis. Exp Cell Res 2016; 349:273-281. [PMID: 27793648 DOI: 10.1016/j.yexcr.2016.10.018] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 10/19/2016] [Accepted: 10/22/2016] [Indexed: 11/21/2022]
Abstract
The general mechanism underlying the tumor suppressor activity of the Hippo signaling pathway remains unclear. In this study, we explore the molecular mechanisms connecting the Hippo signaling pathway with glucose metabolism. We have found that two key regulators of glycolysis, C-MYC and GLUT1, are targets of the Hippo signaling pathway in human leukemia cells. Our results revealed that activation of MST1 by the natural compound shikonin inhibited the expression of GLUT1 and C-MYC. Furthermore, RNAi experiments confirmed the regulation of GLUT1 and C-MYC expression via the MST1-YAP1-TEAD1 axis. Surprisingly, YAP1 was found to positively regulate C-MYC mRNA levels in complex with TEAD1, while it negatively regulates C-MYC levels in cooperation with MST1. Hence, YAP1 serves as a rheostat for C-MYC, which is regulated by MST1. In addition, depletion of MST1 stimulates lactate production, whereas the specific depletion of TEAD1 has an opposite effect. The inhibition of lactate production and cellular proliferation induced by shikonin also depends on the Hippo pathway activity. Finally, a bioinformatic analysis revealed conserved TEAD-binding motifs in the C-MYC and GLUT1 promoters providing another molecular data supporting our observations. In summary, regulation of glucose metabolism could serve as a new tumor suppressor mechanism orchestrated by the Hippo signaling pathway.
Collapse
|