1
|
Chen J, Zhang A, Nie A, Zuo X, Zhang L, Jiao Y, Wang L, Yang Y, Liu K, Xue X, Zhuang Y, Meng Y, Yang JH. Multi-omics analysis to reveal the synergistic mechanism underlying the multiple ingredients of Stephania tetrandra extract on rheumatoid arthritis through the PI3K/Akt signaling pathway. Front Pharmacol 2024; 15:1447283. [PMID: 39221139 PMCID: PMC11361992 DOI: 10.3389/fphar.2024.1447283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
Background: Stephania tetrandra has been used for treating rheumatic diseases for thousands of years in rural areas of China. Several studies have found that tetrandrine and fangchinoline can inactivate the PI3K/Akt signaling pathway by reducing the expression and phosphorylation of AKT. However, the mechanism underlying the therapeutic actions of S. tetrandra on RA is not well known. Methods: In this study, we determined the molecular mechanism of the therapeutic effects of the multiple ingredients of S. tetrandra extract (STE) on collagen-induced arthritic (CIA) rats by integrating pharmacometabolomics, proteomics, and PTMomics. Results: In the multi-omics joint analysis, first, the expression signatures of proteins, PTMs, metabolites, and STE ingredients were profiled in CIA rats PBMCs that underwent STE treatment. Bioinformatics analysis were subsequently probed that STE mainly regulated tryptophan metabolism, inflammatory response, and cell adhesion pathways in CIA rats. The interrelated pathways were further constructed, and the findings revealed that STE attenuated the inflammatory response and proliferation of PBMCs in CIA rats by mediating the key targets of the PI3K/Akt pathway, including Hint1, ACP1, FGR, HSP90@157W + dioxidation, and Prkca@220N + 845.4540 Da. The rheumatic functions of Hint1 and ACP1 were further confirmed by applying a transcriptomic data of RA patients who clinically received abatacept therapy. Furthermore, a cross-ome correlation analysis was performed and major in vivo ingredients of STE, including coclaurine-N-glucuronide, Me,coclaurine-O-glc, N-gluA-schefferine, corydamine, corypamine, tetrandrine, and fangchiniline, were found to act on these targerts to inactivate the PI3K/Akt pathway. Conclusion: These results elucidated the molecular mechanism by which the ingredients of STE mediate the expression of the key targets in the PI3K/Akt pathway, leading to anti-rheumatic functions. The findings of this study provided new insights into the synergistic effect of STE against arthritis in rats.
Collapse
Affiliation(s)
- Jinfeng Chen
- Research Center for Clinical Systems Biology, Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Institute of Infection and Immunity, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, China
| | - An Zhang
- Research Center for Clinical Systems Biology, Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Institute of Infection and Immunity, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, China
| | - Anzheng Nie
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xiaoxiao Zuo
- Radiotherapy Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lei Zhang
- Research Center for Clinical Systems Biology, Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Institute of Infection and Immunity, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, China
| | - Yuxue Jiao
- Research Center for Clinical Systems Biology, Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Institute of Infection and Immunity, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, China
| | - Lulu Wang
- Research Center for Clinical Systems Biology, Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Institute of Infection and Immunity, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, China
| | - Yang Yang
- Research Center for Clinical Systems Biology, Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Institute of Infection and Immunity, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, China
| | - Kun Liu
- Research Center for Clinical Systems Biology, Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Institute of Infection and Immunity, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, China
| | - Xinli Xue
- Research Center for Clinical Systems Biology, Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Institute of Infection and Immunity, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, China
| | - Yuanyuan Zhuang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yansha Meng
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jing-Hua Yang
- Research Center for Clinical Systems Biology, Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Institute of Infection and Immunity, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, China
| |
Collapse
|
2
|
Saetang J, Sukkapat P, Mittal A, Julamanee J, Khopanlert W, Maneechai K, Nazeer RA, Sangkhathat S, Benjakul S. Proteome Analysis of the Antiproliferative Activity of the Novel Chitooligosaccharide-Gallic Acid Conjugate against the SW620 Colon Cancer Cell Line. Biomedicines 2023; 11:1683. [PMID: 37371778 PMCID: PMC10296375 DOI: 10.3390/biomedicines11061683] [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: 05/09/2023] [Revised: 06/07/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
Chitooligosaccharide (COS) and gallic acid (GA) are natural compounds with anti-cancer properties, and their conjugate (COS-GA) has several biological activities. Herein, the anti-cancer activity of COS-GA in SW620 colon cancer cells was investigated. MTT assay was used to evaluate cell viability after treatment with 62.5, 122, and 250 µg/mL of COS, GA, and COS-GA for 24 and 48 h. The number of apoptotic cells was determined using flow cytometry. Proteomic analysis was used to explore the mechanisms of action of different compounds. COS-GA and GA showed a stronger anti-cancer effect than COS by reducing SW620 cell proliferation at 125 and 250 µg/mL within 24 h. Flow cytometry revealed 20% apoptosis after COS-GA treatment for 24 h. Thus, GA majorly contributed to the enhanced anti-cancer activity of COS via conjugation. Proteomic analysis revealed alterations in protein translation and DNA duplication in the COS group and the structural constituents of the cytoskeleton, intermediate filament organization, the mitochondrial nucleoid, and glycolytic processes in the COS-GA group. Anti-cancer-activity-related proteins were altered, including CLTA, HSPA9, HIST2H2BF, KRT18, HINT1, DSP, and VIM. Overall, the COS-GA conjugate can serve as a potential anti-cancer agent for the safe and effective treatment of colon cancer.
Collapse
Affiliation(s)
- Jirakrit Saetang
- International Center of Excellence in Seafood Science and Innovation, Faculty of Agro-Industry, Prince of Songkla University, Hat Yai 90110, Songkhla, Thailand; (J.S.); (P.S.); (A.M.)
| | - Phutthipong Sukkapat
- International Center of Excellence in Seafood Science and Innovation, Faculty of Agro-Industry, Prince of Songkla University, Hat Yai 90110, Songkhla, Thailand; (J.S.); (P.S.); (A.M.)
| | - Ajay Mittal
- International Center of Excellence in Seafood Science and Innovation, Faculty of Agro-Industry, Prince of Songkla University, Hat Yai 90110, Songkhla, Thailand; (J.S.); (P.S.); (A.M.)
| | - Jakrawadee Julamanee
- Stem Cell Laboratory, Hematology Unit, Division of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Hat Yai 90110, Songkhla, Thailand; (J.J.); (W.K.); (K.M.)
| | - Wannakorn Khopanlert
- Stem Cell Laboratory, Hematology Unit, Division of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Hat Yai 90110, Songkhla, Thailand; (J.J.); (W.K.); (K.M.)
| | - Kajornkiat Maneechai
- Stem Cell Laboratory, Hematology Unit, Division of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Hat Yai 90110, Songkhla, Thailand; (J.J.); (W.K.); (K.M.)
| | - Rasool Abdul Nazeer
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chennai 603203, Tamilnadu, India;
| | - Surasak Sangkhathat
- Department of Surgery, Faculty of Medicine, Prince of Songkla University, Hat Yai 90110, Songkhla, Thailand;
- Translational Medicine Research Center, Faculty of Medicine, Prince of Songkla University, Hat Yai 90110, Songkhla, Thailand
| | - Soottawat Benjakul
- International Center of Excellence in Seafood Science and Innovation, Faculty of Agro-Industry, Prince of Songkla University, Hat Yai 90110, Songkhla, Thailand; (J.S.); (P.S.); (A.M.)
- Department of Food and Nutrition, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
3
|
Zhang C, Bao T, Ke Y, Liu X, Wang X, Liao W, He Y, Wang L. Integrated analysis of ceRNA network reveals potential prognostic Hint1-related lncRNAs involved in hepatocellular carcinoma progression. World J Surg Oncol 2022; 20:67. [PMID: 35241097 PMCID: PMC8896107 DOI: 10.1186/s12957-022-02535-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 02/18/2022] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Hint1 is a novel tumor suppressor gene, and inactivation of its expression is closely associated with the carcinogenesis of a variety of malignancies. The effects of Hint1 deficiency on the competing endogenous RNA (ceRNA) regulatory network in the context of HCC remains to be fully characterized. This study aims to explore Hint1-related hub lncRNAs in HCC and to establish a reliable prognostic model for HCC patients based on these hub lncRNAs. METHODS lncRNA + mRNA microarray was used to identify differentially expressed (DE) lncRNAs and mRNAs in Huh7 cells before and after Hint1 knockdown. A Hint1-related ceRNA network was mapped by bioinformation technology. The DEmRNAs in the network were analyzed via GO and KEGG enrichment analyses. Hub DElncRNAs associated with HCC patient prognosis were then detected through univariate and multivariate Cox regression analyses and were incorporated into a prognostic model. The prognostic value of this model was then assessed through the use of Kaplan-Meier curves, time-related ROC analyses, and nomograms. We also utilized Kaplan-Meier curves to validate the relationship between hub lncRNAs and the overall survival (OS) of HCC patients. Finally, A Hint1-related core ceRNA network based on the hub DElncRNAs and DEmRNAs was mapped. RESULTS We identified 417 differentially expressed DElncRNAs and 2096 DEmRNAs in Huh7 cells before and after Hint1 knockdown. Three hub DElncRNAs (LINC00324, SNHG3, and DIO3OS) in the Hint1-associated ceRNA network were screened out using univariate and multivariate Cox regression analyses. A hepatocellular carcinoma (HCC) prognostic risk-scoring model and nomogram were constructed using these three hub lncRNAs, and it was confirmed that the risk score of the model could be used as an independent predictor of HCC prognosis. A Hint1-related core ceRNA network based on the hub DElncRNAs and DEmRNAs was also mapped. CONCLUSION We constructed a reliable prognostic model for HCC patients based on three Hint1-related hub lncRNAs, and we believe these three hub lncRNAs may play critical roles in hepatocarcinogenesis, and progression.
Collapse
Affiliation(s)
- Cheng Zhang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, No. 1168 Chunrongxi Road, Chenggong District, Kunming, Yunnan Province, China
- Department of Hepatobiliary Surgery, The Second People's Hospital of Chengdu, Chengdu, China
| | - Tianhao Bao
- Mental Health Center of Kunming Medical University, Kunming, China
| | - Yang Ke
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, No. 1168 Chunrongxi Road, Chenggong District, Kunming, Yunnan Province, China
| | - Xin Liu
- Department of Dermatology, The Second Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Xinghong Wang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, No. 1168 Chunrongxi Road, Chenggong District, Kunming, Yunnan Province, China
| | - Weiran Liao
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, No. 1168 Chunrongxi Road, Chenggong District, Kunming, Yunnan Province, China
| | - Yutao He
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, No. 1168 Chunrongxi Road, Chenggong District, Kunming, Yunnan Province, China
| | - Lin Wang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, No. 1168 Chunrongxi Road, Chenggong District, Kunming, Yunnan Province, China.
| |
Collapse
|
4
|
Romualdo GR, Leroy K, Costa CJS, Prata GB, Vanderborght B, da Silva TC, Barbisan LF, Andraus W, Devisscher L, Câmara NOS, Vinken M, Cogliati B. In Vivo and In Vitro Models of Hepatocellular Carcinoma: Current Strategies for Translational Modeling. Cancers (Basel) 2021; 13:5583. [PMID: 34771745 PMCID: PMC8582701 DOI: 10.3390/cancers13215583] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 11/02/2021] [Accepted: 11/04/2021] [Indexed: 12/24/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the sixth most common cancer worldwide and the third leading cause of cancer-related death globally. HCC is a complex multistep disease and usually emerges in the setting of chronic liver diseases. The molecular pathogenesis of HCC varies according to the etiology, mainly caused by chronic hepatitis B and C virus infections, chronic alcohol consumption, aflatoxin-contaminated food, and non-alcoholic fatty liver disease associated with metabolic syndrome or diabetes mellitus. The establishment of HCC models has become essential for both basic and translational research to improve our understanding of the pathophysiology and unravel new molecular drivers of this disease. The ideal model should recapitulate key events observed during hepatocarcinogenesis and HCC progression in view of establishing effective diagnostic and therapeutic strategies to be translated into clinical practice. Despite considerable efforts currently devoted to liver cancer research, only a few anti-HCC drugs are available, and patient prognosis and survival are still poor. The present paper provides a state-of-the-art overview of in vivo and in vitro models used for translational modeling of HCC with a specific focus on their key molecular hallmarks.
Collapse
Affiliation(s)
- Guilherme Ribeiro Romualdo
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo (USP), São Paulo 05508-270, Brazil; (G.R.R.); (C.J.S.C.); (T.C.d.S.)
- Department of Structural and Functional Biology, Biosciences Institute, São Paulo State University (UNESP), Botucatu 18618-689, Brazil; (G.B.P.); (L.F.B.)
- Department of Pathology, Botucatu Medical School, São Paulo State University (UNESP), Botucatu 18618-687, Brazil
| | - Kaat Leroy
- Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, 1090 Brussels, Belgium; (K.L.); (M.V.)
| | - Cícero Júlio Silva Costa
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo (USP), São Paulo 05508-270, Brazil; (G.R.R.); (C.J.S.C.); (T.C.d.S.)
| | - Gabriel Bacil Prata
- Department of Structural and Functional Biology, Biosciences Institute, São Paulo State University (UNESP), Botucatu 18618-689, Brazil; (G.B.P.); (L.F.B.)
- Department of Pathology, Botucatu Medical School, São Paulo State University (UNESP), Botucatu 18618-687, Brazil
| | - Bart Vanderborght
- Gut-Liver Immunopharmacology Unit, Basic and Applied Medical Sciences, Liver Research Center Ghent, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium;
- Hepatology Research Unit, Internal Medicine and Paediatrics, Liver Research Center Ghent, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium;
| | - Tereza Cristina da Silva
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo (USP), São Paulo 05508-270, Brazil; (G.R.R.); (C.J.S.C.); (T.C.d.S.)
| | - Luís Fernando Barbisan
- Department of Structural and Functional Biology, Biosciences Institute, São Paulo State University (UNESP), Botucatu 18618-689, Brazil; (G.B.P.); (L.F.B.)
| | - Wellington Andraus
- Department of Gastroenterology, Clinics Hospital, School of Medicine, University of São Paulo (HC-FMUSP), São Paulo 05403-000, Brazil;
| | - Lindsey Devisscher
- Hepatology Research Unit, Internal Medicine and Paediatrics, Liver Research Center Ghent, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium;
| | - Niels Olsen Saraiva Câmara
- Department of Immunology, Institute of Biomedical Sciences IV, University of São Paulo (USP), São Paulo 05508-000, Brazil;
| | - Mathieu Vinken
- Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, 1090 Brussels, Belgium; (K.L.); (M.V.)
| | - Bruno Cogliati
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo (USP), São Paulo 05508-270, Brazil; (G.R.R.); (C.J.S.C.); (T.C.d.S.)
| |
Collapse
|
5
|
Dang J, Gao J, Ma F, Luo Y, Wang J, Wang D, Li W, Sun H, Li L, Liu X, Hu D, Jin Z. Roles of metformin-mediated girdin expression in metastasis of epithelial ovarian cancer. Cell Biol Int 2021; 45:1030-1037. [PMID: 33404163 DOI: 10.1002/cbin.11547] [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: 10/29/2020] [Revised: 12/14/2020] [Accepted: 01/03/2021] [Indexed: 11/11/2022]
Abstract
Antimetastatic effect of Metformin has been documented in epithelial ovarian cancer (EOC). Presently, we investigated the regulatory mechanism of Metformin in EOC metastasis. First, Girdin was significantly enhanced in EOC tumorous tissues and cell lines. Seconded, knockdown of Girdin significantly suppressed EOC cell viability, migration, and invasion, while upregulation of Girdin produced the opposite effects in vitro and facilitated lung metastasis in EOC cell xenograft in vivo. In addition, we confirmed that the inhibitory effect of Metformin on Girdin expression. Mechanistically, the oncogenic effects of Girdin could be reversed by LY294002 (an AKT pathway inhibitor) and Metformin. These results suggested that Metformin attenuated EOC metastasis through Girdin and targeting Girdin may be a promising therapeutic strategy for EOC in the future.
Collapse
Affiliation(s)
- Jianhong Dang
- Department of Obstetrics and Gynecology, Changzheng Hospital, The Naval Military Medical University, Shanghai, China
| | - Jinghai Gao
- Department of Obstetrics and Gynecology, Changzheng Hospital, The Naval Military Medical University, Shanghai, China
| | - Fang Ma
- Department of Obstetrics and Gynecology, Changzheng Hospital, The Naval Military Medical University, Shanghai, China
| | - Yan Luo
- Department of Obstetrics and Gynecology, Changzheng Hospital, The Naval Military Medical University, Shanghai, China
| | - Jing Wang
- Department of Obstetrics and Gynecology, Changzheng Hospital, The Naval Military Medical University, Shanghai, China
| | - Dan Wang
- Department of Obstetrics and Gynecology, Changzheng Hospital, The Naval Military Medical University, Shanghai, China
| | - Weiqing Li
- Department of Pathology, Changzheng Hospital, The Naval Military Medical University, Shanghai, China
| | - Hao Sun
- Department of Obstetrics and Gynecology, Changzheng Hospital, The Naval Military Medical University, Shanghai, China
| | - Lingling Li
- Department of Obstetrics and Gynecology, Changzheng Hospital, The Naval Military Medical University, Shanghai, China
| | - Xiaojun Liu
- Department of Obstetrics and Gynecology, Changzheng Hospital, The Naval Military Medical University, Shanghai, China
| | - Dian Hu
- Department of Obstetrics and Gynecology, Changzheng Hospital, The Naval Military Medical University, Shanghai, China
| | - Zhijun Jin
- Department of Obstetrics and Gynecology, Changzheng Hospital, The Naval Military Medical University, Shanghai, China
| |
Collapse
|
6
|
Carrillo-Najar C, Rembao-Bojórquez D, Tena-Suck ML, Zavala-Vega S, Gelista-Herrera N, Ramos-Peek MA, Gómez-Amador JL, Cazares-Raga F, Hernández-Hernández FDLC, Ortiz-Plata A. Comparative Proteomic Study Shows the Expression of Hint-1 in Pituitary Adenomas. Diagnostics (Basel) 2021; 11:diagnostics11020330. [PMID: 33671384 PMCID: PMC7922225 DOI: 10.3390/diagnostics11020330] [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: 12/31/2020] [Revised: 02/12/2021] [Accepted: 02/14/2021] [Indexed: 12/28/2022] Open
Abstract
Pituitary adenomas (PAs) can be unpredictable and aggressive tumors. No reliable markers of their biological behavior have been found. Here, a proteomic analysis was applied to identify proteins in the expression profile between invasive and non-invasive PAs to search for possible biomarkers. A histopathological and immunohistochemical (adenohypophyseal hormones, Ki-67, p53, CD34, VEGF, Flk1 antibodies) analysis was done; a proteomic map was evaluated in 64 out of 128 tumors. There were 107 (84%) invasive and 21 (16%) non-invasive PAs; 80.5% belonged to III and IV grades of the Hardy–Vezina classification. Invasive PAs (n = 56) showed 105 ± 43 spots; 86 ± 32 spots in non-invasive PAs (n = 8) were observed. The 13 most prominent spots were selected and 11 proteins related to neoplastic process in different types of tumors were identified. Hint1 (Histidine triad nucleotide-binding protein 1) high expression in invasive PA was found (11.8 ± 1.4, p = 0.005), especially at high index (>10; p = 0.0002). High Hint1 expression was found in invasive VEGF positive PA (13.8 ± 2.3, p = 0.005) and in Flk1 positive PA (14.04 ± 2.28, p = 0.006). Hint1 is related to human tumorigenesis by its interaction with signaling pathways and transcription factors. It could be related to invasive behavior in PAs. This is the first report on Hint expression in PAs. More analysis is needed to find out the possible role of Hint in these tumors.
Collapse
Affiliation(s)
- Carolina Carrillo-Najar
- Experimental Neuropathology Laboratory, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Insurgentes Sur 3877, Mexico City 14269, Mexico;
| | - Daniel Rembao-Bojórquez
- Neuropathology Department, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Insurgentes Sur 3877, Mexico City 14269, Mexico; (D.R.-B.); (M.L.T.-S.); (S.Z.-V.); (N.G.-H.)
| | - Martha L. Tena-Suck
- Neuropathology Department, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Insurgentes Sur 3877, Mexico City 14269, Mexico; (D.R.-B.); (M.L.T.-S.); (S.Z.-V.); (N.G.-H.)
| | - Sergio Zavala-Vega
- Neuropathology Department, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Insurgentes Sur 3877, Mexico City 14269, Mexico; (D.R.-B.); (M.L.T.-S.); (S.Z.-V.); (N.G.-H.)
| | - Noemí Gelista-Herrera
- Neuropathology Department, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Insurgentes Sur 3877, Mexico City 14269, Mexico; (D.R.-B.); (M.L.T.-S.); (S.Z.-V.); (N.G.-H.)
| | - Miguel A. Ramos-Peek
- Neurosurgery Division, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Insurgentes Sur 3877, Mexico City 14269, Mexico; (M.A.R.-P.); (J.L.G.-A.)
| | - Juan L. Gómez-Amador
- Neurosurgery Division, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Insurgentes Sur 3877, Mexico City 14269, Mexico; (M.A.R.-P.); (J.L.G.-A.)
| | - Febe Cazares-Raga
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies of National Polytechnic Institute, IPN Avenue 2508, Mexico City 07360, Mexico; (F.C.-R.); (F.d.l.C.H.-H.)
| | - Fidel de la Cruz Hernández-Hernández
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies of National Polytechnic Institute, IPN Avenue 2508, Mexico City 07360, Mexico; (F.C.-R.); (F.d.l.C.H.-H.)
| | - Alma Ortiz-Plata
- Experimental Neuropathology Laboratory, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Insurgentes Sur 3877, Mexico City 14269, Mexico;
- Correspondence: ; Tel.: +52-(55)5606-3822 (ext. 2008)
| |
Collapse
|
7
|
Jiang K, Du F, lv L, Zhuo H, Xu T, Peng L, Chen Y, Li L, Zhang J. Genetic Fine Mapping and Genomic Annotation Defines Causal Mechanisms at A Novel Colorectal Cancer Susceptibility Locus in Han Chinese. J Cancer 2020; 11:6841-6849. [PMID: 33123275 PMCID: PMC7592009 DOI: 10.7150/jca.47189] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 09/14/2020] [Indexed: 12/30/2022] Open
Abstract
Genome-wide association studies of colorectal cancer (CRC) have identified two risk SNPs. The characterization of these risk regions in diverse racial groups with different linkage disequilibrium structure would aid in localizing the causal variants. Herein, fine mapping of the established CRC loci was carried out in 1,508 cases and 1,482 controls obtained from the Han Chinese population. One distinct association signal was identified at these loci, where fine mapping implicated rs1010208 as a functional locus. Next, the candidate target genes of functional SNP rs1010208 were analyzed using data from TCGA databases by expression quantitative trait loci analysis method; the data from Peking University People's Hospital were utilized for verification. The dual-luciferase reporter system analysis confirmed that rs1010208 is a regulatory region that can be mutated to decrease the expression of HINT1, resulting in proliferation and invasiveness of CRC.
Collapse
Affiliation(s)
- Kewei Jiang
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, China
| | - Fengying Du
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Liang lv
- Department of General Surgery, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hongqing Zhuo
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Tao Xu
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Lipan Peng
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Yuezhi Chen
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Leping Li
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Jizhun Zhang
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
8
|
Duan DD, Xie H, Shi HF, Huang WW, Ding F, Hong JK, Fan JS, Hu SY, Wang QW, Zhou MQ. Hint1 Overexpression Inhibits the Cell Cycle and Induces Cell Apoptosis in Human Osteosarcoma Cells. Onco Targets Ther 2020; 13:8223-8232. [PMID: 32884300 PMCID: PMC7443417 DOI: 10.2147/ott.s242344] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 05/10/2020] [Indexed: 12/19/2022] Open
Abstract
Background New evidence suggests that histidine triad nucleotide-binding protein 1 (Hint1) exerts a tumor suppressor effect in various human tumors, such as colorectal cancer and gastric cancer. However, it has not been reported whether Hint1 is involved in the occurrence and development of osteosarcoma (OS). Materials and Methods The present study investigated the role of Hint1 in human OS cells by using cell lines, including 143B, U2OS, KHOS-240S, Saos-2 and MG-63. Cell proliferation and apoptosis were detected by flow cytometry. Results The present result revealed that Hint1 is downregulated in these cell lines. The overexpression of Hint1 by adenovirus transfection in 143B and MG63 cell lines suppressed the proliferation and cell cycle, and increased the cell apoptosis. Mechanically, it was found that Hint1 downregulated the cyclin D1 expression via FOXO1 inhibition. Furthermore, FOXO1 overexpression in the 143B and MG63 cell lines significantly blurred the effects of Hint1 on cellular proliferation and apoptosis. Conclusion The present study indicates that Hint1 inhibits the development of OS by regulating FoxO1-cyclin D1, suggesting that Hint1 may be a new method for the treatment of OS.
Collapse
Affiliation(s)
- Dong-Dong Duan
- Department of Orthopedic Surgery, The First People's Hospital of Jingmen, Jingmen, Hubei 448000, People's Republic of China
| | - Hui Xie
- Department of Orthopedic Surgery, The First People's Hospital of Jingmen, Jingmen, Hubei 448000, People's Republic of China
| | - Hua-Feng Shi
- Department of Orthopedic Surgery, The First People's Hospital of Jingmen, Jingmen, Hubei 448000, People's Republic of China
| | - Wen-Wen Huang
- Department of Orthopedic Surgery, The First People's Hospital of Jingmen, Jingmen, Hubei 448000, People's Republic of China
| | - Fan Ding
- Department of Orthopedic Surgery, The First People's Hospital of Jingmen, Jingmen, Hubei 448000, People's Republic of China
| | - Jia-Kun Hong
- Department of Orthopedic Surgery, The First People's Hospital of Jingmen, Jingmen, Hubei 448000, People's Republic of China
| | - Jun-Sheng Fan
- Department of Orthopedic Surgery, The First People's Hospital of Jingmen, Jingmen, Hubei 448000, People's Republic of China
| | - Shou-Yong Hu
- Department of Orthopedic Surgery, The First People's Hospital of Jingmen, Jingmen, Hubei 448000, People's Republic of China
| | - Qing-Wei Wang
- Department of Orthopedic Surgery, The First People's Hospital of Jingmen, Jingmen, Hubei 448000, People's Republic of China
| | - Meng-Qiao Zhou
- Department of Cardiology, The First People's Hospital of Jingmen, Jingmen, Hubei 448000, People's Republic of China
| |
Collapse
|
9
|
Knockdown of Orphan Transporter SLC22A18 Impairs Lipid Metabolism and Increases Invasiveness of HepG2 Cells. Pharm Res 2019; 36:39. [PMID: 30635741 DOI: 10.1007/s11095-018-2565-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 12/26/2018] [Indexed: 12/19/2022]
Abstract
PURPOSE The aim of this work is to investigate the roles of solute carrier family 22 member 18 (SLC22A18) in lipid metabolism and in establishing the tumor phenotype of HepG2 cells. METHODS SLC22A18-knockdown HepG2 cells were established by stable transfection with shRNA. Protein expression levels were measured by quantitative proteomics and Western blot analysis. Cell growth was examined by cell counting kit. Accumulation of triglyceride-rich lipid droplets was measured by Oil-Red O staining. Cell migration and invasion were examined by Transwell assays. RESULTS SLC22A18-knockdown HepG2 cells accumulated triglyceride-rich lipid droplets and showed decreased expression levels of lysosomal/autophagic proteins, suggesting that lipid degradation is suppressed. Growth of HepG2 cells was decreased by SLC22A18 knockdown, but was restored by free fatty acid supplementation. In addition, SLC22A18 knockdown decreased the expression of insulin-like growth factor-binding protein 1 (IGFBP-1) and increased the invasion ability of HepG2 cells. Exogenous IGFBP-1 blocked the increase of invasion activity induced by SLC22A18 knockdown. CONCLUSION Our results suggest that suppression of SLC22A18 decreased the supply of intracellular free fatty acids from triglyceride-rich lipid droplets by impairing the lysosomal/autophagy degradation pathway and reduced the invasive activity of HepG2 cells by decreasing IGFBP-1 expression.
Collapse
|
10
|
Bao T, Ke Y, Wang Y, Wang W, Li Y, Wang Y, Kui X, Zhou Q, Zhou H, Zhang C, Zhou D, Wang L, Xiao C. Taraxasterol suppresses the growth of human liver cancer by upregulating Hint1 expression. J Mol Med (Berl) 2018; 96:661-672. [PMID: 29806073 DOI: 10.1007/s00109-018-1652-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Revised: 05/07/2018] [Accepted: 05/11/2018] [Indexed: 02/06/2023]
Abstract
UNLABELLED Taraxasterol has potent anti-inflammatory and anti-tumor activity. However, the effect and potential mechanisms of Taraxasterol on the growth of human liver cancer have not been clarified. Histidine triad nucleotide-binding protein 1 (Hint1) is a tumor suppressor and its downregulated expression is associated with the development of cancer. Here, we report that Taraxasterol treatment significantly suppressed cell proliferation and induced cell cycle arrest at G0/G1 phase and apoptosis in liver cancer cells, but not in non-tumor hepatocytes. Furthermore, Taraxasterol upregulated Hint1 and Bax, but downregulated Bcl2 and cyclin D1 expression, accompanied by promoting the demethylation in the Hint1 promoter region in liver cancer cells. The effects of Taraxasterol were abrogated by Hint1 silencing and partially mitigated by Bax silencing, Bcl2 or cyclin D1 over-expression in HepG2 cells. Moreover, oral administration with Taraxasterol did not affect body weight, urinary protein levels, and the heart, liver, and kidney morphology in BALB/c mice but effectively inhibited the growth of implanted SK-Hep1 tumor in vivo. Collectively, we demonstrate that Taraxasterol inhibits the growth of liver cancer at least partially by enhancing Hint1 expression to regulate Bax, Bcl2, and cyclin D1 expression. Taraxasterol may be a drug candidate for the treatment of human liver cancer. KEY MESSAGES Taraxasterol inhibits growth and induces apoptosis in human liver cancer cells. Taraxasterol enhances Hint1 expression by promoting demethylation in Hint1 promoter. Taraxasterol increases Hint1 levels to regulate Bax, Bcl2, and cyclinD1 expression. The effects of Taraxasterol are abrogated by Hint1 silencing in liver cancer cells. Taraxasterol inhibits the growth of subcutaneously implanted liver cancers in mice.
Collapse
Affiliation(s)
- Tianhao Bao
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, 374 Kunrui Road, Wuhua District, Kunming, 650101, Yunnan, China
- Mental Health Center of Kunming Medical University, Kunming, 650224, China
- School of Medicine, Yunnan University, 2 Cuihu North Road, Wuhua District, Kunming, 650091, Yunnan, China
| | - Yang Ke
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, 374 Kunrui Road, Wuhua District, Kunming, 650101, Yunnan, China
| | - Yifan Wang
- School of Stomatology, Shandong University, Jinan, 250012, China
| | - Weiwei Wang
- Department of Cardiology, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101, China
| | - Yuehua Li
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, 374 Kunrui Road, Wuhua District, Kunming, 650101, Yunnan, China
| | - Yan Wang
- Department of Pathology, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101, China
| | - Xiang Kui
- Department of Pathology, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101, China
| | - Qixin Zhou
- Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China
| | - Han Zhou
- Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - Cheng Zhang
- Department of Hepatobiliary Surgery, The Sixth People's Hospital of Chengdu, Chengdu, 610051, China
| | - Dongming Zhou
- Environment and Health Research Center, Southwest China Eco-development Academy, Southwest Forestry University, Kunming, 650224, China
| | - Lin Wang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, 374 Kunrui Road, Wuhua District, Kunming, 650101, Yunnan, China.
| | - Chunjie Xiao
- School of Medicine, Yunnan University, 2 Cuihu North Road, Wuhua District, Kunming, 650091, Yunnan, China.
| |
Collapse
|
11
|
Xie H, Yu H, Tian S, Yang X, Wang X, Wang H, Guo Z. MEIS-1 level in unresectable hepatocellular carcinoma can predict the post-treatment outcomes of radiofrequency ablation. Oncotarget 2018; 9:15252-15265. [PMID: 29632641 PMCID: PMC5880601 DOI: 10.18632/oncotarget.24165] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 12/01/2017] [Indexed: 02/06/2023] Open
Abstract
Radiofrequency ablation (RFA) is a local-ablative therapy for unresectable hepatocellular carcinoma (HCC). At present, there is no predictive marker for RFA treatment outcomes. This work aimed to valuate myeloid ecotropic viral integration site 1 (MEIS-1) in predicting post-RFA treatment outcomes of unresectable HCC patients. The time to progression (TTP) and overall survival (OS) of 81 HCC patients who received RFA treatment were measured. The protein level of MEIS-1 in tumor specimens was measured by western blot. The role of MEIS-1 in RFA-treating HCC in vivo growth nude mouse model was examined via PET/CT imaging. Higher level of MEIS-1 in tumor tissue is associated with better RFA treatment outcomes. The median TTP was 9.0 (95% confidence interval [CI]: 6.8-11.3) months in patients with high MEIS-1 expression (n = 43) versus 6.0 (95% CI: 4.6-7.4) months in patients with low MEIS-1 expression (n = 38). Moreover, in rodent HCC model we found overexpression of MEIS-1 enhanced the anti-tumor effect of RFA treatment. We conclude that high level of MEIS-1 expression predicts better RFA treatment outcome in HCC.
Collapse
Affiliation(s)
- Hui Xie
- Department of Interventional Therapy, Tianjin Medical University Cancer Institute & Hospital, National Clinical Cancer Research Center, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300070, PR China
- Department of interventional therapy, 302 Hospital of People's Liberation Army, Beijing 100039, PR China
| | - Haipeng Yu
- Department of Interventional Therapy, Tianjin Medical University Cancer Institute & Hospital, National Clinical Cancer Research Center, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300070, PR China
| | - Shengtao Tian
- Department of interventional therapy, 302 Hospital of People's Liberation Army, Beijing 100039, PR China
| | - Xueling Yang
- Department of Interventional Therapy, Tianjin Medical University Cancer Institute & Hospital, National Clinical Cancer Research Center, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300070, PR China
| | - Ximing Wang
- Department of interventional therapy, 302 Hospital of People's Liberation Army, Beijing 100039, PR China
| | - Huaming Wang
- Department of interventional therapy, 302 Hospital of People's Liberation Army, Beijing 100039, PR China
| | - Zhi Guo
- Department of Interventional Therapy, Tianjin Medical University Cancer Institute & Hospital, National Clinical Cancer Research Center, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300070, PR China
| |
Collapse
|
12
|
Wang WW, Han JH, Wang L, Bao TH. Scutellarin may alleviate cognitive deficits in a mouse model of hypoxia by promoting proliferation and neuronal differentiation of neural stem cells. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2017; 20:272-279. [PMID: 28392899 PMCID: PMC5378964 DOI: 10.22038/ijbms.2017.8355] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Objective(s): Scutellarin, a flavonoid extracted from the medicinal herb Erigeron breviscapus Hand-Mazz, protects neurons from damage and inhibits glial activation. Here we examined whether scutellarin may also protect neurons from hypoxia-induced damage. Materials and Methods: Mice were exposed to hypoxia for 7 days and then administered scutellarin (50 mg/kg/d) or vehicle for 30 days Cognitive impairment in the two groups was assessed using the Morris water maze test, cell proliferation in the hippocampus was compared using 5-bromo-2-deoxyuridine (BrdU) immunohistochemistry, and hippocampal levels of nestin and neuronal class III β-tubulin (Tuj-1) were measured using Western blotting. These results were validated in vitro by treating cultured neural stem cells (NSCs) with scutellarin (30 μM). Results: Treating mice with scutellarin shortened escape times and increased the number of platform crossings, it increased the number of BrdU-positive proliferating cells in the hippocampus, and it up-regulated expression of nestin and Tuj-1. Treating NSC cultures with scutellarin increased the number of proliferating cells and the proportion of cells differentiating into neurons instead of astrocytes. The increase in NSC proliferation was associated with phosphorylation of extracellular signal-regulated kinase (ERK) 1/2, while neuronal differentiation was associated with altered expression of differentiation-related genes. Conclusion: Scutellarin may alleviate cognitive impairment in a mouse model of hypoxia by promo-ting proliferation and neuronal differentiation of NSCs.
Collapse
Affiliation(s)
- Wei-Wei Wang
- Department of Cardiology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, PR China; Key Laboratory of Stem Cells and Regenerative Medicine, Institute of Molecular and Clinical Medicine, Kunming Medical University, Kunming, Yunnan, PR China; Department of Anatomy and Development Biology, Monash University, Clayton, vic 3800, Australia
| | - Jian-Hong Han
- The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, PR China
| | - Lin Wang
- The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, PR China
| | - Tian-Hao Bao
- The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, PR China; Mental Health Center of Kunming Medical University, Kunming City, Yunnan Province, PR China
| |
Collapse
|
13
|
Ke Y, Bao T, Wu X, Tang H, Wang Y, Ge J, Fu B, Meng X, Chen L, Zhang C, Tan Y, Chen H, Guo Z, Ni F, Lei X, Shi Z, Wei D, Wang L. Scutellarin suppresses migration and invasion of human hepatocellular carcinoma by inhibiting the STAT3/Girdin/Akt activity. Biochem Biophys Res Commun 2017; 483:509-515. [PMID: 27998773 DOI: 10.1016/j.bbrc.2016.12.114] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Accepted: 12/17/2016] [Indexed: 12/15/2022]
Abstract
Scutellarin is an active flavone from Erigeron breviscapine (vant) Hand Mass. This study aimed to investigate the potential role of scutellarin in migration and invasion of human hepatocellular carcinoma (HCC) cells and its possible mechanism. In comparison with the vehicle-treated controls, treatment with scutellarin (50 mg/kg/day) for 35 days significantly mitigated the lung and intrahepatic metastasis of HCC tumors in vivo. Scutellarin treatment significantly reduced HepG2 cell viability in a dose-dependent manner, and inhibited migration and invasion of HCC cells in vitro. Scutellarin treatment significantly reduced STAT3 and Girders of actin filaments (Girdin) expression, STAT3 and Akt phosphorylation in HCC cells. Introduction of STAT3 overexpression restored the scutellarin-downregulated Girdin expression, Akt activation, migration and invasion of HCC cells. Furthermore, induction of Girdin overexpression completely abrogated the inhibition of scutellarin on the Akt phosphorylation, migration and invasion of HCC cells. Scutellarin can inhibit HCC cell metastasis in vivo, and migration and invasion in vitro by down-regulating the STAT3/Girdin/Akt signaling.
Collapse
Affiliation(s)
- Yang Ke
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Tianhao Bao
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, China; Mental Health Center, Kunming Medical University, Kunming, China
| | - Xuesong Wu
- Department of Gastroenterological Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Haoran Tang
- Department of Gastroenterological Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yan Wang
- Department of Pathology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jiayun Ge
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Bimang Fu
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xu Meng
- Intensive Care Unit, The First Hospital of Kunming, Kunming, China
| | - Li Chen
- Department of Pediatric Surgery, The Kunming Children's Hospital, Kunming, China
| | - Cheng Zhang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yuqi Tan
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Haotian Chen
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Zhitang Guo
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Fan Ni
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xuefen Lei
- Department of Oncology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Zhitian Shi
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Dong Wei
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Lin Wang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, China.
| |
Collapse
|