1
|
Todorova VK, Byrum SD, Mackintosh SG, Jamshidi-Parsian A, Gies AJ, Washam CL, Jenkins SV, Spiva T, Bowman E, Reyna NS, Griffin RJ, Makhoul I. Exosomal MicroRNA and Protein Profiles of Hepatitis B Virus-Related Hepatocellular Carcinoma Cells. Int J Mol Sci 2023; 24:13098. [PMID: 37685904 PMCID: PMC10487651 DOI: 10.3390/ijms241713098] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/14/2023] [Accepted: 08/18/2023] [Indexed: 09/10/2023] Open
Abstract
Infection with hepatitis B virus (HBV) is a main risk factor for hepatocellular carcinoma (HCC). Extracellular vesicles, such as exosomes, play an important role in tumor development and metastasis, including regulation of HBV-related HCC. In this study, we have characterized exosome microRNA and proteins released in vitro from hepatitis B virus (HBV)-related HCC cell lines SNU-423 and SNU-182 and immortalized normal hepatocyte cell lines (THLE2 and THLE3) using microRNA sequencing and mass spectrometry. Bioinformatics, including functional enrichment and network analysis, combined with survival analysis using data related to HCC in The Cancer Genome Atlas (TCGA) database, were applied to examine the prognostic significance of the results. More than 40 microRNAs and 200 proteins were significantly dysregulated (p < 0.05) in the exosomes released from HCC cells in comparison with the normal liver cells. The functional analysis of the differentially expressed exosomal miRNAs (i.e., mir-483, mir-133a, mir-34a, mir-155, mir-183, mir-182), their predicted targets, and exosomal differentially expressed proteins (i.e., POSTN, STAM, EXOC8, SNX9, COL1A2, IDH1, FN1) showed correlation with pathways associated with HBV, virus activity and invasion, exosome formation and adhesion, and exogenous protein binding. The results from this study may help in our understanding of the role of HBV infection in the development of HCC and in the development of new targets for treatment or non-invasive predictive biomarkers of HCC.
Collapse
Affiliation(s)
- Valentina K. Todorova
- Department of Internal Medicine/Division of Hematology/Oncology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| | - Stephanie D. Byrum
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (S.D.B.); (S.G.M.); (A.J.G.); (C.L.W.)
| | - Samuel G. Mackintosh
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (S.D.B.); (S.G.M.); (A.J.G.); (C.L.W.)
| | - Azemat Jamshidi-Parsian
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (A.J.-P.); (S.V.J.); (R.J.G.)
| | - Allen J. Gies
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (S.D.B.); (S.G.M.); (A.J.G.); (C.L.W.)
| | - Charity L. Washam
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (S.D.B.); (S.G.M.); (A.J.G.); (C.L.W.)
| | - Samir V. Jenkins
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (A.J.-P.); (S.V.J.); (R.J.G.)
| | - Timothy Spiva
- Biology Department, Ouachita Baptist University, Arkadelphia, AR 71998, USA; (T.S.); (E.B.); (N.S.R.)
| | - Emily Bowman
- Biology Department, Ouachita Baptist University, Arkadelphia, AR 71998, USA; (T.S.); (E.B.); (N.S.R.)
| | - Nathan S. Reyna
- Biology Department, Ouachita Baptist University, Arkadelphia, AR 71998, USA; (T.S.); (E.B.); (N.S.R.)
| | - Robert J. Griffin
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (A.J.-P.); (S.V.J.); (R.J.G.)
| | - Issam Makhoul
- Department of Internal Medicine/Division of Hematology/Oncology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| |
Collapse
|
2
|
LncRNA TLR8-AS1 suppresses miR-34a maturation in hepatocellular carcinoma to suppress cell proliferation and migration. Mamm Genome 2021; 32:530-536. [PMID: 34665305 DOI: 10.1007/s00335-021-09920-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 09/17/2021] [Indexed: 12/24/2022]
Abstract
TLR8-AS1 has been characterized as an oncogenic lncRNA in ovarian cancer, while its role in hepatocellular carcinoma (HCC) is unknown. This study aimed to explore the role of TLR8-AS1 in HCC. TLR8-AS1 expression in HCC and paired non-tumor tissues from 62 HCC patients was determined by RT-qPCR. The prognostic value of TLR8-AS1 for HCC was analyzed by performing a 5-year follow-up. Correlations between TLR8-AS1 and mature miR-34a and miR-34a precursor were analyzed by Pearson's correlation coefficient. The roles of TLR8-AS1 and miR-34a in regulating the proliferation and migration were explored by CCK-8 assay and Transwell migration assay. We found that TLR8-AS1 was upregulated in HCC and predicted poor survival. Across HCC tissues, TLR8-AS1 was inversely correlated with mature miR-34a, but not miR-34a precursor. In HCC cells, TLR8-AS1 overexpression downregulated mature miR-34a, but not miR-34a precursor. Cell proliferation and Transwell migration assay showed that TLR8-AS1 overexpression reduced the enhancing effects of miR-34a on cell proliferation and migration. TLR8-AS1 may suppress miR-34a maturation in HCC to suppress cell proliferation and migration.
Collapse
|
3
|
Efficacy of Sorafenib Combined with Interventional Therapy on Primary Liver Cancer Patients and Its Effect on Serum AFP, VEGF, and GGT. JOURNAL OF ONCOLOGY 2021; 2021:9120265. [PMID: 34434234 PMCID: PMC8382533 DOI: 10.1155/2021/9120265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 07/30/2021] [Indexed: 11/18/2022]
Abstract
Objective To explore the efficacy of sorafenib combined with interventional therapy on primary liver cancer (PLC) patients and its effect on serum AFP, VEGF, and GGT. Methods 120 PLC patients admitted to our hospital from January 2016 to January 2020 were selected as the research object and divided into group A and group B according to the admission order, with 60 cases each. Interventional therapy was performed to both groups, and sorafenib was given to group A additionally to compare their treatment effect, survival, adverse reaction rate (ARR), and serum AFP, VEGF, and GGT levels. Results After treatment, group A obtained significantly higher objective remission rate (ORR) and disease control rate (DCR) (p < 0.05), higher one-year survival rate and two-year survival rate (p < 0.05), lower ARR of skin reactions, gastrointestinal reactions, hepatorenal reactions, and hyperbilirubinemia (p < 0.05), and lower serum AFP, VEGF, and GGT levels (p < 0.001). Conclusion The combination of sorafenib and interventional therapy can inhibit the growth and migration of PLC, improve the immune function, prolong the survival period of patients, and lower ARR, so it should be promoted in practice.
Collapse
|
4
|
Feng YQ, Li BA, Feng F, Chen YS, Ren YX, Zhang H, Cao S. Novel mTOR Inhibitor Enhances the Sensitivity of Hepatocellular Carcinoma Cells to Molecular Targeting Agents. Onco Targets Ther 2020; 13:7165-7176. [PMID: 32801748 PMCID: PMC7394584 DOI: 10.2147/ott.s244474] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Accepted: 07/02/2020] [Indexed: 12/14/2022] Open
Abstract
Background Although molecular-targeted agents are still the first choice for advanced hepatocellular carcinoma (HCC) treatment, the therapeutic efficacy of these agents is not satisfactory. Recently, the mammalian target of rapamycin (mTOR) is considered to be a promising molecular target that can enhance the sensitivity of HCC cells to antitumor therapy. However, the reported mTOR inhibitors have some shortcomings, and novel mTOR inhibitors need to be developed to enhance the antitumor effect of molecularly targeted agents on advanced HCC. Methods In this study, five small-molecular compounds that could serve as potential mTOR-specific inhibitors were identified by virtual screening. The activity of tert-butyl (4-(9-(2-(1,3-dioxolan-2-yl)ethyl)-6-morpholino-9H-purin-2-yl)phenyl)carbamate (compound 4) was measured by enzyme test and Western blot, and its antitumor effect on HCC was examined in nude mice subcutaneous tumor model. Results The results showed that 4 is the most effective one in inhibiting the activation of mTOR kinase (mTOR IC50 = 17.52±3.67 nmol/L) among the five lead compounds. Further research in this study indicated that treatment with 4 enhanced the sensitivity of HCC cells to the molecular-targeted agents, such as sorafenib, regorafenib, lenvatinib, anlotinib, and apatinib. In addition, this research indicated that mTOR was correlated with the poor prognosis in patients with advanced HCC who received sorafenib. Conclusion Our study identified a new type of small-molecular inhibitors of mTOR and confirmed their ability to enhance the antitumor effect of molecular-targeted agents on advanced HCC.
Collapse
Affiliation(s)
- Ying-Qi Feng
- Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan 430072, People's Republic of China
| | - Bo-An Li
- Center for Clinical Laboratory, The Fifth Medical Center, General Hospital of Chinese PLA, Beijing 100039, People's Republic of China
| | - Fan Feng
- Center for Clinical Laboratory, The Fifth Medical Center, General Hospital of Chinese PLA, Beijing 100039, People's Republic of China
| | - Yong-Shou Chen
- Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan 430072, People's Republic of China
| | - Yi-Xin Ren
- Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan 430072, People's Republic of China
| | - Heng Zhang
- Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan 430072, People's Republic of China
| | - Shuang Cao
- Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan 430072, People's Republic of China
| |
Collapse
|
5
|
Feng YQ, Gu SX, Chen YS, Gao XD, Ren YX, Chen JC, Lu YY, Zhang H, Cao S. Virtual Screening and Optimization of Novel mTOR Inhibitors for Radiosensitization of Hepatocellular Carcinoma. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:1779-1798. [PMID: 32440103 PMCID: PMC7220363 DOI: 10.2147/dddt.s249156] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 04/09/2020] [Indexed: 12/12/2022]
Abstract
Background Radiotherapy has an ameliorative effect on a wide variety of tumors, but hepatocellular carcinoma (HCC) is insensitive to this treatment. Overactivated mammalian target of rapamycin (mTOR) plays an important part in the resistance of HCC to radiotherapy; thus, mTOR inhibitors have potential as novel radiosensitizers to enhance the efficacy of radiotherapy for HCC. Methods A lead compound was found based on pharmacophore modeling and molecular docking, and optimized according to the differences between the ATP-binding pockets of mTOR and PI3K. The radiosensitizing effect of the optimized compound (2a) was confirmed by colony formation assays and DNA double-strand break assays in vitro. The discovery and preclinical characteristics of this compound are described. Results The key amino acid residues in mTOR were identified, and a precise virtual screening model was constructed. Compound 2a, with a 4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidine scaffold, exhibited promising potency against mTOR (mTOR IC50=7.1 nmol/L (nM)) with 126-fold selectivity over PI3Kα. Moreover, 2a significantly enhanced the sensitivity of HCC to radiotherapy in vitro in a dose-dependent manner. Conclusion A new class of selective mTOR inhibitors was developed and their radiosensitization effects were confirmed. This study also provides a basis for developing mTOR-specific inhibitors for use as radiosensitizers for HCC radiotherapy.
Collapse
Affiliation(s)
- Ying-Qi Feng
- Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan 430072, People's Republic of China
| | - Shuang-Xi Gu
- Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan 430072, People's Republic of China
| | - Yong-Shou Chen
- Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan 430072, People's Republic of China
| | - Xu-Dong Gao
- Comprehensive Liver Cancer Department, The Fifth Medical Center, Chinese PLA General Hospital, Beijing 100039, People's Republic of China
| | - Yi-Xin Ren
- Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan 430072, People's Republic of China
| | - Jian-Chao Chen
- Key Laboratory of Structure-Based Drug Design and Discovery, Shenyang Pharmaceutical University, Ministry of Education, Shenyang 110016, People's Republic of China
| | - Yin-Ying Lu
- Comprehensive Liver Cancer Department, The Fifth Medical Center, Chinese PLA General Hospital, Beijing 100039, People's Republic of China
| | - Heng Zhang
- Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan 430072, People's Republic of China
| | - Shuang Cao
- Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan 430072, People's Republic of China.,National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, People's Republic of China
| |
Collapse
|
6
|
Ferrín G, Guerrero M, Amado V, Rodríguez-Perálvarez M, De la Mata M. Activation of mTOR Signaling Pathway in Hepatocellular Carcinoma. Int J Mol Sci 2020; 21:ijms21041266. [PMID: 32070029 PMCID: PMC7072933 DOI: 10.3390/ijms21041266] [Citation(s) in RCA: 215] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Revised: 02/10/2020] [Accepted: 02/11/2020] [Indexed: 12/17/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the most frequent primary liver cancer and occurs mainly in patients with liver cirrhosis. The mammalian target of rapamycin (mTOR) signaling pathway is involved in many hallmarks of cancer including cell growth, metabolism re-programming, proliferation and inhibition of apoptosis. The mTOR pathway is upregulated in HCC tissue samples as compared with the surrounding liver cirrhotic tissue. In addition, the activation of mTOR is more intense in the tumor edge, thus reinforcing its role in HCC proliferation and spreading. The inhibition of the mTOR pathway by currently available pharmacological compounds (i.e., sirolimus or everolimus) is able to hamper tumor progression both in vitro and in animal models. The use of mTOR inhibitors alone or in combination with other therapies is a very attractive approach, which has been extensively investigated in humans. However, results are contradictory and there is no solid evidence suggesting a true benefit in clinical practice. As a result, neither sirolimus nor everolimus are currently approved to treat HCC or to prevent tumor recurrence after curative surgery. In the present comprehensive review, we analyzed the most recent scientific evidence while providing some insights to understand the gap between experimental and clinical studies.
Collapse
Affiliation(s)
- Gustavo Ferrín
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Universidad de Córdoba, 14004 Córdoba, Spain; (G.F.); (M.G.); (V.A.); (M.D.l.M.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 14004 Córdoba, Spain
| | - Marta Guerrero
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Universidad de Córdoba, 14004 Córdoba, Spain; (G.F.); (M.G.); (V.A.); (M.D.l.M.)
- Department of Hepatology and Liver Transplantaton, Hospital Universitario Reina Sofía, 14004 Córdoba, Spain
| | - Víctor Amado
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Universidad de Córdoba, 14004 Córdoba, Spain; (G.F.); (M.G.); (V.A.); (M.D.l.M.)
- Department of Hepatology and Liver Transplantaton, Hospital Universitario Reina Sofía, 14004 Córdoba, Spain
| | - Manuel Rodríguez-Perálvarez
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Universidad de Córdoba, 14004 Córdoba, Spain; (G.F.); (M.G.); (V.A.); (M.D.l.M.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 14004 Córdoba, Spain
- Department of Hepatology and Liver Transplantaton, Hospital Universitario Reina Sofía, 14004 Córdoba, Spain
- Correspondence: ; Tel.: +34-617854692
| | - Manuel De la Mata
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Universidad de Córdoba, 14004 Córdoba, Spain; (G.F.); (M.G.); (V.A.); (M.D.l.M.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 14004 Córdoba, Spain
- Department of Hepatology and Liver Transplantaton, Hospital Universitario Reina Sofía, 14004 Córdoba, Spain
| |
Collapse
|
7
|
Protein Kinase B Inactivation Is Associated with Magnolol-Enhanced Therapeutic Efficacy of Sorafenib in Hepatocellular Carcinoma In Vitro and In Vivo. Cancers (Basel) 2019; 12:cancers12010087. [PMID: 31905887 PMCID: PMC7017147 DOI: 10.3390/cancers12010087] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 12/03/2019] [Accepted: 12/20/2019] [Indexed: 12/11/2022] Open
Abstract
Although sorafenib, an oral multikinase inhibitor, was approved as a treatment drug of advance hepatocellular carcinoma (HCC), treatment efficacy still requires improvement. Searching for the adjuvant reagent for enhancing sorafenib efficacy remains as a critical issue. Sorafenib has been proved to suppress extracellular signal-regulated kinases (ERK) in HCC; however, protein kinase B (AKT) was not affected by it. Targeting AKT in combination with sorafenib could be an important breakthrough point of HCC treatment. Many herbal compounds and composite formulas have been shown to enhance anti-HCC activity of sorafenib. Magnolol is a bioactive compound extracted from the bark of the Magnolia officinalis and has been shown to induce apoptosis and inhibit cell invasion in HCC in vitro. However, whether magnolol sensitizes HCC to sorafenib is ambiguous. In this study, we indicated that magnolol significantly enhanced sorafenib-diminished tumor cell growth, expression of anti-apoptotic proteins, and migration/invasion ability compared to sorafenib alone. Magnolol significantly boosted sorafenib-induced extrinsic/intrinsic dependent apoptosis pathways in HCC. Notably sorafenib could not reduce protein level of AKT (Ser473), but expression of AKT (Ser473) was significantly decreased by magnolol or magnolol combined with sorafenib. LY294002 as specific AKT inhibitor was used to confirm that AKT inactivation may promote anticancer effect of sorafenib. Taken together, AKT inhibition is associated with magnolol-enhanced the therapeutic effect of sorafenib in HCC. We suggested magnolol as the potential adjuvant which may enhance therapeutic benefits of sorafenib in patients with HCC.
Collapse
|
8
|
Ruso-Julve F, Pombero A, Pilar-Cuéllar F, García-Díaz N, Garcia-Lopez R, Juncal-Ruiz M, Castro E, Díaz Á, Vazquez-Bourgón J, García-Blanco A, Garro-Martinez E, Pisonero H, Estirado A, Ayesa-Arriola R, López-Giménez J, Mayor F, Valdizán E, Meana J, Gonzalez-Maeso J, Martínez S, Vaqué JP, Crespo-Facorro B. Dopaminergic control of ADAMTS2 expression through cAMP/CREB and ERK: molecular effects of antipsychotics. Transl Psychiatry 2019; 9:306. [PMID: 31740729 PMCID: PMC6861307 DOI: 10.1038/s41398-019-0647-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 10/08/2019] [Accepted: 10/20/2019] [Indexed: 02/07/2023] Open
Abstract
A better understanding of the molecular mechanisms that participate in the development and clinical manifestations of schizophrenia can lead to improve our ability to diagnose and treat this disease. Previous data strongly associated the levels of deregulated ADAMTS2 expression in peripheral blood mononuclear cells (PBMCs) from patients at first episode of psychosis (up) as well as in clinical responders to treatment with antipsychotic drugs (down). In this current work, we performed an independent validation of such data and studied the mechanisms implicated in the control of ADAMTS2 gene expression. Using a new cohort of drug-naïve schizophrenia patients with clinical follow-up, we confirmed that the expression of ADAMTS2 was highly upregulated in PBMCs at the onset (drug-naïve patients) and downregulated, in clinical responders, after treatment with antipsychotics. Mechanistically, ADAMTS2 expression was activated by dopaminergic signalling (D1-class receptors) and downstream by cAMP/CREB and mitogen-activated protein kinase (MAPK)/ERK signalling. Incubation with antipsychotic drugs and selective PKA and MEK inhibitors abrogated D1-mediated activation of ADAMTS2 in neuronal-like cells. Thus, D1 receptors signalling towards CREB activation might participate in the onset and clinical responses to therapy in schizophrenia patients, by controlling ADAMTS2 expression and activity. The unbiased investigation of molecular mechanisms triggered by antipsychotic drugs may provide a new landscape of novel targets potentially associated with clinical efficacy.
Collapse
Affiliation(s)
- Fulgencio Ruso-Julve
- Department of Psychiatry, University Hospital Marqués de Valdecilla-IDIVAL, Santander, 39011, Cantabria, Spain
- Department of Molecular Biology, School of Medicine, University of Cantabria, Santander, 39011, Cantabria, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, 28029, Spain
| | - Ana Pombero
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, 28029, Spain
- Instituto de Neurociencias, UMH-CSIC, Alicante, 3550, Spain
| | - Fuencisla Pilar-Cuéllar
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, 28029, Spain
- Instituto de Biomedicina y Biotecnología de Cantabria, IBBTEC (Universidad de Cantabria, CSIC, SODERCAN), 39011, Santander, Cantabria, Spain
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria, Santander, 39011, Cantabria, Spain
| | - Nuria García-Díaz
- Department of Molecular Biology, School of Medicine, University of Cantabria, Santander, 39011, Cantabria, Spain
- Infection, Immunity and Digestive Pathology Group, University Hospital Marqués de Valdecilla-IDIVAL, Santander, 39011, Cantabria, Spain
| | - Raquel Garcia-Lopez
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, 28029, Spain
- Instituto de Neurociencias, UMH-CSIC, Alicante, 3550, Spain
| | - María Juncal-Ruiz
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, 28029, Spain
- Department of Psychiatry, Sierrallana Hospital, Torrelavega, 39300, Cantabria, Spain
| | - Elena Castro
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, 28029, Spain
- Instituto de Biomedicina y Biotecnología de Cantabria, IBBTEC (Universidad de Cantabria, CSIC, SODERCAN), 39011, Santander, Cantabria, Spain
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria, Santander, 39011, Cantabria, Spain
| | - Álvaro Díaz
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, 28029, Spain
- Instituto de Biomedicina y Biotecnología de Cantabria, IBBTEC (Universidad de Cantabria, CSIC, SODERCAN), 39011, Santander, Cantabria, Spain
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria, Santander, 39011, Cantabria, Spain
| | - Javier Vazquez-Bourgón
- Department of Psychiatry, University Hospital Marqués de Valdecilla-IDIVAL, Santander, 39011, Cantabria, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, 28029, Spain
| | - Agustín García-Blanco
- Department of Molecular Biology, School of Medicine, University of Cantabria, Santander, 39011, Cantabria, Spain
- Infection, Immunity and Digestive Pathology Group, University Hospital Marqués de Valdecilla-IDIVAL, Santander, 39011, Cantabria, Spain
| | - Emilio Garro-Martinez
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, 28029, Spain
- Instituto de Biomedicina y Biotecnología de Cantabria, IBBTEC (Universidad de Cantabria, CSIC, SODERCAN), 39011, Santander, Cantabria, Spain
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria, Santander, 39011, Cantabria, Spain
| | - Helena Pisonero
- Department of Molecular Biology, School of Medicine, University of Cantabria, Santander, 39011, Cantabria, Spain
- Infection, Immunity and Digestive Pathology Group, University Hospital Marqués de Valdecilla-IDIVAL, Santander, 39011, Cantabria, Spain
| | - Alicia Estirado
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, 28029, Spain
- Instituto de Neurociencias, UMH-CSIC, Alicante, 3550, Spain
| | - Rosa Ayesa-Arriola
- Department of Psychiatry, University Hospital Marqués de Valdecilla-IDIVAL, Santander, 39011, Cantabria, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, 28029, Spain
| | - Juan López-Giménez
- Institute of Parasitology and Biomedicine "López-Neyra" (IPBLN-CSIC), Armilla, 18016, Granada, Spain
| | - Federico Mayor
- Department of Molecular Biology, Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Universidad Autónoma de Madrid, Madrid, 28049, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, 28029, Spain
| | - Elsa Valdizán
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, 28029, Spain
- Instituto de Biomedicina y Biotecnología de Cantabria, IBBTEC (Universidad de Cantabria, CSIC, SODERCAN), 39011, Santander, Cantabria, Spain
- Department of Physiology and Pharmacology, School of Medicine, University of Cantabria, Santander, 39011, Cantabria, Spain
| | - Javier Meana
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, 28029, Spain
- Department of Pharmacology, University of the Basque Country UPV/EHU, Leioa, 48940, Bizkaia, Spain
| | - Javier Gonzalez-Maeso
- Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, P.O. Box 980551, Molecular Medicine Research Building 5-038, Richmond, 23298, Virginia, USA
| | - Salvador Martínez
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, 28029, Spain
- Instituto de Neurociencias, UMH-CSIC, Alicante, 3550, Spain
| | - José Pedro Vaqué
- Department of Molecular Biology, School of Medicine, University of Cantabria, Santander, 39011, Cantabria, Spain.
- Infection, Immunity and Digestive Pathology Group, University Hospital Marqués de Valdecilla-IDIVAL, Santander, 39011, Cantabria, Spain.
| | - Benedicto Crespo-Facorro
- Department of Psychiatry, University Hospital Marqués de Valdecilla-IDIVAL, Santander, 39011, Cantabria, Spain.
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, 28029, Spain.
- Department of Psychiatry, School of Medicine, University Hospital Virgen del Rocio-IBiS, Sevilla, 41013, Spain.
| |
Collapse
|
9
|
Delman M, Avcı ST, Akçok İ, Kanbur T, Erdal E, Çağır A. Antiproliferative activity of (R)-4'-methylklavuzon on hepatocellular carcinoma cells and EpCAM +/CD133 + cancer stem cells via SIRT1 and Exportin-1 (CRM1) inhibition. Eur J Med Chem 2019; 180:224-237. [PMID: 31306909 DOI: 10.1016/j.ejmech.2019.07.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 07/07/2019] [Accepted: 07/08/2019] [Indexed: 12/12/2022]
Abstract
Cytotoxic effects of (R)-4'-methylklavuzon were investigated on hepatocellular carcinoma cells (HuH-7 and HepG2) and HuH-7 EpCAM+/CD133+ cancer stem cells. IC50 of (R)-4'-methylklavuzon was found as 1.25 μM for HuH-7 parental cells while it was found as 2.50 μM for HuH-7 EpCAM+/CD133+ cancer stem cells. (R)-4'-methylklavuzon tended to show more efficient in vitro cytotoxicity with its lower IC50 values on hepatocellular carcinoma cell lines compared to its lead molecule, goniothalamin and FDA-approved drugs, sorafenib and regorafenib. Cell-based Sirtuin/HDAC enzyme activity measurements revealed that endogenous Sirtuin/HDAC enzymes were reduced by 40% compared to control. SIRT1 protein levels were upregulated indicating triggered DNA repair mechanism. p53 was overexpressed in HepG2 cells. (R)-4'-methylklavuzon inhibited CRM1 protein providing increased retention of p53 and RIOK2 protein in the nucleus. HuH-7 parental and EpCAM+/CD133+ cancer stem cell spheroids lost intact morphology. 3D HepG2 spheroid viabilities were decreased in a correlation with upregulation in p53 protein levels.
Collapse
Affiliation(s)
- Murat Delman
- Department of Biotechnology and Bioengineering, Izmir Institute of Technology, 35430, Urla, Izmir, Turkey
| | - Sanem Tercan Avcı
- Izmir Biomedicine and Genome Center, 35340, Balcova, Izmir, Turkey; Department of Medical Biology and Genetics, Faculty of Medicine, Dokuz Eylul University, 35340, Balcova, Izmir, Turkey
| | - İsmail Akçok
- Department of Chemistry, Faculty of Science, Izmir Institute of Technology, 35430, Urla, Izmir, Turkey
| | - Tuğçe Kanbur
- Department of Chemistry, Faculty of Science, Izmir Institute of Technology, 35430, Urla, Izmir, Turkey
| | - Esra Erdal
- Izmir Biomedicine and Genome Center, 35340, Balcova, Izmir, Turkey; Department of Medical Biology and Genetics, Faculty of Medicine, Dokuz Eylul University, 35340, Balcova, Izmir, Turkey.
| | - Ali Çağır
- Department of Chemistry, Faculty of Science, Izmir Institute of Technology, 35430, Urla, Izmir, Turkey.
| |
Collapse
|