1
|
Ingels A, Scott R, Hooper AR, van der Westhuyzen AE, Wagh SB, de Meester J, Maddau L, Marko D, Aichinger G, Berger W, Vermeersch M, Pérez-Morga D, Maslivetc VA, Evidente A, van Otterlo WAL, Kornienko A, Mathieu V. New hemisynthetic derivatives of sphaeropsidin phytotoxins triggering severe endoplasmic reticulum swelling in cancer cells. Sci Rep 2024; 14:14674. [PMID: 38918539 PMCID: PMC11199504 DOI: 10.1038/s41598-024-65335-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 06/19/2024] [Indexed: 06/27/2024] Open
Abstract
Sphaeropsidins are iso-pimarane diterpenes produced by phytopathogenic fungi that display promising anticancer activities. Sphaeropsidin A, in particular, has been shown to counteract regulatory volume increase, a process used by cancer cells to avoid apoptosis. This study reports the hemi-synthesis of new lipophilic derivatives obtained by modifications of the C15,C16-alkene moiety. Several of these compounds triggered severe ER swelling associated with strong proteasomal inhibition and consequently cell death, a feature that was not observed with respect to mode of action of the natural product. Significantly, an analysis from the National Cancer Institute sixty cell line testing did not reveal any correlations between the most potent derivative and any other compound in the database, except at high concentrations (LC50). This study led to the discovery of a new set of sphaeropsidin derivatives that may be exploited as potential anti-cancer agents, notably due to their maintained activity towards multidrug resistant models.
Collapse
Affiliation(s)
- Aude Ingels
- Department of Pharmacotherapy and Pharmaceutics, Chemistry and Biochemistry, Faculté de Pharmacie, Université Libre de Bruxelles, Brussels, Belgium
- ULB Cancer Research Center, U-CRC, Université Libre de Bruxelles, Brussels, Belgium
| | - Robert Scott
- Department of Chemistry and Biochemistry, Texas State University, San Marcos, TX, 78666, USA
| | - Annie R Hooper
- Department of Chemistry and Biochemistry, Texas State University, San Marcos, TX, 78666, USA
| | - Aletta E van der Westhuyzen
- Department of Chemistry and Polymer Science, University of Stellenbosch, Matieland, Stellenbosch, 7600, South Africa
| | - Sachin B Wagh
- Department of Chemistry and Biochemistry, Texas State University, San Marcos, TX, 78666, USA
| | - Joséphine de Meester
- Department of Chemistry and Polymer Science, University of Stellenbosch, Matieland, Stellenbosch, 7600, South Africa
| | - Lucia Maddau
- Department of Agriculture, Section of Plant Pathology and Entomology, University of Sassari, Sassari, Italy
| | - Doris Marko
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Georg Aichinger
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Walter Berger
- Medical University of Vienna Center for Cancer Research, Vienna, Austria
| | - Marjorie Vermeersch
- Electron Microscopy Laboratory, Center for Microscopy and Molecular Imaging (CMMI), Université Libre de Bruxelles (ULB), Gosselies, Belgium
| | - David Pérez-Morga
- Electron Microscopy Laboratory, Center for Microscopy and Molecular Imaging (CMMI), Université Libre de Bruxelles (ULB), Gosselies, Belgium
| | - Vladimir A Maslivetc
- Department of Chemistry and Biochemistry, Texas State University, San Marcos, TX, 78666, USA
| | - Antonio Evidente
- Institute of Biomolecular Chemistry, National Research Council, Pozzuoli, Italy
| | - Willem A L van Otterlo
- Department of Chemistry and Polymer Science, University of Stellenbosch, Matieland, Stellenbosch, 7600, South Africa
| | - Alexander Kornienko
- Department of Chemistry and Biochemistry, Texas State University, San Marcos, TX, 78666, USA.
| | - Véronique Mathieu
- Department of Pharmacotherapy and Pharmaceutics, Chemistry and Biochemistry, Faculté de Pharmacie, Université Libre de Bruxelles, Brussels, Belgium.
- ULB Cancer Research Center, U-CRC, Université Libre de Bruxelles, Brussels, Belgium.
| |
Collapse
|
2
|
Röth S, Kocaturk NM, Sathyamurthi PS, Carton B, Watt M, Macartney TJ, Chan KH, Isidro-Llobet A, Konopacka A, Queisser MA, Sapkota GP. Identification of KLHDC2 as an efficient proximity-induced degrader of K-RAS, STK33, β-catenin, and FoxP3. Cell Chem Biol 2023; 30:1261-1276.e7. [PMID: 37591251 DOI: 10.1016/j.chembiol.2023.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 05/09/2023] [Accepted: 07/16/2023] [Indexed: 08/19/2023]
Abstract
Targeted protein degradation (TPD), induced by enforcing target proximity to an E3 ubiquitin ligase using small molecules has become an important drug discovery approach for targeting previously undruggable disease-causing proteins. However, out of over 600 E3 ligases encoded by the human genome, just over 10 E3 ligases are currently utilized for TPD. Here, using the affinity-directed protein missile (AdPROM) system, in which an anti-GFP nanobody was linked to an E3 ligase, we screened over 30 E3 ligases for their ability to degrade 4 target proteins, K-RAS, STK33, β-catenin, and FoxP3, which were endogenously GFP-tagged. Several new E3 ligases, including CUL2 diGly receptor KLHDC2, emerged as effective degraders, suggesting that these E3 ligases can be taken forward for the development of small-molecule degraders, such as proteolysis targeting chimeras (PROTACs). As a proof of concept, we demonstrate that a KLHDC2-recruiting peptide-based PROTAC connected to chloroalkane is capable of degrading HALO-GFP protein in cells.
Collapse
Affiliation(s)
- Sascha Röth
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, UK
| | - Nur Mehpare Kocaturk
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, UK
| | - Preethi S Sathyamurthi
- Protein Degradation Group, Medicines Research Centre, GSK, Gunnels Wood Road, Stevenage, UK
| | - Bill Carton
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, UK
| | - Matthew Watt
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, UK
| | - Thomas J Macartney
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, UK
| | - Kwok-Ho Chan
- Protein Degradation Group, Medicines Research Centre, GSK, Gunnels Wood Road, Stevenage, UK
| | - Albert Isidro-Llobet
- Chemical Biology, Medicines Research Centre, GSK, Gunnels Wood Road, Stevenage, UK
| | - Agnieszka Konopacka
- Protein Degradation Group, Medicines Research Centre, GSK, Gunnels Wood Road, Stevenage, UK
| | - Markus A Queisser
- Protein Degradation Group, Medicines Research Centre, GSK, Gunnels Wood Road, Stevenage, UK
| | - Gopal P Sapkota
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, UK.
| |
Collapse
|
3
|
Zheng Z, Wang X, Chen D. Proteasome inhibitor MG132 enhances the sensitivity of human OSCC cells to cisplatin via a ROS/DNA damage/p53 axis. Exp Ther Med 2023; 25:224. [PMID: 37123203 PMCID: PMC10133788 DOI: 10.3892/etm.2023.11924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 02/27/2023] [Indexed: 05/02/2023] Open
Abstract
Cis-diamine-dichloroplatinum II (cisplatin, CDDP) is a key chemotherapeutic regimen in the treatment of oral squamous cell carcinoma (OSCC). However, the therapeutic efficacy of cisplatin in OSCC may be hampered by chemoresistance. Therefore, the development of novel combination therapy strategies to overcome the limitations of CDDP is of great importance. The proteasome inhibitor MG132 exhibits anti-cancer properties against various types of cancer. However, our knowledge of its anti-cancer effects in combination with CDDP in OSCC cells remains limited. In the current study, the synergetic effects of MG132 and CDDP were evaluated in the human CAL27 OSCC cell line. CAL27 cells were treated with CDDP alone or in combination with MG132. The results showed that MG132 significantly reduced cell viability in a dose-dependent manner. Additionally, cell viability was significantly reduced in CAL27 cells treated with 0.2 µM MG132 and 2 µM CDDP compared with cells treated with MG132 or CDDP alone. In addition, MG132 significantly enhanced the CDDP-induced generation of intracellular reactive oxygen species and DNA damage in OSCC cells. Furthermore, treatment with CDDP or MG132 alone notably inhibited colony formation and proliferation of OSCC cells. However, co-treatment of OSCC cells with MG132 and CDDP further hampered colony formation and proliferation compared with cells treated with either MG132 or CDDP alone. Finally, in cells co-treated with MG132 and CDDP, the expression of p53 was markedly elevated and the p53-mediated apoptotic pathway was further activated compared with cells treated with MG132 or CDDP alone, as shown by the enhanced cell apoptosis, Bax upregulation, and Bcl-2 downregulation. Overall, the results of the current study support the synergistic anti-cancer effects of a combination of MG132 and CDDP against OSCC, thus suggesting that the combination of MG132 and CDDP may be a promising therapeutic strategy for the management of OSCC.
Collapse
Affiliation(s)
- Zheng Zheng
- Department of Stomatology, The First People's Hospital of Nantong, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu 226000, P.R. China
| | - Xiang Wang
- Department of Stomatology, The First People's Hospital of Nantong, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu 226000, P.R. China
- Correspondence to: Dr Donglei Chen or Dr Xiang Wang, Department of Stomatology, The First People's Hospital of Nantong, Affiliated Hospital 2 of Nantong University, 6 Haierxiang Road, Nantong, Jiangsu 226000, P.R. China
| | - Donglei Chen
- Department of Stomatology, The First People's Hospital of Nantong, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu 226000, P.R. China
- Correspondence to: Dr Donglei Chen or Dr Xiang Wang, Department of Stomatology, The First People's Hospital of Nantong, Affiliated Hospital 2 of Nantong University, 6 Haierxiang Road, Nantong, Jiangsu 226000, P.R. China
| |
Collapse
|
4
|
Ji J, Zhou BR, Zhang RH, Li HM, Guo Q, Zhu J, Luo D. MG-132 treatment promotes TRAIL-mediated apoptosis in SEB-1 sebocytes. Life Sci 2018; 210:150-157. [PMID: 30176247 DOI: 10.1016/j.lfs.2018.08.068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 08/22/2018] [Accepted: 08/30/2018] [Indexed: 11/19/2022]
Abstract
AIMS This study aimed to identify the mechanism of how MG-132 stimulates cell death in SEB-1 sebocytes. MATERIALS AND METHODS TUNEL staining and annexin-FITC/PI flow cytometry were utilized to examine the apoptotic cell number of SEB-1 sebocytes and HaCaT keratinocytes upon MG-132 and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) treatment. MTT assay and CCK-8 assay monitored the proliferative rate and viability of both cell lines with different treatment. Western blotting (WB) and qPCR were performed to detect the expression of TRAIL and members of Bcl-2 family at protein and gene level. Additionally, RNA interfering was used to knockdown the mRNA transcription of TRAIL and BIK gene. KEY FINDINGS MG-132 treatment enhanced cell death in SEB-1 sebocytes but not in HaCaT keratinocytes. Meanwhile, TRAIL concentrations in SEB-1 sebocytes treated with MG-132 were markedly elevated. Furthermore, treatment with TRAIL or the TRAIL receptor-specific monoclonal antibody AY4 at various doses stimulated cell death in SEB-1 sebocytes in a time- and dose-dependent manner. Silencing of TRAIL restored the cell viability of SEB-1 cells to a normal level after MG-132 treatment. Combined treatment of SEB-1 sebocytes with TRAIL and MG-132 synergistically triggered cell death, suppressed cell proliferation and survival, and promoted BIK expression. Furthermore, BCL2 Interacting Killer (BIK) knockdown via RNA interference participated in the recovery of cell survival reduced by treatment with TRAIL and MG-132. SIGNIFICANCE These findings suggest that treatment with the selective proteasome suppressor MG-132 and TRAIL induces cell death in sebocytes through upregulation of BIK, a member of the Bcl-2 family.
Collapse
Affiliation(s)
- Jin Ji
- Department of Dermatology, the First Affiliated Hospital of Nanjing Medical University
| | - Bing-Rong Zhou
- Department of Dermatology, the First Affiliated Hospital of Nanjing Medical University
| | - Ruo-Hua Zhang
- Department of Dermatology, The Affiliated Hospital of Nanjing University of Chinese Medicine
| | - Hong-Min Li
- Department of Dermatology, The Affiliated Hospital of Nanjing University of Chinese Medicine
| | - Qin Guo
- Department of Dermatology, The Affiliated Hospital of Nanjing University of Chinese Medicine
| | - Jie Zhu
- Department of Dermatology, The Affiliated Hospital of Nanjing University of Chinese Medicine
| | - Dan Luo
- Department of Dermatology, the First Affiliated Hospital of Nanjing Medical University.
| |
Collapse
|
5
|
Wu X, Chen Z, Yang Y, Dong Y, Liu H, Kuang S, Luo K. Impact of proteasome inhibitor MG-132 on expression of NF-κB, IL-1β and histological remodeling after myocardial infarction. Exp Ther Med 2018; 16:1365-1372. [PMID: 30112065 DOI: 10.3892/etm.2018.6308] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 09/01/2017] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to investigate the impact of carbobenzoxy-Leu-Leu-leucinal (MG-132) on myocardial remodeling in rats with myocardial infarction (MI) and investigate the possible underlying mechanisms. The rat model of MI was established, followed by administration of MG-132 (MG group), pyrrolidine dithiocarbamic acid (PDTC group) or normal saline (MI group) for 28 days. The expression of nuclear factor-κB (NF-κB) p65, interleukin 1β (IL-1β) and matrix metalloproteinase 2 (MMP-2), as well as the total volume of collagen and the ratio of type I/III collagen were then detected. Total collagen, including type I and III collagen, and the ratio of type I/III collagen were significantly increased in MI rats compared with those in the sham group (P<0.01), while it was significantly decreased in the PDTC and MG groups compared with that in the MI group (P<0.01). A similar trend was identified for the expression of NF-κB, IL-1β and MMP-2, which was significantly increased in the MI group compared with that in the sham group (P<0.01), while it was significantly decreased in the MG and PDTC groups compared with that in the MI group (P<0.01). In conclusion, MG-132 was demonstrated to improve post-MI tissue remodeling, and the mechanism may be associated with the inhibition of NF-κB activation and the downregulation of inflammatory cytokines, such as IL-1β.
Collapse
Affiliation(s)
- Xinhua Wu
- Department of Cardiology, The First Affiliated Hospital of Dali University, Dali, Yunnan 671000, P.R. China
| | - Zhangrong Chen
- Department of Cardiology, The First Affiliated Hospital of Dali University, Dali, Yunnan 671000, P.R. China
| | - Ying Yang
- Department of Cardiology, The First Affiliated Hospital of Dali University, Dali, Yunnan 671000, P.R. China
| | - Yu Dong
- Department of Cardiology, The First Affiliated Hospital of Dali University, Dali, Yunnan 671000, P.R. China
| | - Hong Liu
- Department of Cardiology, The First Affiliated Hospital of Dali University, Dali, Yunnan 671000, P.R. China
| | - Shiquan Kuang
- Department of Cardiology, The First Affiliated Hospital of Dali University, Dali, Yunnan 671000, P.R. China
| | - Kailiang Luo
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 404100, P.R. China
| |
Collapse
|
6
|
Chen H, Yang H, Pan L, Wang W, Liu X, Ren X, Liu Y, Liu W, Zhang Y, Jiang L, Li K, Zhang B, Wang LX. The molecular mechanisms of XBP-1 gene silencing on IRE1α-TRAF2-ASK1-JNK pathways in oral squamous cell carcinoma under endoplasmic reticulum stress. Biomed Pharmacother 2016; 77:108-13. [DOI: 10.1016/j.biopha.2015.12.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 10/31/2015] [Accepted: 12/15/2015] [Indexed: 11/16/2022] Open
|
7
|
Chen HY, Ren XY, Wang WH, Zhang YX, Chen SF, Zhang B, Wang LX. Upregulated ROS production induced by the proteasome inhibitor MG-132 on XBP1 gene expression and cell apoptosis in Tca-8113 cells. Biomed Pharmacother 2014; 68:709-13. [PMID: 25092240 DOI: 10.1016/j.biopha.2014.07.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 07/08/2014] [Indexed: 12/01/2022] Open
Abstract
Exposure of Tca-8113 cells to proteasome inhibitor carbobenzoxy-Leu-Leu-leucinal (MG-132) causing apoptosis is associated with endoplasmic reticulum (ER) stress. X-box-binding protein-1 (XBP1) is an important regulator of a subset of genes active during ER stress, which is related to cell survival and is required for tumor growth. The present study is to evaluate the effect of MG-132 on ROS production, XBP1 gene expression, tumor necrosis factor receptor-associated factor 2 (TRAF2), ASK1 and c-jun protein expression in tongue squamous cell carcinoma cell line Tca-8113 cells. ROS production was measured by reactive oxygen species assay. X-box binding protein-1 (XBP1) mRNA was analyzed by real-time-PCR, TRAF2, ASK1 and c-jun protein were investigated by western blot and immunocytochemistry respectively. The result indicated that ROS production, TRAF2, ASK1 and c-jun were elevated in MG-132 treated cells. Giving ROS scavenger N-acetyl-L-cysteine (NAC) largely prevented the effects of MG-132. Furthermore, treating with MG-132 lead to decreased XBP1 mRNA expression but could not completely block the expression of XBP1. Taken together, these findings provide the evidence that MG-132 induced ER stress lead to Tca-8113 cells apoptosis through ROS generation and TRAF2-ASK1-JNK signal pathway activation.
Collapse
Affiliation(s)
- Hai-ying Chen
- Oral Maxillofacial Head-Neck Key Laboratory of Medical Biology, and central laboratory of Liaocheng People's Hospital, Liaocheng, Shandong 252000, China
| | - Xiao-yan Ren
- Oral Maxillofacial Head-Neck Key Laboratory of Medical Biology, and central laboratory of Liaocheng People's Hospital, Liaocheng, Shandong 252000, China
| | - Wei-hua Wang
- Oral Maxillofacial Head-Neck Key Laboratory of Medical Biology, and central laboratory of Liaocheng People's Hospital, Liaocheng, Shandong 252000, China
| | - Ying-xin Zhang
- Oral Maxillofacial Head-Neck Key Laboratory of Medical Biology, and central laboratory of Liaocheng People's Hospital, Liaocheng, Shandong 252000, China
| | - Shuang-feng Chen
- Oral Maxillofacial Head-Neck Key Laboratory of Medical Biology, and central laboratory of Liaocheng People's Hospital, Liaocheng, Shandong 252000, China
| | - Bin Zhang
- Department of Oral and Maxillofacial Surgery, Liaocheng People's Hospital, Liaocheng, Shandong 252000, China.
| | - Le-xin Wang
- School of Biomedical Sciences, Charles Sturt University, Wagga Wagga, NSW 2650, Australia.
| |
Collapse
|
8
|
Liu W, Han B, Sun B, Gao Y, Huang Y, Hu M. Overexpression of Interleukin-18 Induces Growth Inhibition, Apoptosis and Gene Expression Changes in a Human Tongue Squamous Cell Carcinoma Cell Line. J Int Med Res 2012; 40:537-44. [PMID: 22613414 DOI: 10.1177/147323001204000215] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
OBJECTIVE: To investigate the role of interleukin-18 (IL-18) in regulating the growth of the human tongue squamous cell carcinoma cell line CRL-1623™. METHODS: The human IL18 gene was cloned and transfected into CRL-1623™ cells using the transfection vector pcDNA3.1(+). Investigations included analysis of cell viability, detection of apoptosis using annexin V—fluorescein isothiocyanate, assessment of caspase 3/7 activity and real-time reverse transcription—polymerase chain reaction to assess expression of the IL18, CCND1 (cyclin D1), CCNA1 (cyclin A1) and IFNG (interferon-γ) genes. RESULTS: Introduction of the IL18 gene inhibited cell proliferation at 24, 48 and 72 h after transfection compared with untransfected cells and cells transfected with blank pcDNA3.1(+) vector. Apoptotic cell numbers and caspase 3/7 activity were significantly enhanced by IL18 transfection. Levels of IL18 and IFNG mRNA were elevated and CCND1 mRNA was reduced after 48 h in IL18 transfected cells compared with wild-type cells. CONCLUSIONS: These findings suggest that IL-18 plays a role in the regulation of tongue squamous cell carcinoma and may represent a potential therapeutic target.
Collapse
Affiliation(s)
- W Liu
- Stomatology Medical College, Jilin University, Changchun, China
| | - B Han
- Stomatology Medical College, Jilin University, Changchun, China
| | - B Sun
- China-Japan Friendship Hospital, Jilin University, Changchun, China
| | - Y Gao
- Stomatology Medical College, Jilin University, Changchun, China
| | - Y Huang
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun, China
| | - M Hu
- Stomatology Medical College, Jilin University, Changchun, China
| |
Collapse
|