1
|
Palumbo V, Treglia M, Scimeca M, Servadei F, Giacobbi E, Bonfiglio R, Pallocci M, Passalacqua P, Del Duca F, Tittarelli R, Coppeta L, Schiaroli S, Cervelli G, Mauriello A, Marsella LT, Mauriello S. Cocaine-Induced Cardiac Alterations: Histological and Immunohistochemical Post-Mortem Analysis. Diagnostics (Basel) 2025; 15:999. [PMID: 40310391 PMCID: PMC12026069 DOI: 10.3390/diagnostics15080999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 03/04/2025] [Accepted: 04/01/2025] [Indexed: 05/02/2025] Open
Abstract
Background: Cocaine abuse represents a serious health issue. The cardiovascular system is one of the main sites on which cocaine elicits its toxicity, as indicated by deadly events mainly related to myocardial infarction. The main aim of this study was to characterize the histological and immunohistochemical alterations related to cocaine abuse in cardiac tissue. Methods: Cardiac tissue samples derived from cocaine-related (n = 30) and not-cocaine-related deaths (n = 30). Histomorphology evaluations and immunohistochemistry for inflammatory biomarkers (CD45 and CD3) have been performed on formalin-fixed, paraffin-embedded (FFPE) cardiac tissue samples. Results: A higher frequency of cardiac alterations, such as wavy fibers, interstitial edema, fibrosis and hemorrhagic extravasation, were found in the group of cocaine users compared to the control group. Moreover, immunohistochemical analysis showed higher levels of inflammatory cells infiltrate within the cocaine-related deaths group. Conclusions: These data could shed new light on the complex relationship between cocaine use and cardiac alterations. Specifically, our data support the evidence that cocaine abuse is related to cardiac inflammation. Therefore, the generation of an inflammatory state could promote functional and structural cardiac alterations and lead ultimately to myocardial infarction. This would explain the high frequency of acute myocardial infarction in cocaine users.
Collapse
Affiliation(s)
- Valeria Palumbo
- Department of Experimental Medicine, Tor Vergata Oncoscience Research, University of Rome “Tor Vergata”, 00133 Rome, Italy (F.S.)
| | - Michele Treglia
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, 00133 Rome, Italy
| | - Manuel Scimeca
- Department of Experimental Medicine, Tor Vergata Oncoscience Research, University of Rome “Tor Vergata”, 00133 Rome, Italy (F.S.)
| | - Francesca Servadei
- Department of Experimental Medicine, Tor Vergata Oncoscience Research, University of Rome “Tor Vergata”, 00133 Rome, Italy (F.S.)
| | - Erica Giacobbi
- Department of Experimental Medicine, Tor Vergata Oncoscience Research, University of Rome “Tor Vergata”, 00133 Rome, Italy (F.S.)
| | - Rita Bonfiglio
- Department of Experimental Medicine, Tor Vergata Oncoscience Research, University of Rome “Tor Vergata”, 00133 Rome, Italy (F.S.)
| | - Margherita Pallocci
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, 00133 Rome, Italy
- PhD School of Applied Medical-Surgical Sciences, University of Rome “Tor Vergata”, 00133 Rome, Italy
| | - Pierluigi Passalacqua
- Department of Public Health and Infectious Diseases, “Sapienza” University of Rome, 00185 Rome, Italy
| | - Fabio Del Duca
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Sapienza University of Rome, 00185 Rome, Italy
| | - Roberta Tittarelli
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, 00133 Rome, Italy
| | - Luca Coppeta
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, 00133 Rome, Italy
| | - Stefania Schiaroli
- Department of Experimental Medicine, Tor Vergata Oncoscience Research, University of Rome “Tor Vergata”, 00133 Rome, Italy (F.S.)
| | - Giulio Cervelli
- Department of Experimental Medicine, Tor Vergata Oncoscience Research, University of Rome “Tor Vergata”, 00133 Rome, Italy (F.S.)
| | - Alessandro Mauriello
- Department of Experimental Medicine, Tor Vergata Oncoscience Research, University of Rome “Tor Vergata”, 00133 Rome, Italy (F.S.)
| | - Luigi Tonino Marsella
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, 00133 Rome, Italy
| | - Silvestro Mauriello
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, 00133 Rome, Italy
| |
Collapse
|
2
|
Qin H, Li Z, Wu J, Liu X, Wang R, Xu J, Zhu X. Diclofenac Enhances the Response of BRAF Inhibitor to Melanoma Through ROS/p38/p53 Signaling. Clin Exp Pharmacol Physiol 2025; 52:e70022. [PMID: 39788129 DOI: 10.1111/1440-1681.70022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 12/18/2024] [Accepted: 12/24/2024] [Indexed: 01/12/2025]
Abstract
BRAF inhibitors (BRAFi) represent a cornerstone in melanoma therapy due to their high efficacy. However, the emergence of resistance causes a significant challenge to their clinical utility. This study aims to investigate the potential of diclofenac as a sensitizer for BRAFi therapy in melanoma and to elucidate its underlying mechanism. BRAFi-acquired resistant melanoma cell lines SK-MEL-5R and A375R were established and treated with diclofenac in combination with BRAFi PLX4032. Cell viability was assessed using the MTT assay, cell proliferation was determined by crystal violet staining, cell apoptosis was evaluated by flow cytometry, and intracellular ROS levels were measured using the DCFH-DA probe-labeled and flow cytometry. Mitochondrial membrane potential was assessed by JC-1 staining and flow cytometry, and protein expression levels were detected by western blotting. Our results demonstrated that diclofenac significantly augmented the cytotoxicity of PLX4032 and enhanced its ability to induce apoptosis in SK-MEL-5R and A375R cells. Diclofenac treatment led to the release of intracellular reactive oxygen species (ROS), consequently reducing transmembrane potential, promoting mitochondrial apoptosis, and activating the ROS downstream p38/p53 signaling pathway. Pretreatment with N-acetylcysteine significantly reversed the sensitizing effect of diclofenac on PLX4032 in SK-MEL-5R cells. These findings suggested that diclofenac sensitized BRAFi-resistant melanoma cells to BRAFi by increasing ROS release and activating p38/p53 signaling pathway. Diclofenac might serve as a promising adjunct therapy to overcome BRAFi resistance in melanoma treatment.
Collapse
Affiliation(s)
- Haihong Qin
- Department of Dermatology, Fudan University Huashan Hospital, Shanghai, China
| | - Zheng Li
- Department of Dermatology, Fudan University Huashan Hospital, Shanghai, China
| | - Jinfeng Wu
- Department of Dermatology, Fudan University Huashan Hospital, Shanghai, China
| | - Xiao Liu
- Department of Dermatology, Fudan University Huashan Hospital, Shanghai, China
| | - Ruilong Wang
- Department of Dermatology, Fudan University Huashan Hospital, Shanghai, China
| | - Jinhua Xu
- Department of Dermatology, Fudan University Huashan Hospital, Shanghai, China
| | - Xiaohua Zhu
- Department of Dermatology, Fudan University Huashan Hospital, Shanghai, China
| |
Collapse
|
3
|
Aloke C, Onisuru OO, Achilonu I. Glutathione S-transferase: A versatile and dynamic enzyme. Biochem Biophys Res Commun 2024; 734:150774. [PMID: 39366175 DOI: 10.1016/j.bbrc.2024.150774] [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: 07/20/2024] [Revised: 09/19/2024] [Accepted: 09/30/2024] [Indexed: 10/06/2024]
Abstract
The dynamic and versatile group of enzymes referred to as glutathione S-transferases (GSTs) play diverse roles in cellular detoxification, safeguarding hosts from oxidative damage, and performing various other functions. This review explores different classes of GST, existence of polymorphisms in GST, functions of GST and utilizations of GST inhibitors in treatment of human diseases. The study indicates that the cytosolic GSTs, mitochondrial GSTs, microsomal GSTs, and bacterial proteins that provide resistance to Fosfomycin are the major classes. Given a GST, variation in its expression and function among individuals is due to the presence of polymorphic alleles that encode it. Genetic polymorphism might result in the modification of GST activity, thereby increasing individuals' vulnerability to harmful chemical compounds. GSTs have been demonstrated to play a regulatory function in cellular signalling pathways through kinases, S-Glutathionylation, and in detoxification processes. Various applications of bacterial GSTs and their potential roles in plants were examined. Targeting GSTs, especially GSTP1-1, is considered a potential therapeutic strategy for treating cancer and diseases linked to abnormal cell proliferation. Their role in cancer cell growth, differentiation, and resistance to anticancer agents makes them promising targets for drug development, offering prospects for the future.
Collapse
Affiliation(s)
- Chinyere Aloke
- Protein Structure-Function and Research Unit, School of Molecular and Cell Biology, Faculty of Science, University of the Witwatersrand, Braamfontein, Johannesburg, 2050, South Africa; Department of Medical Biochemistry, Alex Ekwueme Federal University Ndufu-Alike, Ebonyi State, Nigeria.
| | - Olalekan Olugbenga Onisuru
- Protein Structure-Function and Research Unit, School of Molecular and Cell Biology, Faculty of Science, University of the Witwatersrand, Braamfontein, Johannesburg, 2050, South Africa
| | - Ikechukwu Achilonu
- Protein Structure-Function and Research Unit, School of Molecular and Cell Biology, Faculty of Science, University of the Witwatersrand, Braamfontein, Johannesburg, 2050, South Africa
| |
Collapse
|
4
|
Lv N, Huang C, Huang H, Dong Z, Chen X, Lu C, Zhang Y. Overexpression of Glutathione S-Transferases in Human Diseases: Drug Targets and Therapeutic Implications. Antioxidants (Basel) 2023; 12:1970. [PMID: 38001822 PMCID: PMC10668987 DOI: 10.3390/antiox12111970] [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: 09/25/2023] [Revised: 10/25/2023] [Accepted: 11/03/2023] [Indexed: 11/26/2023] Open
Abstract
Glutathione S-transferases (GSTs) are a major class of phase II metabolic enzymes. Besides their essential role in detoxification, GSTs also exert diverse biological activities in the occurrence and development of various diseases. In the past few decades, much research interest has been paid to exploring the mechanisms of GST overexpression in tumor drug resistance. Correspondingly, many GST inhibitors have been developed and applied, solely or in combination with chemotherapeutic drugs, for the treatment of multi-drug resistant tumors. Moreover, novel roles of GSTs in other diseases, such as pulmonary fibrosis and neurodegenerative diseases, have been recognized in recent years, although the exact regulatory mechanisms remain to be elucidated. This review, firstly summarizes the roles of GSTs and their overexpression in the above-mentioned diseases with emphasis on the modulation of cell signaling pathways and protein functions. Secondly, specific GST inhibitors currently in pre-clinical development and in clinical stages are inventoried. Lastly, applications of GST inhibitors in targeting cell signaling pathways and intracellular biological processes are discussed, and the potential for disease treatment is prospected. Taken together, this review is expected to provide new insights into the interconnection between GST overexpression and human diseases, which may assist future drug discovery targeting GSTs.
Collapse
Affiliation(s)
- Ning Lv
- Clinical Pharmacology Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; (N.L.); (H.H.)
| | - Chunyan Huang
- Clinical Pharmacology Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; (N.L.); (H.H.)
| | - Haoyan Huang
- Clinical Pharmacology Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; (N.L.); (H.H.)
| | - Zhiqiang Dong
- Department of Pharmacy, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing 211100, China;
| | - Xijing Chen
- Clinical Pharmacology Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; (N.L.); (H.H.)
| | - Chengcan Lu
- Department of Pharmacy, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing 211100, China;
- Jiangning Clinical Medical College, Jiangsu University, Nanjing 211100, China
| | - Yongjie Zhang
- Clinical Pharmacology Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; (N.L.); (H.H.)
| |
Collapse
|
5
|
Sorbi C, Belluti S, Atene CG, Marocchi F, Linciano P, Roy N, Paradiso E, Casarini L, Ronsisvalle S, Zanocco-Marani T, Brasili L, Lanfrancone L, Imbriano C, Di Rocco G, Franchini S. BS148 Reduces the Aggressiveness of Metastatic Melanoma via Sigma-2 Receptor Targeting. Int J Mol Sci 2023; 24:ijms24119684. [PMID: 37298633 DOI: 10.3390/ijms24119684] [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: 03/21/2023] [Revised: 05/18/2023] [Accepted: 05/30/2023] [Indexed: 06/12/2023] Open
Abstract
The management of advanced-stage melanoma is clinically challenging, mainly because of its resistance to the currently available therapies. Therefore, it is important to develop alternative therapeutic strategies. The sigma-2 receptor (S2R) is overexpressed in proliferating tumor cells and represents a promising vulnerability to target. Indeed, we have recently identified a potent S2R modulator (BS148) that is effective in melanoma. To elucidate its mechanism of action, we designed and synthesized a BS148 fluorescent probe that enters SK-MEL-2 melanoma cells as assessed using confocal microscopy analysis. We show that S2R knockdown significantly reduces the anti-proliferative effect induced by BS148 administration, indicating the engagement of S2R in BS148-mediated cytotoxicity. Interestingly, BS148 treatment showed similar molecular effects to S2R RNA interference-mediated knockdown. We demonstrate that BS148 administration activates the endoplasmic reticulum stress response through the upregulation of protein kinase R-like ER kinase (PERK), activating transcription factor 4 (ATF4) genes, and C/EBP homologous protein (CHOP). Furthermore, we show that BS148 treatment downregulates genes related to the cholesterol pathway and activates the MAPK signaling pathway. Finally, we translate our results into patient-derived xenograft (PDX) cells, proving that BS148 treatment reduces melanoma cell viability and migration. These results demonstrate that BS148 is able to inhibit metastatic melanoma cell proliferation and migration through its interaction with the S2R and confirm its role as a promising target to treat cancer.
Collapse
Affiliation(s)
- Claudia Sorbi
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Silvia Belluti
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Claudio Giacinto Atene
- Hematology Section, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Federica Marocchi
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, 20139 Milan, Italy
| | - Pasquale Linciano
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy
| | - Neena Roy
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Ospedale di Baggiovara, 41126 Modena, Italy
| | - Elia Paradiso
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Ospedale di Baggiovara, 41126 Modena, Italy
| | - Livio Casarini
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Ospedale di Baggiovara, 41126 Modena, Italy
- Center for Genomic Research, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Simone Ronsisvalle
- Department of Drug and Health Sciences, University of Catania, 95125 Catania, Italy
| | - Tommaso Zanocco-Marani
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Livio Brasili
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Luisa Lanfrancone
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, 20139 Milan, Italy
| | - Carol Imbriano
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Giulia Di Rocco
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Silvia Franchini
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| |
Collapse
|
6
|
Sha H, Zou R, Lu Y, Gan Y, Ma R, Feng J, Chen D. NBDHEX re-sensitizes adriamycin-resistant breast cancer by inhibiting glutathione S-transferase pi. Cancer Med 2023; 12:5833-5845. [PMID: 36266920 PMCID: PMC10028113 DOI: 10.1002/cam4.5370] [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: 12/15/2021] [Revised: 08/26/2022] [Accepted: 10/08/2022] [Indexed: 11/06/2022] Open
Abstract
PURPOSE Adriamycin is a novel chemotherapeutic agent of great benefit for treating breast cancer. However, adriamycin -resistance remains a major obstacle. The vital Glutathione transferase P1 (GSTPi) inhibitor 6-(7-nitro-2,1,3-benzoxadiazol-4-ylthio) hexanol (NBDHEX) has recently shown antitumor activity in various cancers. In this study, we analyzed the effect of NBDHEX and adriamycin combination against breast cancer in vitro and in vivo. METHODS CCK-8 assay was performed to test cell viability. The location and expression level of GSTpi was determined by immunofluorescence and Western blot in cells and immunohistochemistry staining in tissues. The enzyme activity test was applied to detect the effect of NBDHEX on the activity of GSTpi. The apoptosis related proteins' expression was tested using Western blot. The phosphorylation sites of GSTpi were detected by mass spectrometry. Antitumor effects of single treatment or co-administration of adriamycin and NBDHEX were evaluated in nude mice. RESULTS NBDHEX treatment inhibited GSTpi enzyme activity and co-administration of adriamycin and NBDHEX promoted apoptosis of adriamycin-resistance breast cancer cell. Moreover, drug combination of NBDHEX and adriamycin significantly enhanced tumor growth inhibition compared with single agent. CONCLUSION NBDHEX serves as a good candidate for combination with adriamycin, offering new insights for breast cancer treatment.
Collapse
Affiliation(s)
- Huanhuan Sha
- Department of ChemotherapyJiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer ResearchNanjingJiangsuChina
| | - Renrui Zou
- The Fourth Clinical School of The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer ResearchNanjingJiangsuChina
| | - Ya Lu
- The Fourth Clinical School of The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer ResearchNanjingJiangsuChina
| | - Yujie Gan
- The Fourth Clinical School of The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer ResearchNanjingJiangsuChina
| | - Rong Ma
- Research Center of Clinical Oncology, Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer ResearchNanjingJiangsuChina
| | - Jifeng Feng
- The Fourth Clinical School of The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer ResearchNanjingJiangsuChina
| | - Dan Chen
- Research Center of Clinical Oncology, Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer ResearchNanjingJiangsuChina
| |
Collapse
|
7
|
Identification of Dihydrolipoamide Dehydrogenase as Potential Target of Vemurafenib-Resistant Melanoma Cells. Molecules 2022; 27:molecules27227800. [PMID: 36431901 PMCID: PMC9698468 DOI: 10.3390/molecules27227800] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/07/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Despite recent improvements in therapy, the five-year survival rate for patients with advanced melanoma is poor, mainly due to the development of drug resistance. The aim of the present study was to investigate the mechanisms underlying this phenomenon, applying proteomics and structural approaches to models of melanoma cells. METHODS Sublines from two human (A375 and SK-MEL-28) cells with acquired vemurafenib resistance were established, and their proteomic profiles when exposed to denaturation were identified through LC-MS/MS analysis. The pathways derived from bioinformatics analyses were validated by in silico and functional studies. RESULTS The proteomic profiles of resistant melanoma cells were compared to parental counterparts by taking into account protein folding/unfolding behaviors. Several proteins were found to be involved, with dihydrolipoamide dehydrogenase (DLD) being the only one similarly affected by denaturation in all resistant cell sublines compared to parental ones. DLD expression was observed to be increased in resistant cells by Western blot analysis. Protein modeling analyses of DLD's catalytic site coupled to in vitro assays with CPI-613, a specific DLD inhibitor, highlighted the role of DLD enzymatic functions in the molecular mechanisms of BRAFi resistance. CONCLUSIONS Our proteomic and structural investigations on resistant sublines indicate that DLD may represent a novel and potent target for overcoming vemurafenib resistance in melanoma cells.
Collapse
|
8
|
NIITSU Y, SATO Y, TAKAYAMA T. Implications of glutathione-S transferase P1 in MAPK signaling as a CRAF chaperone: In memory of Dr. Irving Listowsky. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2022; 98:72-86. [PMID: 35153270 PMCID: PMC8890996 DOI: 10.2183/pjab.98.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 12/14/2021] [Indexed: 06/14/2023]
Abstract
Glutathione-S transferase P1 (GSTP1) is one of the glutathione-S transferase isozymes that belong to a family of phase II metabolic isozymes. The unique feature of GSTP1 compared with other GST isozymes is its relatively high expression in malignant tissues. Thus, clinically, GSTP1 serves as a tumor marker and as a refractory factor against certain types of anticancer drugs through its primary function as a detoxifying enzyme. Additionally, recent studies have identified a chaperone activity of GSTP1 involved in the regulation the function of various intracellular proteins, including factors of the growth signaling pathway. In this review, we will first describe the function of GSTP1 and then extend the details onto its role in the mitogen-activated protein kinase signal pathway, referring to the results of our recent study that proposed a novel autocrine signal loop formed by the CRAF/GSTP1 complex in mutated KRAS and BRAF cancers. Finally, the possibilities of new therapeutic approaches for these cancers by targeting this complex will be discussed.
Collapse
Affiliation(s)
- Yoshiro NIITSU
- Oncology Section, Center of Advanced Medicine, Shonan Kamakura Innovation Park, Shonan Kamakura General Hospital, Kamakura, Kanagawa, Japan
- Sapporo Medical University, Sapporo, Hokkaido, Japan
| | - Yasushi SATO
- Department of Community Medicine for Gastroenterology and Oncology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Tetsuji TAKAYAMA
- Department of Community Medicine for Gastroenterology and Oncology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| |
Collapse
|
9
|
Fiorentino F, De Angelis M, Menna M, Rovere A, Caccuri AM, D'Acunzo F, Palamara AT, Nencioni L, Rotili D, Mai A. Anti-influenza A virus activity and structure-activity relationship of a series of nitrobenzoxadiazole derivatives. J Enzyme Inhib Med Chem 2021; 36:2128-2138. [PMID: 34583607 PMCID: PMC8480593 DOI: 10.1080/14756366.2021.1982932] [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] [Indexed: 11/08/2022] Open
Abstract
Influenza viruses represent a major threat to human health and are responsible for seasonal epidemics, along with pandemics. Currently, few therapeutic options are available, with most drugs being at risk of the insurgence of resistant strains. Hence, novel approaches targeting less explored pathways are urgently needed. In this work, we assayed a library of nitrobenzoxadiazole derivatives against the influenza virus A/Puerto Rico/8/34 H1N1 (PR8) strain. We identified three promising 4-thioether substituted nitrobenzoxadiazoles (12, 17, and 25) that were able to inhibit viral replication at low micromolar concentrations in two different infected cell lines using a haemagglutination assay. We further assessed these molecules using an In-Cell Western assay, which confirmed their potency in the low micromolar range. Among the three molecules, 12 and 25 displayed the most favourable profile of activity and selectivity and were selected as hit compounds for future optimisation studies.
Collapse
Affiliation(s)
- Francesco Fiorentino
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Rome, Italy
| | - Marta De Angelis
- Department of Public Health and Infectious Diseases, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - Martina Menna
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Rome, Italy
| | - Annarita Rovere
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Rome, Italy
| | - Anna Maria Caccuri
- Department of Chemical Sciences and Technologies, University of Rome Tor Vergata, Rome, Italy
| | - Francesca D'Acunzo
- CNR, Istituto di Metodologie Chimiche, Sezione Meccanismi di Reazione, Sapienza University of Rome, Rome, Italy
| | - Anna Teresa Palamara
- Department of Public Health and Infectious Diseases, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy.,Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Lucia Nencioni
- Department of Public Health and Infectious Diseases, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - Dante Rotili
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Rome, Italy
| | - Antonello Mai
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
10
|
Abstract
Incorporation of heterocycles into drug molecules can enhance physical properties and biological activity. A variety of heterocyclic groups is available to medicinal chemists, many of which have been reviewed in detail elsewhere. Oxadiazoles are a class of heterocycle containing one oxygen and two nitrogen atoms, available in three isomeric forms. While the 1,2,4- and 1,3,4-oxadiazoles have seen widespread application in medicinal chemistry, 1,2,5-oxadiazoles (furazans) are less common. This Review provides a summary of the application of furazan-containing molecules in medicinal chemistry and drug development programs from analysis of both patent and academic literature. Emphasis is placed on programs that reached clinical or preclinical stages of development. The examples provided herein describe the pharmacology and biological activity of furazan derivatives with comparative data provided where possible for other heterocyclic groups and pharmacophores commonly used in medicinal chemistry.
Collapse
Affiliation(s)
| | | | - Donald F Weaver
- Department of Fundamental Neurobiology, Krembil Research Institute, Toronto, Ontario M5T 0S8, Canada.,Department of Chemistry, University of Toronto, Toronto, Ontario M5S 3H6, Canada.,Department of Medicine, University of Toronto, Toronto, Ontario M5S 3H2, Canada
| | - Mark A Reed
- Treventis Corporation, Toronto, Ontario M5T 0S8, Canada.,Department of Fundamental Neurobiology, Krembil Research Institute, Toronto, Ontario M5T 0S8, Canada
| |
Collapse
|
11
|
Proietti I, Skroza N, Bernardini N, Tolino E, Balduzzi V, Marchesiello A, Michelini S, Volpe S, Mambrin A, Mangino G, Romeo G, Maddalena P, Rees C, Potenza C. Mechanisms of Acquired BRAF Inhibitor Resistance in Melanoma: A Systematic Review. Cancers (Basel) 2020; 12:E2801. [PMID: 33003483 PMCID: PMC7600801 DOI: 10.3390/cancers12102801] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 09/21/2020] [Accepted: 09/25/2020] [Indexed: 12/18/2022] Open
Abstract
This systematic review investigated the literature on acquired v-raf murine sarcoma viral oncogene homolog B1 (BRAF) inhibitor resistance in patients with melanoma. We searched MEDLINE for articles on BRAF inhibitor resistance in patients with melanoma published since January 2010 in the following areas: (1) genetic basis of resistance; (2) epigenetic and transcriptomic mechanisms; (3) influence of the immune system on resistance development; and (4) combination therapy to overcome resistance. Common resistance mutations in melanoma are BRAF splice variants, BRAF amplification, neuroblastoma RAS viral oncogene homolog (NRAS) mutations and mitogen-activated protein kinase kinase 1/2 (MEK1/2) mutations. Genetic and epigenetic changes reactivate previously blocked mitogen-activated protein kinase (MAPK) pathways, activate alternative signaling pathways, and cause epithelial-to-mesenchymal transition. Once BRAF inhibitor resistance develops, the tumor microenvironment reverts to a low immunogenic state secondary to the induction of programmed cell death ligand-1. Combining a BRAF inhibitor with a MEK inhibitor delays resistance development and increases duration of response. Multiple other combinations based on known mechanisms of resistance are being investigated. BRAF inhibitor-resistant cells develop a range of 'escape routes', so multiple different treatment targets will probably be required to overcome resistance. In the future, it may be possible to personalize combination therapy towards the specific resistance pathway in individual patients.
Collapse
Affiliation(s)
- Ilaria Proietti
- Dermatology Unit “Daniele Innocenzi”, Department of Medical-Surgical Sciences and Bio-Technologies, Sapienza University of Rome, Fiorini Hospital, Polo Pontino, 04019 Terracina, Italy; (N.S.); (N.B.); (E.T.); (V.B.); (A.M.); (S.M.); (S.V.); (A.M.); (P.M.); (C.P.)
| | - Nevena Skroza
- Dermatology Unit “Daniele Innocenzi”, Department of Medical-Surgical Sciences and Bio-Technologies, Sapienza University of Rome, Fiorini Hospital, Polo Pontino, 04019 Terracina, Italy; (N.S.); (N.B.); (E.T.); (V.B.); (A.M.); (S.M.); (S.V.); (A.M.); (P.M.); (C.P.)
| | - Nicoletta Bernardini
- Dermatology Unit “Daniele Innocenzi”, Department of Medical-Surgical Sciences and Bio-Technologies, Sapienza University of Rome, Fiorini Hospital, Polo Pontino, 04019 Terracina, Italy; (N.S.); (N.B.); (E.T.); (V.B.); (A.M.); (S.M.); (S.V.); (A.M.); (P.M.); (C.P.)
| | - Ersilia Tolino
- Dermatology Unit “Daniele Innocenzi”, Department of Medical-Surgical Sciences and Bio-Technologies, Sapienza University of Rome, Fiorini Hospital, Polo Pontino, 04019 Terracina, Italy; (N.S.); (N.B.); (E.T.); (V.B.); (A.M.); (S.M.); (S.V.); (A.M.); (P.M.); (C.P.)
| | - Veronica Balduzzi
- Dermatology Unit “Daniele Innocenzi”, Department of Medical-Surgical Sciences and Bio-Technologies, Sapienza University of Rome, Fiorini Hospital, Polo Pontino, 04019 Terracina, Italy; (N.S.); (N.B.); (E.T.); (V.B.); (A.M.); (S.M.); (S.V.); (A.M.); (P.M.); (C.P.)
| | - Anna Marchesiello
- Dermatology Unit “Daniele Innocenzi”, Department of Medical-Surgical Sciences and Bio-Technologies, Sapienza University of Rome, Fiorini Hospital, Polo Pontino, 04019 Terracina, Italy; (N.S.); (N.B.); (E.T.); (V.B.); (A.M.); (S.M.); (S.V.); (A.M.); (P.M.); (C.P.)
| | - Simone Michelini
- Dermatology Unit “Daniele Innocenzi”, Department of Medical-Surgical Sciences and Bio-Technologies, Sapienza University of Rome, Fiorini Hospital, Polo Pontino, 04019 Terracina, Italy; (N.S.); (N.B.); (E.T.); (V.B.); (A.M.); (S.M.); (S.V.); (A.M.); (P.M.); (C.P.)
| | - Salvatore Volpe
- Dermatology Unit “Daniele Innocenzi”, Department of Medical-Surgical Sciences and Bio-Technologies, Sapienza University of Rome, Fiorini Hospital, Polo Pontino, 04019 Terracina, Italy; (N.S.); (N.B.); (E.T.); (V.B.); (A.M.); (S.M.); (S.V.); (A.M.); (P.M.); (C.P.)
| | - Alessandra Mambrin
- Dermatology Unit “Daniele Innocenzi”, Department of Medical-Surgical Sciences and Bio-Technologies, Sapienza University of Rome, Fiorini Hospital, Polo Pontino, 04019 Terracina, Italy; (N.S.); (N.B.); (E.T.); (V.B.); (A.M.); (S.M.); (S.V.); (A.M.); (P.M.); (C.P.)
| | - Giorgio Mangino
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 00185 Rome, Italy; (G.M.); (G.R.)
| | - Giovanna Romeo
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 00185 Rome, Italy; (G.M.); (G.R.)
- Department of Infectious, Parasitic and Immune-Mediated Diseases, Istituto Superiore di Sanità, 00185 Rome, Italy
- Institute of Molecular Biology and Pathology, Consiglio Nazionale delle Ricerche, 00185 Rome, Italy
| | - Patrizia Maddalena
- Dermatology Unit “Daniele Innocenzi”, Department of Medical-Surgical Sciences and Bio-Technologies, Sapienza University of Rome, Fiorini Hospital, Polo Pontino, 04019 Terracina, Italy; (N.S.); (N.B.); (E.T.); (V.B.); (A.M.); (S.M.); (S.V.); (A.M.); (P.M.); (C.P.)
| | | | - Concetta Potenza
- Dermatology Unit “Daniele Innocenzi”, Department of Medical-Surgical Sciences and Bio-Technologies, Sapienza University of Rome, Fiorini Hospital, Polo Pontino, 04019 Terracina, Italy; (N.S.); (N.B.); (E.T.); (V.B.); (A.M.); (S.M.); (S.V.); (A.M.); (P.M.); (C.P.)
| |
Collapse
|
12
|
Di Paolo V, Fulci C, Rotili D, De Luca A, Tomassi S, Serra M, Scimeca M, Geroni C, Quintieri L, Caccuri AM. Characterization of water-soluble esters of nitrobenzoxadiazole-based GSTP1-1 inhibitors for cancer treatment. Biochem Pharmacol 2020; 178:114060. [DOI: 10.1016/j.bcp.2020.114060] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 05/25/2020] [Indexed: 12/11/2022]
|
13
|
Huebner K, Procházka J, Monteiro AC, Mahadevan V, Schneider-Stock R. The activating transcription factor 2: an influencer of cancer progression. Mutagenesis 2020; 34:375-389. [PMID: 31799611 PMCID: PMC6923166 DOI: 10.1093/mutage/gez041] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 11/18/2019] [Indexed: 12/26/2022] Open
Abstract
In contrast to the continuous increase in survival rates for many cancer entities, colorectal cancer (CRC) and pancreatic cancer are predicted to be ranked among the top 3 cancer-related deaths in the European Union by 2025. Especially, fighting metastasis still constitutes an obstacle to be overcome in CRC and pancreatic cancer. As described by Fearon and Vogelstein, the development of CRC is based on sequential mutations leading to the activation of proto-oncogenes and the inactivation of tumour suppressor genes. In pancreatic cancer, genetic alterations also attribute to tumour development and progression. Recent findings have identified new potentially important transcription factors in CRC, among those the activating transcription factor 2 (ATF2). ATF2 is a basic leucine zipper protein and is involved in physiological and developmental processes, as well as in tumorigenesis. The mutation burden of ATF2 in CRC and pancreatic cancer is rather negligible; however, previous studies in other tumours indicated that ATF2 expression level and subcellular localisation impact tumour progression and patient prognosis. In a tissue- and stimulus-dependent manner, ATF2 is activated by upstream kinases, dimerises and induces target gene expression. Dependent on its dimerisation partner, ATF2 homodimers or heterodimers bind to cAMP-response elements or activator protein 1 consensus motifs. Pioneering work has been performed in melanoma in which the dual role of ATF2 is best understood. Even though there is increasing interest in ATF2 recently, only little is known about its involvement in CRC and pancreatic cancer. In this review, we summarise the current understanding of the underestimated ‘cancer gene chameleon’ ATF2 in apoptosis, epithelial-to-mesenchymal transition and microRNA regulation and highlight its functions in CRC and pancreatic cancer. We further provide a novel ATF2 3D structure with key phosphorylation sites and an updated overview of all so-far available mouse models to study ATF2 in vivo.
Collapse
Affiliation(s)
- Kerstin Huebner
- Experimental Tumorpathology, Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Jan Procházka
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the ASCR, Prague, Czech Republic
| | - Ana C Monteiro
- Experimental Tumorpathology, Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Vijayalakshmi Mahadevan
- Institute of Bioinformatics and Applied Biotechnology, Biotech Park, Electronic City Phase I, Bangalore, India
| | - Regine Schneider-Stock
- Experimental Tumorpathology, Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
14
|
Li Z, Liu X, Li M, Chai J, He S, Wu J, Xu J. Juglone potentiates BRAF inhibitor‑induced apoptosis in melanoma through reactive oxygen species and the p38‑p53 pathway. Mol Med Rep 2020; 22:566-574. [PMID: 32377702 DOI: 10.3892/mmr.2020.11095] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 03/02/2020] [Indexed: 11/06/2022] Open
Abstract
BRAF inhibitors are some of the most effective drugs against melanoma; however, their clinical application is largely limited by drug resistance. Juglone, isolated from walnut trees, has demonstrated anti‑tumour activity. In the present study, it was investigated whether juglone could enhance the responses to a BRAF inhibitor in melanoma cells (A375R and SK‑MEL‑5R) with an acquired resistance. These cells were treated with juglone alone, BRAF inhibitor (PLX4032) alone, or juglone combined with PLX4032. It was demonstrated that the combination of juglone and PLX4032 had synergistic effects on BRAF inhibitor‑resistant melanoma cells. Juglone potentiated PLX4032‑induced cytotoxicity and mitochondrial apoptosis in both A375R and SK‑MEL‑5R cells, which was accompanied by a decline in mitochondrial membrane potential and a decrease in Bcl‑2/Bax ratio. Moreover, juglone combined with PLX4032 markedly increased the intracellular level of reactive oxygen species (ROS) and activated p38 and p53, as compared with juglone alone or PLX4032 alone. Pre‑treatment with N‑acetyl‑L‑cysteine, a ROS scavenger, completely reversed the cytotoxicity induced by juglone combined with PLX4032. In conclusion, juglone potentiated BRAF inhibitor‑induced apoptosis in resistant melanoma cells, and these effects occurred partially through ROS and the p38‑p53 pathway, suggesting the potential of juglone as a sensitizer to BRAF inhibitors in the treatment of melanoma.
Collapse
Affiliation(s)
- Zheng Li
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Xiao Liu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Ming Li
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Jingxiu Chai
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Shan He
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Jinfeng Wu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Jinhua Xu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| |
Collapse
|
15
|
Sciarretta F, Fulci C, Palumbo C, Rotili D, Tentori L, Graziani G, Caccuri AM. Effects of Glutathione Transferase-Targeting Nitrobenzoxadiazole Compounds in Relation to PD-L1 Status in Human Melanoma Cells. Chemotherapy 2019; 64:138-145. [PMID: 31639786 DOI: 10.1159/000503339] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 09/12/2019] [Indexed: 11/19/2022]
Abstract
BACKGROUND PD-L1 is a membrane protein with inhibitory effects on immune responses, whose expression has been correlated with high aggressiveness and the propensity of melanoma to metastasize. The nitrobenzoxadiazole (NBD) NBDHEX and its analog MC3181 are endowed with strong antitumor activity towards melanoma and a significant ability to reduce its adhesion and invasiveness. Therefore, we investigated whether PD-L1 status could affect cell sensitivity to the cytotoxic effects of NBDs. We then evaluated the effects of NBDHEX on PD-L1 expression and autophagy in melanoma cells. We used the BRAF-mutated A375 melanoma cell line and an A375 variant population enriched for PD-L1+ cells as a model. The cytotoxic effects of NBDs were evaluated in comparison to those of the BRAF inhibitor vemurafenib and the autophagy inhibitor chloroquine. METHODS The effect of NBDHEX on autophagy was determined by measuring LC3-II and p62 protein levels by Western blot. The cytotoxic activity of the compounds was evaluated by sulforhodamine B assay. PD-L1 expression and plasma membrane localization were analyzed by FACS and Western blot analysis. RESULTS NBDHEX behaves as a late-autophagy inhibitor in A375 melanoma cells, as previously found in other tumor cell lines. NBDHEX and MC3181 showed strong and comparable cytotoxic activity in both parental and PD-L1+ A375 cells, with IC50 values in the sub-micromolar range. Conversely, cells sorted for high PD-L1 expression had lower sensitivity to both the BRAF inhibitor vemurafenib and the autophagy inhibitor chloroquine. NBDHEX treatment did not change the total expression and cell surface localization of PD-L1 in both parental and PD-L1+ A375 cells. CONCLUSIONS Our data suggest that NBDs may represent a promising treatment strategy for melanoma with elevated PD-L1 expression.
Collapse
Affiliation(s)
- Francesca Sciarretta
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Chiara Fulci
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Camilla Palumbo
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Dante Rotili
- Department of Drug Chemistry and Technologies, University of "Sapienza,", Rome, Italy
| | - Lucio Tentori
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Grazia Graziani
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Anna Maria Caccuri
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy, .,The NAST Centre for Nanoscience and Nanotechnology and Innovative Instrumentation, University of Rome Tor Vergata, Rome, Italy,
| |
Collapse
|
16
|
Sciarretta F, Fulci C, Palumbo C, Aquilano K, Pastore A, Iorio E, Lettieri-Barbato D, Cicconi R, Minutolo A, Parravano M, Gilardi M, Varano M, Caccuri AM. Glutathione transferase P silencing promotes neuronal differentiation of retinal R28 cells. J Cell Physiol 2019; 234:15885-15897. [PMID: 30741416 DOI: 10.1002/jcp.28246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 01/14/2019] [Accepted: 01/15/2019] [Indexed: 01/24/2023]
Abstract
Glutathione transferases (GSTs) play an important role in retinal pathophysiology. Within this family, the GSTP isoform is known as an endogenous regulator of cell survival and proliferation pathways and of cellular responses to oxidative stress. In the present study we silenced GSTP in R28 cells, a retinal precursor cell line with markers of both glial and neuronal origin, and obtained stable clones which were viable and, unexpectedly, characterized by a more neuronal phenotype. The degree of neuronal differentiation was inversely correlated with GSTP residual expression levels. The clone with the lowest expression of GSTP showed metabolic reprogramming, a more favorable redox status and, despite its neuronal phenotype, a sensitivity to glutamate and 4-hydroxynonenal toxicity comparable to that of control cells. Altogether, our evidence shows that near full depletion of GSTP in retinal precursor cells, triggers neuronal differentiation and prosurvival metabolic changes.
Collapse
Affiliation(s)
- Francesca Sciarretta
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Chiara Fulci
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Camilla Palumbo
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Katia Aquilano
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Anna Pastore
- Laboratory of Molecular Genetics and Functional Genomics, Division of Genetic and Rare Disease, Children's Hospital and Research Institute Bambino Gesù, Rome, Italy
| | - Egidio Iorio
- Core Facilities, High Resolution NMR Unit, Istituto Superiore di Sanità, Rome, Italy
| | | | - Rosella Cicconi
- Interdepartmental Service Centre - Station for Animal Technology (STA), University of Rome Tor Vergata, Rome, Italy
| | | | | | - Marta Gilardi
- Ophthalmology, IRCCS-G.B. Bietti Foundation, Rome, Italy
| | - Monica Varano
- Ophthalmology, IRCCS-G.B. Bietti Foundation, Rome, Italy
| | - Anna Maria Caccuri
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy.,Interdepartmental Centre for Nanosciences, Nanotechnologies, Innovative Instrumentation (NAST), University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
17
|
Di Paolo V, Fulci C, Rotili D, Sciarretta F, Lucidi A, Morozzo Della Rocca B, De Luca A, Rosato A, Quintieri L, Caccuri AM. Synthesis and characterisation of a new benzamide-containing nitrobenzoxadiazole as a GSTP1-1 inhibitor endowed with high stability to metabolic hydrolysis. J Enzyme Inhib Med Chem 2019; 34:1131-1139. [PMID: 31169043 PMCID: PMC6566875 DOI: 10.1080/14756366.2019.1617287] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The antitumor agent 6-((7-nitrobenzo[c][1,2,5]oxadiazol-4-yl)thio)hexan-1-ol (1) is a potent inhibitor of GSTP1-1, a glutathione S-transferase capable of inhibiting apoptosis by binding to JNK1 and TRAF2. We recently demonstrated that, unlike its parent compound, the benzoyl ester of 1 (compound 3) exhibits negligible reactivity towards GSH, and has a different mode of interaction with GSTP1-1. Unfortunately, 3 is susceptible to rapid metabolic hydrolysis. In an effort to improve the metabolic stability of 3, its ester group has been replaced by an amide, leading to N-(6-((7-nitrobenzo[c][1,2,5]oxadiazol-4-yl)thio)hexyl)benzamide (4). Unlike 3, compound 4 was stable to human liver microsomal carboxylesterases, but retained the ability to disrupt the interaction between GSTP1-1 and TRAF2 regardless of GSH levels. Moreover, 4 exhibited both a higher stability in the presence of GSH and a greater cytotoxicity towards cultured A375 melanoma cells, in comparison with 1 and its analog 2. These findings suggest that 4 deserves further preclinical testing.
Collapse
Affiliation(s)
- Veronica Di Paolo
- a Department of Pharmaceutical and Pharmacological Sciences , University of Padova , Padova , Italy
| | - Chiara Fulci
- b Department of Experimental Medicine , "Tor Vergata" University of Rome , Rome , Italy
| | - Dante Rotili
- c Department of Drug Chemistry and Technologies , "Sapienza" University of Rome , Rome , Italy
| | - Francesca Sciarretta
- b Department of Experimental Medicine , "Tor Vergata" University of Rome , Rome , Italy
| | - Alessia Lucidi
- c Department of Drug Chemistry and Technologies , "Sapienza" University of Rome , Rome , Italy
| | | | - Anastasia De Luca
- d Department of Biology , "Tor Vergata" University of Rome , Rome , Italy
| | - Antonio Rosato
- e Department of Surgery, Oncology and Gastroenterology , University of Padova , Padova , Italy.,f Istituto Oncologico Veneto IRCCS , Padova , Italy
| | - Luigi Quintieri
- a Department of Pharmaceutical and Pharmacological Sciences , University of Padova , Padova , Italy
| | - Anna Maria Caccuri
- b Department of Experimental Medicine , "Tor Vergata" University of Rome , Rome , Italy.,g The NAST Centre for Nanoscience and Nanotechnology and Innovative Instrumentation , "Tor Vergata" University of Rome , Rome , Italy
| |
Collapse
|
18
|
Luebker SA, Koepsell SA. Diverse Mechanisms of BRAF Inhibitor Resistance in Melanoma Identified in Clinical and Preclinical Studies. Front Oncol 2019; 9:268. [PMID: 31058079 PMCID: PMC6478763 DOI: 10.3389/fonc.2019.00268] [Citation(s) in RCA: 141] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 03/25/2019] [Indexed: 12/13/2022] Open
Abstract
BRAF inhibitor therapy may provide profound initial tumor regression in metastatic melanoma with BRAF V600 mutations, but treatment resistance often leads to disease progression. A multi-center analysis of BRAF inhibitor resistant patient tissue samples detected genomic changes after disease progression including multiple secondary mutations in the MAPK/Erk signaling pathway, mutant BRAF copy number gains, and BRAF alternative splicing as the predominant putative mechanisms of resistance, but 41.7% of samples had no known resistance drivers. In vitro models of BRAF inhibitor resistance have been developed under a wide variety of experimental conditions to investigate unknown drivers of resistance. Several in vitro models developed genetic alterations observed in patient tissue, but others modulate the response to BRAF inhibitors through increased expression of receptor tyrosine kinases. Both secondary genetic alterations and expression changes in receptor tyrosine kinases may increase activation of MAPK/Erk signaling in the presence of BRAF inhibitors as well as activate PI3K/Akt signaling to support continued growth. Melanoma cells that develop resistance in vitro may have increased dependence on serine or glutamine metabolism and have increased cell motility and metastatic capacity. Future studies of BRAF inhibitor resistance in vitro would benefit from adhering to experimental parameters that reflect development of BRAF inhibitor resistance in patients through using multiple cell lines, fully characterizing the dosing strategy, and reporting the fold change in drug sensitivity.
Collapse
Affiliation(s)
- Stephen A Luebker
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Scott A Koepsell
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
19
|
Aldaghi SA, Jalal R. Concentration-Dependent Dual Effects of Ciprofloxacin on SB-590885-Resistant BRAF V600E A375 Melanoma Cells. Chem Res Toxicol 2019; 32:645-658. [PMID: 30829029 DOI: 10.1021/acs.chemrestox.8b00335] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BRAF inhibitors (BRAFi) have been applied to treat melanoma harboring V600E mutations. Several studies showed that BRAFi-resistant melanomas are dependent on mitochondrial biogenesis. Therefore, the present study aimed to investigate the influence of ciprofloxacin (CIP), a mitochondria-targeting antibiotic, on SB-590885-resistant BRAFV600E A375 melanoma (A375/SB) cells. The cytotoxicity activity of CIP and SB-590885, a potent and specific BRAFi, on A375 and A375/SB cells was evaluated by MTT, colony formation, migration, and spheroid formation assays. Moreover, SB-590885-induced cell death in A375 cells was analyzed. SB-590885 showed time- and concentration-dependent cytotoxic effects on A375 cells. Twenty-five μg/mL CIP decreased the cell viability of A375 and A375/SB cells in a time-dependent manner. This concentration of CIP markedly decreased clonogenicity in both cells and caused a reduction in the growth of A375/SB spheroids. The cytotoxicity of 5 μg/mL CIP on A375/SB cells was less than that of A375 cells. The colony formation and migration ability of A375/SB cells was increased in the presence of 5 μg/mL CIP. Ten μM SB-590885 induced a massive vacuolization in A375 cells. Cell death assays suggested a simultaneous activation of autophagy, paraptosis, apoptosis, and necrosis. For the first time, this study reveals that CIP at the maximum concentration in serum (5 μg/mL) can enhance the colony formation and migration abilities in BRAFi-resistant melanoma cells, while it has cytotoxic activity against these cells at a higher concentration than serum level. This study suggests that CIP may promote aggressive growth properties in BRAFi-resistant melanomas, at a concentration present in serum.
Collapse
Affiliation(s)
- Seyyede Araste Aldaghi
- Department of Chemistry, Faculty of Science , Ferdowsi University of Mashhad , Mashhad , Iran
| | - Razieh Jalal
- Department of Chemistry, Faculty of Science , Ferdowsi University of Mashhad , Mashhad , Iran.,Department of Research Cell and Molecular Biology, Institute of Biotechnology , Ferdowsi University of Mashhad , Mashhad , Iran
| |
Collapse
|
20
|
Hodgkinson A, Le Cam L, Trucu D, Radulescu O. Spatio-Genetic and phenotypic modelling elucidates resistance and re-sensitisation to treatment in heterogeneous melanoma. J Theor Biol 2019; 466:84-105. [PMID: 30503930 DOI: 10.1016/j.jtbi.2018.11.037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 11/06/2018] [Accepted: 11/29/2018] [Indexed: 12/11/2022]
|
21
|
Dong SC, Sha HH, Xu XY, Hu TM, Lou R, Li H, Wu JZ, Dan C, Feng J. Glutathione S-transferase π: a potential role in antitumor therapy. Drug Des Devel Ther 2018; 12:3535-3547. [PMID: 30425455 PMCID: PMC6204874 DOI: 10.2147/dddt.s169833] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Glutathione S-transferase π (GSTπ) is a Phase II metabolic enzyme that is an important facilitator of cellular detoxification. Traditional dogma asserts that GSTπ functions to catalyze glutathione (GSH)-substrate conjunction to preserve the macromolecule upon exposure to oxidative stress, thus defending cells against various toxic compounds. Over the past 20 years, abnormal GSTπ expression has been linked to the occurrence of tumor resistance to chemotherapy drugs, demonstrating that this enzyme possesses functions beyond metabolism. This revelation reveals exciting possibilities in the realm of drug discovery, as GSTπ inhibitors and its prodrugs offer a feasible strategy in designing anticancer drugs with the primary purpose of reversing tumor resistance. In connection with the authors' current research, we provide a review on the biological function of GSTπ and current developments in GSTπ-targeting drugs, as well as the prospects of future strategies.
Collapse
Affiliation(s)
- Shu-Chen Dong
- Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210009, China, ;
| | - Huan-Huan Sha
- Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210009, China, ;
| | - Xiao-Yue Xu
- Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210009, China, ;
| | - Tian-Mu Hu
- Department of Biological Science, Purdue University, West Lafayette, IN, USA
| | - Rui Lou
- Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210009, China, ;
| | - Huizi Li
- Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210009, China, ;
| | - Jian-Zhong Wu
- Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210009, China, ;
| | - Chen Dan
- Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210009, China, ;
| | - Jifeng Feng
- Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210009, China, ;
| |
Collapse
|
22
|
Wang X, Qu H, Dong Y, Wang G, Zhen Y, Zhang L. Targeting signal-transducer-and-activator-of-transcription 3 sensitizes human cutaneous melanoma cells to BRAF inhibitor. Cancer Biomark 2018; 23:67-77. [PMID: 30010109 DOI: 10.3233/cbm-181365] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Melanoma treatment with the BRAF V600E inhibitor vemurafenib provides therapeutic benefits but the common emergence of drug resistance remains a challenge. To define molecular mechanisms of vemurafenib resistance, we generated A375-R, WM35-R cell lines resistant to vemurafenib and show that the phosphorylated (p)-STAT3 was upregulated in these cells in vitro and in vivo. In particular, activation of the Signal-transducer-and-activator-of-transcription 3 (STAT3) pathway was associated with vemurafenib resistance. Inhibition of this pathway with STAT3-specific siRNA (shRNA) sensitized A375-R, WM35-R cells to vemurafenib and induced apoptosis in vitro and in vivo. Moreover, targeting STAT3 induced expression of miR-579-3p and elicited resistance to vemurafenib. However, targeting microRNA (miR)-579-3p with anti-miR-579-3p reversed the resistance to vemurafenib. Together, these results indicated that STAT3-mediated downexpression of miR-579-3p caused resistance to vemurafenib. Our findings suggest novel approaches to overcome resistance to vemurafenib by combining vemurafenib with STAT3 sliencing or miR-579-3p overexpression.
Collapse
Affiliation(s)
- Xiaohui Wang
- Department of Orthopedics, The Central Hospital of Linyi, Linyi, Shandong, China.,Department of Orthopedics, The Central Hospital of Linyi, Linyi, Shandong, China
| | - Huajun Qu
- Department of Cosmetic Plastic Surgery, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, Shandong, China.,Department of Orthopedics, The Central Hospital of Linyi, Linyi, Shandong, China
| | - Yinghe Dong
- Department of Cosmetic Plastic Surgery, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, Shandong, China
| | - Guozhi Wang
- Department of Cosmetic Plastic Surgery, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, Shandong, China
| | - Yuchen Zhen
- Department of Cosmetic Plastic Surgery, The Second Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Linxia Zhang
- Department of Medcine, The People's Hospital of Weifang, Weifang, Shandong, China
| |
Collapse
|
23
|
6-(7-nitro-2,1,3-benzoxadiazol-4-ylthio) hexanol: a promising new anticancer compound. Biosci Rep 2018; 38:BSR20171440. [PMID: 29358310 PMCID: PMC5809612 DOI: 10.1042/bsr20171440] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 01/20/2018] [Accepted: 01/22/2018] [Indexed: 12/23/2022] Open
Abstract
The 7-nitro-2,1,3-nitrobenzoxadiazole (NBD) derivatives are a series of compounds containing the NBD scaffold that are not glutathione (GSH) peptidomimetics, and result in a strong inhibition of glutathione S-transferases (GSTs). Growing evidences highlight their pivotal roles and outstanding anticancer activity in different tumor models. In particular, 6-(7-nitro-2,1,3-benzoxadiazol-4-ylthio) hexanol (NBDHEX) is extensively studied, which is a very efficient inhibitor of GSTP1-1. It triggers apoptosis in several tumor cell lines and this cytotoxic activity is observed at micro and submicromolar concentrations. Importantly, studies have shown that NBDHEX acts as an anticancer drug by inhibiting GSTs catalytic activity, avoiding inconvenience of the inhibitor extrusion from the cell by specific pumps and disrupting the interaction between the GSTP1-1 and key signaling effectors. Additionally, some researchers also have discovered that NBDHEX can act as late-phase autophagy inhibitor, which opens new opportunities to fully exploit its therapeutic potential. In this review, we summarize the advantages, anticancer mechanisms, and analogs of this compound, which will establish the basis on the usage of NBDHEX in clinical applications in future.
Collapse
|
24
|
Allocati N, Masulli M, Di Ilio C, Federici L. Glutathione transferases: substrates, inihibitors and pro-drugs in cancer and neurodegenerative diseases. Oncogenesis 2018; 7:8. [PMID: 29362397 PMCID: PMC5833873 DOI: 10.1038/s41389-017-0025-3] [Citation(s) in RCA: 379] [Impact Index Per Article: 54.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 12/12/2017] [Indexed: 12/12/2022] Open
Abstract
Glutathione transferase classical GSH conjugation activity plays a critical role in cellular detoxification against xenobiotics and noxious compounds as well as against oxidative stress. However, this feature is also exploited by cancer cells to acquire drug resistance and improve their survival. As a result, various members of the family were found overexpressed in a number of different cancers. Moreover several GST polymorphisms, ranging from null phenotypes to point mutations, were detected in members of the family and found to correlate with the onset of neuro-degenerative diseases. In the last decades, a great deal of research aimed at clarifying the role played by GSTs in drug resistance, at developing inhibitors to counteract this activity but also at exploiting GSTs for prodrugs specific activation in cancer cells. Here we summarize some of the most important achievements reached in this lively area of research.
Collapse
Affiliation(s)
- Nerino Allocati
- Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio", Chieti, Italy.
| | - Michele Masulli
- Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio", Chieti, Italy
| | - Carmine Di Ilio
- Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio", Chieti, Italy
| | - Luca Federici
- Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio", Chieti, Italy.,CESI-MET, University "G. d'Annunzio", Chieti, Italy
| |
Collapse
|
25
|
De Luca A, Carpanese D, Rapanotti MC, Viguria TMS, Forgione MA, Rotili D, Fulci C, Iorio E, Quintieri L, Chimenti S, Bianchi L, Rosato A, Caccuri AM. The nitrobenzoxadiazole derivative MC3181 blocks melanoma invasion and metastasis. Oncotarget 2017; 8:15520-15538. [PMID: 28107182 PMCID: PMC5362503 DOI: 10.18632/oncotarget.14690] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 12/27/2016] [Indexed: 02/01/2023] Open
Abstract
The novel nitrobenzoxadiazole (NBD) derivative MC3181 is endowed with remarkable therapeutic activity in mice bearing both sensitive and vemurafenib-resistant human melanoma xenografts. Here, we report that subtoxic concentrations of this compound significantly reduced invasiveness of BRAF-V600D mutated WM115 and WM266.4 melanoma cell lines derived from the primary lesion and related skin metastasis of the same patient, respectively. The strong antimetastatic activity of MC3181 was observed in both 2D monolayer cultures and 3D multicellular tumor spheroids, and confirmed in vivo by the significant decrease in the number of B16-F10 melanoma lung metastases in drug-treated mice. Our data also show that MC3181 affects the lactate production in the high glycolytic WM266.4 cell line. To unveil the MC3181 mechanism of action, we analyzed the ability of MC3181 to affect the degree of activation of different MAPK pathways, as well as the expression/activity levels of several proteins involved in angiogenesis, invasion, and survival (i.e. AP2, MCAM/MUC18, N-cadherin, VEGF and MMP-2). Our data disclosed both a decrease of the phospho-active form of JNK and an increased expression of the transcription factor AP2, events that occur in the very early phase of drug treatment and may be responsible of the antimetastatic effects of MC3181.
Collapse
Affiliation(s)
- Anastasia De Luca
- Department of Experimental Medicine and Surgery, University of Tor Vergata, 00133 Rome, Italy
| | - Debora Carpanese
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy
| | | | | | | | - Dante Rotili
- Department of Drug Chemistry and Technologies, "Sapienza" University, 00185 Rome, Italy
| | - Chiara Fulci
- Department of Experimental Medicine and Surgery, University of Tor Vergata, 00133 Rome, Italy
| | - Egidio Iorio
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Luigi Quintieri
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy
| | - Sergio Chimenti
- Department of Dermatology, University of Tor Vergata, 00133 Rome, Italy
| | - Luca Bianchi
- Department of Dermatology, University of Tor Vergata, 00133 Rome, Italy
| | - Antonio Rosato
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy.,Istituto Oncologico Veneto IOV-IRCCS, 35128 Padova, Italy
| | - Anna Maria Caccuri
- Department of Experimental Medicine and Surgery, University of Tor Vergata, 00133 Rome, Italy
| |
Collapse
|
26
|
Cesarini V, Guida E, Todaro F, Di Agostino S, Tassinari V, Nicolis S, Favaro R, Caporali S, Lacal PM, Botti E, Costanzo A, Rossi P, Jannini EA, Dolci S. Sox2 is not required for melanomagenesis, melanoma growth and melanoma metastasis in vivo. Oncogene 2017; 36:4508-4515. [PMID: 28368402 DOI: 10.1038/onc.2017.53] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 01/31/2017] [Accepted: 02/02/2017] [Indexed: 12/13/2022]
Abstract
Melanoma is a dangerous form of skin cancer derived from the malignant transformation of melanocytes. The transcription factor SOX2 is not expressed in melanocytes, however, it has been shown to be differentially expressed between benign nevi and malignant melanomas and to be essential for melanoma stem cell maintenance and expansion in vitro and in xenograft models. By using a mouse model in which BRafV600E mutation cooperates with Pten loss to induce the development of metastatic melanoma, we investigated if Sox2 is required during the process of melanomagenesis, melanoma growth and metastasis and in the acquisition of resistance to BRAF inhibitors (BRAFi) treatments. We found that deletion of Sox2 specifically in Pten null and BRafV600E-expressing melanocytes did not prevent tumor formation and did not modify the temporal kinetics of melanoma occurrence compared to Sox2 wt mice. In addition, tumor growth was similar between Sox2 wt and Sox2 deleted (del) melanomas. By querying publicly available databases, we did not find statistically significant differences in SOX2 expression levels between benign nevi and melanomas, and analysis on two melanoma patient cohorts confirmed that Sox2 levels did not significantly change between primary and metastatic melanomas. Melanoma cell lines derived from both Sox2 genotypes showed a similar sensitivity to vemurafenib treatment and the same ability to develop vemurafenib resistance in long-term cultures. Development of vemurafenib resistance was not dependent on SOX2 expression also in human melanoma cell lines in vitro. Our findings exclude an oncogenic function for Sox2 during melanoma development and do not support a role for this transcription factor in the acquisition of resistance to BRAFi treatments.
Collapse
Affiliation(s)
- V Cesarini
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - E Guida
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - F Todaro
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - S Di Agostino
- Regina Elena National Cancer Institute - IFO, Oncogenomic and Epigenetic Unit, Rome, Italy
| | - V Tassinari
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - S Nicolis
- Department of Biotechnology and Bioscience, University of Milan-Bicocca, Milan, Italy
| | - R Favaro
- Department of Biotechnology and Bioscience, University of Milan-Bicocca, Milan, Italy
| | - S Caporali
- Molecular Oncology Laboratory, Istituto Dermopatico dell'Immacolata - IRCCS, Rome, Italy
| | - P M Lacal
- Molecular Oncology Laboratory, Istituto Dermopatico dell'Immacolata - IRCCS, Rome, Italy
| | - E Botti
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - A Costanzo
- Department of Biomedicine, Humanitas University, Milan, Italy
| | - P Rossi
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - E A Jannini
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - S Dolci
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
27
|
Ceci C, Tentori L, Atzori MG, Lacal PM, Bonanno E, Scimeca M, Cicconi R, Mattei M, de Martino MG, Vespasiani G, Miano R, Graziani G. Ellagic Acid Inhibits Bladder Cancer Invasiveness and In Vivo Tumor Growth. Nutrients 2016; 8:744. [PMID: 27879653 PMCID: PMC5133127 DOI: 10.3390/nu8110744] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 11/11/2016] [Accepted: 11/16/2016] [Indexed: 12/15/2022] Open
Abstract
Ellagic acid (EA) is a polyphenolic compound that can be found as a naturally occurring hydrolysis product of ellagitannins in pomegranates, berries, grapes, green tea and nuts. Previous studies have reported the antitumor properties of EA mainly using in vitro models. No data are available about EA influence on bladder cancer cell invasion of the extracellular matrix triggered by vascular endothelial growth factor-A (VEGF-A), an angiogenic factor associated with disease progression and recurrence, and tumor growth in vivo. In this study, we have investigated EA activity against four different human bladder cancer cell lines (i.e., T24, UM-UC-3, 5637 and HT-1376) by in vitro proliferation tests (measuring metabolic and foci forming activity), invasion and chemotactic assays in response to VEGF-A and in vivo preclinical models in nude mice. Results indicate that EA exerts anti-proliferative effects as a single agent and enhances the antitumor activity of mitomycin C, which is commonly used for the treatment of bladder cancer. EA also inhibits tumor invasion and chemotaxis, specifically induced by VEGF-A, and reduces VEGFR-2 expression. Moreover, EA down-regulates the expression of programmed cell death ligand 1 (PD-L1), an immune checkpoint involved in immune escape. EA in vitro activity was confirmed by the results of in vivo studies showing a significant reduction of the growth rate, infiltrative behavior and tumor-associated angiogenesis of human bladder cancer xenografts. In conclusion, these results suggest that EA may have a potential role as an adjunct therapy for bladder cancer.
Collapse
Affiliation(s)
- Claudia Ceci
- Department of Systems Medicine, University of Rome Tor Vergata, Rome 00173, Italy.
| | - Lucio Tentori
- Department of Systems Medicine, University of Rome Tor Vergata, Rome 00173, Italy.
| | - Maria Grazia Atzori
- Department of Systems Medicine, University of Rome Tor Vergata, Rome 00173, Italy.
| | - Pedro M Lacal
- Laboratory of Molecular Oncology, "Istituto Dermopatico dell'Immacolata"-IRCCS, Rome 00167, Italy.
| | - Elena Bonanno
- Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, Rome 00173, Italy.
| | - Manuel Scimeca
- Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, Rome 00173, Italy.
| | - Rosella Cicconi
- "Centro di Servizi Interdipartimentale, Stazione per la Tecnologia Animale", Department of Biology, University of Rome Tor Vergata, Rome 00173, Italy.
| | - Maurizio Mattei
- "Centro di Servizi Interdipartimentale, Stazione per la Tecnologia Animale", Department of Biology, University of Rome Tor Vergata, Rome 00173, Italy.
| | - Maria Gabriella de Martino
- Laboratory of Signal Transduction, Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome 00173, Italy.
| | - Giuseppe Vespasiani
- Urology Unit, Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, Rome 00173, Italy.
| | - Roberto Miano
- Urology Unit, Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, Rome 00173, Italy.
| | - Grazia Graziani
- Department of Systems Medicine, University of Rome Tor Vergata, Rome 00173, Italy.
| |
Collapse
|
28
|
Sun D, Han Y, Wang W, Wang Z, Ma X, Hou Y, Bai G. Screening and identification of Caulis Sinomenii bioactive ingredients with dual-target NF-κB inhibition and β 2- AR agonizing activities. Biomed Chromatogr 2016; 30:1843-1853. [PMID: 27187693 DOI: 10.1002/bmc.3761] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 05/02/2016] [Accepted: 05/06/2016] [Indexed: 11/07/2022]
Abstract
Caulis Sinomenii (CS) is a valuable traditional medicine in China. Its extract can act as an anti-inflammatory agent and a vascular smooth muscle relaxant. However, the underlying mechanisms remain unknown. In this study, we developed a simple dual-target method based on ultra-performance liquid chromatography/quadrupole time-of-flight mass spectrometry combined with a dual-target bioactive screening assay for anti-inflammatory and antispasmodic activities to characterize the chemical structure of various bioactive compounds of CS rapidly. Seven potential NF-κB inhibitors were identified, including laudanosoline-1-O-xylopyranose, 6-O-methyl-laudanosoline-1-O-glucopyranoside, menisperine, sinomenine, laurifoline, magnoflorine and norsinoacutin. Furthermore, IL-6 and IL-8 assays confirmed the anti-inflammatory effects of these potential NF-κB inhibitors, in which laudanosoline-1-O-d-xylopyranose and menisperine were revealed as novel NF-κB inhibitors. Among the seven identified alkaloids, three potential β2 -adrenergic receptor agonists, including sinomenine, magnoflorine and laurifoline, were characterized using a luciferase reporter system to measure for the activity of β2 -adrenergic receptor agonists. Finally, sinomenine, magnoflorine and laurifoline were identified not only as potential NF-κB inhibitors but also as potential β2 -adrenegic receptor agonists, which is the first time this has been reported. Molecular dynamic simulation and docking results suggest that the three dual-bioactive constituents could not only inhibit Pseudomonas aeruginosa PAK strain-induced inflammatory responses via a negative regulation of the Braf protein that participates in MAPK signaling pathway but also activate the β2 -adrenegic receptor. These results suggest that CS extract has dual signaling activities with potential clinical application as a novel drug for asthma.
Collapse
Affiliation(s)
- Dan Sun
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, People's Republic of China
- Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, People's Republic of China
| | - Yanqi Han
- Department of Traditional Chinese Medicine, Tianjin Institute of Pharmaceutical Research Co. Ltd, Tianjin, People's Republic of China
| | - Weiya Wang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, People's Republic of China
- Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, People's Republic of China
| | - Zengyong Wang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, People's Republic of China
- Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, People's Republic of China
| | - Xiaoyao Ma
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, People's Republic of China
- Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, People's Republic of China
| | - Yuanyuan Hou
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, People's Republic of China.
- Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, People's Republic of China.
| | - Gang Bai
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, People's Republic of China
- Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, People's Republic of China
| |
Collapse
|
29
|
Palumbo C, De Luca A, Rosato N, Forgione M, Rotili D, Caccuri AM. c-Jun N-terminal kinase activation by nitrobenzoxadiazoles leads to late-stage autophagy inhibition. J Transl Med 2016; 14:37. [PMID: 26847645 PMCID: PMC4743117 DOI: 10.1186/s12967-016-0796-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Accepted: 01/20/2016] [Indexed: 03/23/2023] Open
Abstract
BACKGROUND Nitrobenzoxadiazole derivatives (NBDs), including NBDHEX and the recently developed MC3181, are promising anticancer agents able to target glutathione transferase and inhibit both its catalytic activity and ability to sequester TNF-receptor associated factor 2 (TRAF2) and c-Jun N-terminal kinase (JNK). NBDs have been shown to impair the growth and survival of a broad-spectrum of tumor types, in vitro and in vivo. Herein, we evaluated the effects of the new compound MC3181 on U-2OS osteosarcoma cells and investigated the impact of both NBDHEX and MC3181 on autophagy. METHODS Cell viability was evaluated by sulforhodamine B assay. The dissociation of the TRAF2-GSTP1-1 complex was detected by proximity ligation assay, while the phospho-activation of JNK was assessed by western blotting. The effects of NBDs on autophagy were evaluated by GFP-LC3 puncta formation, western blotting for LC3-II and p62, and LC3 turnover assay in the presence of bafilomycin A1. The role of JNK in the reduction of autophagic flux caused by NBDs was investigated using JNK1 shRNA-transfected cells. Fluorogenic caspase activity assay and flow cytometric analysis of DNA content were used to determine the cytotoxic effects of NBDs on JNK1-silenced cells. RESULTS Similar to NBDHEX, MC3181 reduced viability and activated TRAF2/JNK signaling in U-2OS cells. Moreover, NBDs induced the accumulation of autophagic vesicles and LC3-II while reducing both basal and nutritional stress-induced autophagic flux. Furthermore, increased levels of both LC3-II and the autophagy selective substrate p62 were observed in different tumor cell lines treated with NBDs, the concurrent increase of these markers being consistent with an impairment of autophagosome clearance. Autophagy inhibition by NBDs required JNK activity: NBDs caused autophagy inhibition and caspase-3 activation in JNK-positive U-2OS, but no autophagic flux inhibition or caspase-3 activation in JNK-silenced cells. CONCLUSIONS Our demonstration that NBDs can act as late-phase autophagy inhibitors opens new opportunities to fully exploit their therapeutic potential. This may not rely solely on their effectiveness in inducing cell cycle arrest and apoptosis, but also on their ability to weaken the capacity of tumor cells to endure stress conditions via autophagy. In addition, this study provides evidence that JNK can participate in impairing autophagy.
Collapse
Affiliation(s)
- Camilla Palumbo
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133, Rome, Italy.
| | - Anastasia De Luca
- Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, Via Montpellier 1, 00133, Rome, Italy.
| | - Nicola Rosato
- Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, Via Montpellier 1, 00133, Rome, Italy. .,The NAST Centre for Nanoscience and Nanotechnology and Innovative Instrumentation, University of Rome Tor Vergata, Via della Ricerca Scientifica 1, 00133, Rome, Italy.
| | - Mariantonietta Forgione
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185, Rome, Italy. .,Center for Life Nano Science@Sapienza, Italian Institute of Technology, Viale Regina Elena 291, 00161, Rome, Italy.
| | - Dante Rotili
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185, Rome, Italy.
| | - Anna Maria Caccuri
- Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, Via Montpellier 1, 00133, Rome, Italy. .,The NAST Centre for Nanoscience and Nanotechnology and Innovative Instrumentation, University of Rome Tor Vergata, Via della Ricerca Scientifica 1, 00133, Rome, Italy.
| |
Collapse
|