1
|
Riegel G, Orvain C, Recberlik S, Spaety ME, Poschet G, Venkatasamy A, Yamamoto M, Nomura S, Tsukamoto T, Masson M, Gross I, Le Lagadec R, Mellitzer G, Gaiddon C. The unfolded protein response-glutathione metabolism axis: A novel target of a cycloruthenated complexes bypassing tumor resistance mechanisms. Cancer Lett 2024; 585:216671. [PMID: 38290658 DOI: 10.1016/j.canlet.2024.216671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 12/22/2023] [Accepted: 01/20/2024] [Indexed: 02/01/2024]
Abstract
Platinum-based drugs remain the reference treatment for gastric cancer (GC). However, the frequency of resistance, due to mutations in TP53 or alterations in the energy and redox metabolisms, impairs the efficacy of current treatments, highlighting the need for alternative therapeutic options. Here, we show that a cycloruthenated compound targeting the redox metabolism, RDC11, induces higher cytotoxicity than oxaliplatin in GC cells and is more potent in reducing tumor growth in vivo. Detailed investigations into the mode of action of RDC11 indicated that it targets the glutathione (GSH) metabolism, which is an important drug resistance mechanism. We demonstrate that cycloruthenated complexes regulate the expression of enzymes of the transsulfuration pathway via the Unfolded Protein Response (UPR) and its effector ATF4. Furthermore, RDC11 induces the expression of SLC7A11 encoding for the cystine/glutamate antiporter xCT. These effects lead to a lower cellular GSH content and elevated oxygen reactive species production, causing the activation of a caspase-independent apoptosis. Altogether, this study provides the first evidence that cycloruthenated complexes target the GSH metabolism, neutralizing thereby a major resistance mechanism towards platinum-based chemotherapies and anticancer immune response.
Collapse
Affiliation(s)
- Gilles Riegel
- University of Strasbourg, INSERM UMR_S 1113, "Streinth" Laboratory, Strasbourg, France
| | - Christophe Orvain
- University of Strasbourg, INSERM UMR_S 1113, "Streinth" Laboratory, Strasbourg, France; INSERM, UMR 1260, CRBS, Regenerative Nanomedicine, "HERIIT" Laboratory, University of Strasbourg, Strasbourg, France
| | - Sevda Recberlik
- University of Strasbourg, INSERM UMR_S 1113, "Streinth" Laboratory, Strasbourg, France; INSERM, UMR 1260, CRBS, Regenerative Nanomedicine, "HERIIT" Laboratory, University of Strasbourg, Strasbourg, France
| | - Marie-Elodie Spaety
- University of Strasbourg, INSERM UMR_S 1113, "Streinth" Laboratory, Strasbourg, France
| | - Gernot Poschet
- Centre for Organismal Studies (COS), University of Heidelberg, Heidelberg, Germany
| | - Aina Venkatasamy
- University of Strasbourg, INSERM UMR_S 1113, "Streinth" Laboratory, Strasbourg, France; IHU-Strasbourg, Institute of Image-Guided Surgery, Strasbourg, France
| | - Masami Yamamoto
- Department of Laboratory of Physiological Pathology, Nippon Veterinary and Life Science University, Tokyo, Japan
| | - Sachiyo Nomura
- Department of Gastrointestinal Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tetsyua Tsukamoto
- Department of Diagnostic Pathology, Graduate School of Medicine, Fujita Health University, Toyoake, Japan
| | - Murielle Masson
- University of Strasbourg, INSERM UMR_S 1113, "Streinth" Laboratory, Strasbourg, France; University of Strasbourg, CNRS BSC-UMR 7242, Ecole Supérieure de Biotechnologie, Illkirch, France
| | - Isabelle Gross
- University of Strasbourg, INSERM UMR_S 1113, "SMART" Laboratory, Strasbourg, France; INSERM, UMR 1260, CRBS, Regenerative Nanomedicine, "HERIIT" Laboratory, University of Strasbourg, Strasbourg, France
| | - Ronan Le Lagadec
- Instituto de Química, Universidad Nacional Autónoma de México, Circuito Exterior s/n, Ciudad Universitaria, 04510, Ciudad de México, Mexico
| | - Georg Mellitzer
- University of Strasbourg, INSERM UMR_S 1113, "Streinth" Laboratory, Strasbourg, France; INSERM, UMR 1260, CRBS, Regenerative Nanomedicine, "HERIIT" Laboratory, University of Strasbourg, Strasbourg, France.
| | - Christian Gaiddon
- University of Strasbourg, INSERM UMR_S 1113, "Streinth" Laboratory, Strasbourg, France.
| |
Collapse
|
2
|
Klemt I, Varzatskii O, Selin R, Vakarov S, Kovalska V, Bila G, Bilyy R, Voloshin Y, Cuartero IC, Hidalgo A, Frey B, Becker I, Friedrich B, Tietze R, Friedrich RP, Alexiou C, Ursu EL, Rotaru A, Solymosi I, Pérez-Ojeda ME, Mokhir A. 3D-Shaped Binders of Unfolded Proteins Inducing Cancer Cell-Specific Endoplasmic Reticulum Stress In Vitro and In Vivo. J Am Chem Soc 2023; 145:22252-22264. [PMID: 37773090 DOI: 10.1021/jacs.3c08827] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/30/2023]
Abstract
The amount of unfolded proteins is increased in cancer cells, leading to endoplasmic reticulum (ER) stress. Therefore, cancer cells are sensitive to drugs capable of further enhancing ER stress. Examples of such drugs include the clinically approved proteosome inhibitors bortezomib and carfilzomib. Unfortunately, the known ER stress inducers exhibit dose-limiting side effects that justify the search for better, more cancer-specific drugs of this type. Herein, we report on FeC 2, which binds to unfolded proteins prevents their further processing, thereby leading to ER stress and ROS increase in cancer cells, but not in normal cells. FeC 2 exhibits low micromolar toxicity toward human acute promyelocytic leukemia HL-60, Burkitt's lymphoma BL-2, T-cell leukemia Jurkat, ovarian carcinoma A2780, lung cancer SK-MES-1, and murine lung cancer LLC1 cells. Due to the cancer-specific mode of action, 2 is not toxic in vivo up to the dose of 147 mg/kg, does not affect normal blood and bone marrow cells at the therapeutically active dose, but strongly suppresses both primary tumor growth (confirmed in Nemeth-Kellner lymphoma and LLC1 lung cancer models of murine tumor) and spreading of metastases (LLC1).
Collapse
Affiliation(s)
- Insa Klemt
- Department of Chemistry and Pharmacy, Organic Chemistry II, Friedrich-Alexander-University of Erlangen-Nürnberg (FAU), 91058 Erlangen, Germany
| | - Oleg Varzatskii
- Princeton Biomolecular Research Laboratories, 26A Saperne Pole Street, 01042 Kyiv, Ukraine
- V.I. Vernadsky Institute of General and Inorganic Chemistry, NASU, 32/34 Palladin Av., 03142 Kyiv, Ukraine
| | - Roman Selin
- Department of Chemistry and Pharmacy, Organic Chemistry II, Friedrich-Alexander-University of Erlangen-Nürnberg (FAU), 91058 Erlangen, Germany
| | - Serhii Vakarov
- Princeton Biomolecular Research Laboratories, 26A Saperne Pole Street, 01042 Kyiv, Ukraine
- V.I. Vernadsky Institute of General and Inorganic Chemistry, NASU, 32/34 Palladin Av., 03142 Kyiv, Ukraine
| | - Vladyslava Kovalska
- Princeton Biomolecular Research Laboratories, 26A Saperne Pole Street, 01042 Kyiv, Ukraine
- Institute of Molecular Biology and Genetics, NASU, 150 Zabolotnogo Street, 03143 Kyiv, Ukraine
| | - Galyna Bila
- Department of Histology, Cytology and Embryology, Danylo Halytsky Lviv National Medical University, Pekarska Street 69, 79010 Lviv, Ukraine
- Lectinotest R&D, Mechanichna Street 2, 79024 Lviv, Ukraine
| | - Rostyslav Bilyy
- Department of Histology, Cytology and Embryology, Danylo Halytsky Lviv National Medical University, Pekarska Street 69, 79010 Lviv, Ukraine
- Lectinotest R&D, Mechanichna Street 2, 79024 Lviv, Ukraine
| | - Yan Voloshin
- Nesmeyanov Institute of Organoelement Compounds, RAS, 28 Vavilova Street, 119334 Moscow, Russia
| | - Itziar Cossío Cuartero
- Program of Cardiovascular Regeneration, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), C. Melchor Fernández Almagro, 3, 28029 Madrid, Spain
| | - Andrés Hidalgo
- Program of Cardiovascular Regeneration, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), C. Melchor Fernández Almagro, 3, 28029 Madrid, Spain
| | - Benjamin Frey
- Department of Radiation Oncology, Translational Radiobiology, Universitaetsklinikum Erlangen, Friedrich-Alexander-University of Erlangen-Nürnberg (FAU), Glueckstrasse 4A, 91054 Erlangen, Germany
| | - Ina Becker
- Department of Radiation Oncology, Translational Radiobiology, Universitaetsklinikum Erlangen, Friedrich-Alexander-University of Erlangen-Nürnberg (FAU), Glueckstrasse 4A, 91054 Erlangen, Germany
| | - Bernhard Friedrich
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), University Hospital, Friedrich-Alexander-University of Erlangen-Nürnberg (FAU), Glückstraße 10a, 91054 Erlangen, Germany
| | - Rainer Tietze
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), University Hospital, Friedrich-Alexander-University of Erlangen-Nürnberg (FAU), Glückstraße 10a, 91054 Erlangen, Germany
| | - Ralf P Friedrich
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), University Hospital, Friedrich-Alexander-University of Erlangen-Nürnberg (FAU), Glückstraße 10a, 91054 Erlangen, Germany
| | - Christoph Alexiou
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), University Hospital, Friedrich-Alexander-University of Erlangen-Nürnberg (FAU), Glückstraße 10a, 91054 Erlangen, Germany
| | - Elena-Laura Ursu
- "Petru Poni" Institute of Macromolecular Chemistry, Romanian Academy, Centre of Advanced Research in Bionanoconjugates and Biopolymers, Grigore Ghica Voda Alley 41 A, 700487 Iasi, Romania
| | - Alexandru Rotaru
- "Petru Poni" Institute of Macromolecular Chemistry, Romanian Academy, Centre of Advanced Research in Bionanoconjugates and Biopolymers, Grigore Ghica Voda Alley 41 A, 700487 Iasi, Romania
| | - Iris Solymosi
- Department of Chemistry and Pharmacy, Organic Chemistry II, Friedrich-Alexander-University of Erlangen-Nürnberg (FAU), 91058 Erlangen, Germany
| | - M Eugenia Pérez-Ojeda
- Department of Chemistry and Pharmacy, Organic Chemistry II, Friedrich-Alexander-University of Erlangen-Nürnberg (FAU), 91058 Erlangen, Germany
| | - Andriy Mokhir
- Department of Chemistry and Pharmacy, Organic Chemistry II, Friedrich-Alexander-University of Erlangen-Nürnberg (FAU), 91058 Erlangen, Germany
| |
Collapse
|
3
|
Łomzik M, Błauż A, Głodek M, Makal A, Tchoń D, Ayine-Tora DM, Hartinger C, Rychlik B, Plażuk D. Organometallic Ru, Os, Rh and Ir half-sandwich conjugates of ispinesib - impact of the organometallic group on the antimitotic activity. Dalton Trans 2023; 52:11859-11874. [PMID: 37464882 DOI: 10.1039/d3dt01217d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
Antimitotic agents are among the most important drugs used in anticancer therapy. Kinesin spindle protein (KSP) was proposed as a promising target for new antimitotic drugs. Herein, we report the synthesis of Ru, Os, Rh, and Ir half-sandwich complexes with the KSP inhibitor ispinesib and its (S)-enantiomer. Conjugation of the organometallic moiety with ispinesib and its (S)-enantiomer resulted in a significantly increased cytotoxicity of up to 5.6-fold compared to the parent compounds, with IC50 values in the nanomolar range. The most active derivatives were the ispinesib Ru and Rh conjugates which were able to generate reactive oxygen species (ROS), which may at least partially explain their high cytotoxicity. At the same time, the Os and Ir derivatives acted as KSP inhibitors with no effects on ROS generation.
Collapse
Affiliation(s)
- Michał Łomzik
- Department of Organic Chemistry, Faculty of Chemistry, University of Lodz, ul. Tamka 12, 91-403 Łódź, Poland.
| | - Andrzej Błauż
- Cytometry Lab, Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, ul. Pomorska 141/143, 90-236 Łódź, Poland
| | - Marta Głodek
- Department of Organic Chemistry, Faculty of Chemistry, University of Lodz, ul. Tamka 12, 91-403 Łódź, Poland.
| | - Anna Makal
- Laboratory for Structural and Biochemical Research (LBSBio), Biological and Chemical Research Centre, Department of Chemistry, University of Warsaw, ul. Zwirki i Wigury 101, 02-089 Warszawa, Poland
| | - Daniel Tchoń
- Laboratory for Structural and Biochemical Research (LBSBio), Biological and Chemical Research Centre, Department of Chemistry, University of Warsaw, ul. Zwirki i Wigury 101, 02-089 Warszawa, Poland
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | | | - Christian Hartinger
- School of Chemical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Błażej Rychlik
- Cytometry Lab, Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, ul. Pomorska 141/143, 90-236 Łódź, Poland
| | - Damian Plażuk
- Department of Organic Chemistry, Faculty of Chemistry, University of Lodz, ul. Tamka 12, 91-403 Łódź, Poland.
| |
Collapse
|
4
|
Targeting emerging cancer hallmarks by transition metal complexes: Epigenetic reprogramming and epitherapies. Part II. Coord Chem Rev 2023. [DOI: 10.1016/j.ccr.2022.214899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
5
|
Synthesis, characterization and anticancer activities of cationic η6-p-cymene ruthenium(II) complexes containing phosphine and nitrogenous ligands. Polyhedron 2022. [DOI: 10.1016/j.poly.2022.115980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
6
|
ER Stress Response and Induction of Apoptosis in Malignant Pleural Mesothelioma: The Achilles Heel Targeted by the Anticancer Ruthenium Drug BOLD-100. Cancers (Basel) 2022; 14:cancers14174126. [PMID: 36077664 PMCID: PMC9454852 DOI: 10.3390/cancers14174126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/02/2022] [Accepted: 08/23/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Malignant mesothelioma is a rare cancer arising from the serosal surfaces of the body, mainly from the pleural layer. This cancer, strongly linked to asbestos exposure, shows a very inauspicious prognosis. In fact, there is no efficient therapeutic treatment for malignant pleural mesothelioma (MPM). Thus, there is an urgent need to develop novel therapeutic approaches to treat this form of cancer. Our previous study showed the importance of GRP78 in MPM survival. BOLD-100 is a specific modulator of GRP78 and we have observed that it shows cytotoxicity against MPM cells. In particular, we describe that BOLD-100 increases oxidative stress and deregulates the calcium homeostasis leading to cell stress and, ultimately, to cell death. Our in vitro data strongly suggest that BOLD-100 inhibits the growth of MPM cell lines, proposing the application as a single agent, or in combination with other standard-of-care drugs, to treat MPM. Abstract Malignant mesothelioma is a rare cancer arising from the serosal surfaces of the body, mainly from the pleural layer. This cancer is strongly related to asbestos exposure and shows a very inauspicious prognosis, because there are scarce therapeutic options for this rare disease. Thus, there is an urgent need to develop novel therapeutic approaches to treat this form of cancer. To explore the biology of malignant pleural mesothelioma (MPM), we previously observed that MPM cell lines show high expression of the GRP78 protein, which is a chaperone protein and the master regulator of the unfolded protein response (UPR) that resides in the endoplasmic reticulum (ER). Based on our previous studies showing the importance of GRP78 in MPM, we observed that BOLD-100, a specific modulator of GRP78 and the UPR, shows cytotoxicity against MPM cells. Our studies demonstrated that BOLD-100 increases ROS production and Ca2+ release from the ER, leading to ER stress activation and, ultimately, to cell death. Our in vitro data strongly suggest that BOLD-100 inhibits the growth of MPM cell lines, proposing the application as a single agent, or in combination with other standard-of-care drugs, to treat MPM.
Collapse
|
7
|
Gaiddon C, Gross I, Meng X, Sidhoum M, Mellitzer G, Romain B, Delhorme JB, Venkatasamy A, Jung AC, Pfeffer M. Bypassing the Resistance Mechanisms of the Tumor Ecosystem by Targeting the Endoplasmic Reticulum Stress Pathway Using Ruthenium- and Osmium-Based Organometallic Compounds: An Exciting Long-Term Collaboration with Dr. Michel Pfeffer. Molecules 2021; 26:molecules26175386. [PMID: 34500819 PMCID: PMC8434532 DOI: 10.3390/molecules26175386] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 08/30/2021] [Accepted: 08/31/2021] [Indexed: 12/27/2022] Open
Abstract
Metal complexes have been used to treat cancer since the discovery of cisplatin and its interaction with DNA in the 1960’s. Facing the resistance mechanisms against platinum salts and their side effects, safer therapeutic approaches have been sought through other metals, including ruthenium. In the early 2000s, Michel Pfeffer and his collaborators started to investigate the biological activity of organo-ruthenium/osmium complexes, demonstrating their ability to interfere with the activity of purified redox enzymes. Then, they discovered that these organo-ruthenium/osmium complexes could act independently of DNA damage and bypass the requirement for the tumor suppressor gene TP53 to induce the endoplasmic reticulum (ER) stress pathway, which is an original cell death pathway. They showed that other types of ruthenium complexes—as well complexes with other metals (osmium, iron, platinum)—can induce this pathway as well. They also demonstrated that ruthenium complexes accumulate in the ER after entering the cell using passive and active mechanisms. These particular physico-chemical properties of the organometallic complexes designed by Dr. Pfeffer contribute to their ability to reduce tumor growth and angiogenesis. Taken together, the pioneering work of Dr. Michel Pfeffer over his career provides us with a legacy that we have yet to fully embrace.
Collapse
Affiliation(s)
- Christian Gaiddon
- Université de Strasbourg-Inserm, UMR_S 1113 IRFAC, 67200 Strasbourg, France; (I.G.); (G.M.); (B.R.); (J.-B.D.); (A.V.); (J.A.C.)
- Correspondence: ; Tel.: +33-6-8352-5356
| | - Isabelle Gross
- Université de Strasbourg-Inserm, UMR_S 1113 IRFAC, 67200 Strasbourg, France; (I.G.); (G.M.); (B.R.); (J.-B.D.); (A.V.); (J.A.C.)
| | - Xiangjun Meng
- Department of Gastro-Oncology, 7th Hospital, Shanghai 200137, China;
| | | | - Georg Mellitzer
- Université de Strasbourg-Inserm, UMR_S 1113 IRFAC, 67200 Strasbourg, France; (I.G.); (G.M.); (B.R.); (J.-B.D.); (A.V.); (J.A.C.)
| | - Benoit Romain
- Université de Strasbourg-Inserm, UMR_S 1113 IRFAC, 67200 Strasbourg, France; (I.G.); (G.M.); (B.R.); (J.-B.D.); (A.V.); (J.A.C.)
| | - Jean-Batiste Delhorme
- Université de Strasbourg-Inserm, UMR_S 1113 IRFAC, 67200 Strasbourg, France; (I.G.); (G.M.); (B.R.); (J.-B.D.); (A.V.); (J.A.C.)
| | - Aïna Venkatasamy
- Université de Strasbourg-Inserm, UMR_S 1113 IRFAC, 67200 Strasbourg, France; (I.G.); (G.M.); (B.R.); (J.-B.D.); (A.V.); (J.A.C.)
| | - Alain C. Jung
- Université de Strasbourg-Inserm, UMR_S 1113 IRFAC, 67200 Strasbourg, France; (I.G.); (G.M.); (B.R.); (J.-B.D.); (A.V.); (J.A.C.)
| | - Michel Pfeffer
- CNRS UMR 7177, Institute of Chemistry, 67000 Strasbourg, France;
| |
Collapse
|
8
|
Wei W, Jia G. Metal-Carbon Bonds of Heavier Group 7 and 8 Metals (Tc, Re, Ru, Os): Mononuclear Tc/Re/Ru/Os Complexes With Metal-Carbon Bonds. COMPREHENSIVE COORDINATION CHEMISTRY III 2021:123-439. [DOI: 10.1016/b978-0-08-102688-5.00049-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
9
|
Estrada-Montaño AS, Gries A, Oviedo-Fortino JA, Torres-Gutierrez C, Grain-Hayton A, Marcial-Hernández R, Shen L, Ryabov AD, Gaiddon C, Le Lagadec R. Dibromine Promoted Transmetalation of an Organomercurial by Fe(CO)5: Synthesis, Properties, and Cytotoxicity of Bis(2-C6H4-2′-py-κC,N)dicarbonyliron(II). Organometallics 2020. [DOI: 10.1021/acs.organomet.0c00107] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Aldo S. Estrada-Montaño
- Instituto de Quı́mica UNAM, Circuito Exterior s/n, Ciudad Universitaria, 04510 Ciudad de México, Mexico
| | - Alexandre Gries
- Strasbourg Université, Inserm UMR_S U1113, IRFAC, 3 Avenue Molière, 67200 Strasbourg, France
| | - José A. Oviedo-Fortino
- Instituto de Quı́mica UNAM, Circuito Exterior s/n, Ciudad Universitaria, 04510 Ciudad de México, Mexico
| | - Carolina Torres-Gutierrez
- Instituto de Quı́mica UNAM, Circuito Exterior s/n, Ciudad Universitaria, 04510 Ciudad de México, Mexico
| | - Amira Grain-Hayton
- Instituto de Quı́mica UNAM, Circuito Exterior s/n, Ciudad Universitaria, 04510 Ciudad de México, Mexico
| | | | - Longzhu Shen
- University of Cambridge, CB2 3EJ Cambridge, United Kingdom
| | - Alexander D. Ryabov
- Department of Chemistry, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, Pennsylvania 15213, United States
| | - Christian Gaiddon
- Strasbourg Université, Inserm UMR_S U1113, IRFAC, 3 Avenue Molière, 67200 Strasbourg, France
| | - Ronan Le Lagadec
- Instituto de Quı́mica UNAM, Circuito Exterior s/n, Ciudad Universitaria, 04510 Ciudad de México, Mexico
| |
Collapse
|
10
|
Solís-Ruiz JA, Barthe A, Riegel G, Saavedra-Díaz RO, Gaiddon C, Le Lagadec R. Light activation of cyclometalated ruthenium complexes drives towards caspase 3 dependent apoptosis in gastric cancer cells. J Inorg Biochem 2020; 208:111080. [PMID: 32330762 DOI: 10.1016/j.jinorgbio.2020.111080] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 03/24/2020] [Accepted: 03/24/2020] [Indexed: 12/20/2022]
Abstract
Polypyridyl ruthenium complexes have been intensively investigated for their remarkable antiproliferative properties and some are currently being tested in clinical trials. Here, we investigated the impact of illumination on the biological properties of a series of new cyclometalated ruthenium compounds with increased π-conjugation. We determined that various of these complexes display a bivalent biological activity as they are highly cytotoxic by themselves in absence of light while their cytotoxicity can significantly be elevated towards an IC50 in the nanomolar range upon illumination. In particular, we showed that these complexes are particularly active (IC50 < 1 μM) on two gastric cancer cell lines (AGS, KATO III) that are resistant towards cisplatin (IC50 > 25 μM). As expected, light activation leads to increased production of singlet oxygen species in vitro and accumulation of reactive oxygen species in vivo. Importantly, we established that light exposure shifts the mode of action of the complexes towards activation of a caspase 3-dependent apoptosis that correlates with increased DNA damage. Altogether, this study characterizes novel ruthenium complexes with dual activity that can be tuned towards different mode of action in order to bypass cancer cell resistance mechanisms.
Collapse
Affiliation(s)
- Jorge Andrés Solís-Ruiz
- Instituto de Química, Universidad Nacional Autónoma de México, Circuito Exterior s/n, Ciudad Universitaria, 04510 Ciudad de México, Mexico
| | - Anaïs Barthe
- Strasbourg Université, Inserm UMR_S U1113, IRFAC, 3 Avenue Molière, 67200 Strasbourg, France
| | - Gilles Riegel
- Strasbourg Université, Inserm UMR_S U1113, IRFAC, 3 Avenue Molière, 67200 Strasbourg, France
| | - Rafael Omar Saavedra-Díaz
- Universidad Juárez Autónoma de Tabasco, División Académica de Ciencias Básicas, Carretera Cunduacán-Jalpa Km. 1, 86690 Cunduacán, Tabasco, Mexico
| | - Christian Gaiddon
- Strasbourg Université, Inserm UMR_S U1113, IRFAC, 3 Avenue Molière, 67200 Strasbourg, France.
| | - Ronan Le Lagadec
- Instituto de Química, Universidad Nacional Autónoma de México, Circuito Exterior s/n, Ciudad Universitaria, 04510 Ciudad de México, Mexico.
| |
Collapse
|
11
|
Cullinane D, Gkika KS, Byrne A, Keyes TE. Photostable NIR emitting ruthenium(II) conjugates; uptake and biological activity in live cells. J Inorg Biochem 2020; 207:111032. [PMID: 32311630 DOI: 10.1016/j.jinorgbio.2020.111032] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 02/11/2020] [Accepted: 02/12/2020] [Indexed: 01/19/2023]
Abstract
A photostable Ru(2,2-biquinoline)2(3-(2-pyridyl)-5-(4-carboxyphenyl)-1,2,4-triazolate) (Ru(biq)2(trzbenzCOOH)) complex that exhibits near-infrared (NIR) emission centred at 786 nm is reported. The parent complex was conjugated via amide coupling to a cell-penetrating peptide sequence octa-arginine (R8), and two signal peptide sequences; the nuclear localizing sequence (NLS) VQRKRQKLMP and the mitochondria penetrating peptide (MPP) FrFKFrFK(Ac) (r = D isomer of arginine, Ac = terminal lysine amine acetyl blocked). Notably, none of the peptide conjugates were cell-permeable as chloride salts but efficient and rapid membrane permeation was observed post ion exchange with perchlorate counterion. Also, surprisingly, all three peptide conjugates exhibited potent dark cytotoxicity in both CHO and HeLa cell lines. The peptide conjugates induce cell death through a caspase dependent apoptotic pathway. At the minimum concentration of dye (approx. 15 μM) required for cell imaging, only 20% of the cells were viable after a 24 h incubation period. To overcome cytotoxicity, the parent complex was PEGylated; this dramatically decreased cytotoxicity, where 50% of cells were viable even at 150 μM concentration after 24 h. Confocal luminescence microscopy indicated that all four bioconjugates, peptides in perchlorate form and polyethylene glycol (PEG) in chloride form, were rapidly internalized within the cell. However, interestingly the precise localisation by the signal peptides observed in related complexes was not observed here and the peptide conjugates were unsuitable as luminescent probes for cell microscopy due to their high cell toxicity. The poor targeting of signal peptides in this instance is attributed to the high lipophilicity of the metal centre.
Collapse
Affiliation(s)
- David Cullinane
- School of Chemical Sciences, National Centre for Sensor Research, Dublin City University, Dublin 9, Ireland
| | - Karmel Sofia Gkika
- School of Chemical Sciences, National Centre for Sensor Research, Dublin City University, Dublin 9, Ireland
| | - Aisling Byrne
- School of Chemical Sciences, National Centre for Sensor Research, Dublin City University, Dublin 9, Ireland
| | - Tia E Keyes
- School of Chemical Sciences, National Centre for Sensor Research, Dublin City University, Dublin 9, Ireland.
| |
Collapse
|
12
|
Licona C, Delhorme JB, Riegel G, Vidimar V, Cerón-Camacho R, Boff B, Venkatasamy A, Tomasetto C, da Silva Figueiredo Celestino Gomes P, Rognan D, Freund JN, Le Lagadec R, Pfeffer M, Gross I, Mellitzer G, Gaiddon C. Anticancer activity of ruthenium and osmium cyclometalated compounds: identification of ABCB1 and EGFR as resistance mechanisms. Inorg Chem Front 2020. [DOI: 10.1039/c9qi01148j] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Switching from ruthenium to osmium reduces sensitivity towards ABCB1 resistance for cyclometalated anticancer drugs.
Collapse
|
13
|
King AP, Wilson JJ. Endoplasmic reticulum stress: an arising target for metal-based anticancer agents. Chem Soc Rev 2020; 49:8113-8136. [DOI: 10.1039/d0cs00259c] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Metal anticancer agents are rapidly emerging as selective, potent therapeutics that exhibit anticancer activity by inducing endoplasmic reticulum stress.
Collapse
Affiliation(s)
- A. Paden King
- Department of Chemistry and Chemical Biology
- Cornell University
- Ithaca
- USA
| | - Justin J. Wilson
- Department of Chemistry and Chemical Biology
- Cornell University
- Ithaca
- USA
| |
Collapse
|
14
|
Spaety ME, Gries A, Badie A, Venkatasamy A, Romain B, Orvain C, Yanagihara K, Okamoto K, Jung AC, Mellitzer G, Pfeffer S, Gaiddon C. HDAC4 Levels Control Sensibility toward Cisplatin in Gastric Cancer via the p53-p73/BIK Pathway. Cancers (Basel) 2019; 11:cancers11111747. [PMID: 31703394 PMCID: PMC6896094 DOI: 10.3390/cancers11111747] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 10/23/2019] [Accepted: 10/31/2019] [Indexed: 02/08/2023] Open
Abstract
Gastric cancer (GC) remains a health issue due to the low efficiency of therapies, such as cisplatin. This unsatisfactory situation highlights the necessity of finding factors impacting GC sensibility to therapies. We analyzed the cisplatin pangenomic response in cancer cells and found HDAC4 as a major epigenetic regulator being inhibited. HDAC4 mRNA repression was partly mediated by the cisplatin-induced expression of miR-140. At a functional level, HDAC4 inhibition favored cisplatin cytotoxicity and reduced tumor growth. Inversely, overexpression of HDAC4 inhibits cisplatin cytotoxicity. Importantly, HDAC4 expression was found to be elevated in gastric tumors compared to healthy tissues, and in particular in specific molecular subgroups. Furthermore, mutations in HDAC4 correlate with good prognosis. Pathway analysis of genes whose expression in patients correlated strongly with HDAC4 highlighted DNA damage, p53 stabilization, and apoptosis as processes downregulated by HDAC4. This was further confirmed by silencing of HDAC4, which favored cisplatin-induced apoptosis characterized by cleavage of caspase 3 and induction of proapoptotic genes, such as BIK, in part via a p53-dependent mechanism. Altogether, these results reveal HDAC4 as a resistance factor for cisplatin in GC cells that impacts on patients' survival.
Collapse
Affiliation(s)
- Marie-Elodie Spaety
- Laboratory STREINTH (Stress Response and Innovative Therapies), Inserm IRFAC UMR_S1113, Université de Strasbourg, 3 av. Molière, 67200 Strasbourg, France; (M.-E.S.); (A.G.); (A.B.); (A.V.); (B.R.); (C.O.); (A.C.J.); (G.M.)
- Architecture and Reactivity of RNA, Institut de biologie moléculaire et cellulaire du CNRS, Université de Strasbourg, 15 rue René Descartes, 67084 Strasbourg, France;
| | - Alexandre Gries
- Laboratory STREINTH (Stress Response and Innovative Therapies), Inserm IRFAC UMR_S1113, Université de Strasbourg, 3 av. Molière, 67200 Strasbourg, France; (M.-E.S.); (A.G.); (A.B.); (A.V.); (B.R.); (C.O.); (A.C.J.); (G.M.)
| | - Amandine Badie
- Laboratory STREINTH (Stress Response and Innovative Therapies), Inserm IRFAC UMR_S1113, Université de Strasbourg, 3 av. Molière, 67200 Strasbourg, France; (M.-E.S.); (A.G.); (A.B.); (A.V.); (B.R.); (C.O.); (A.C.J.); (G.M.)
| | - Aina Venkatasamy
- Laboratory STREINTH (Stress Response and Innovative Therapies), Inserm IRFAC UMR_S1113, Université de Strasbourg, 3 av. Molière, 67200 Strasbourg, France; (M.-E.S.); (A.G.); (A.B.); (A.V.); (B.R.); (C.O.); (A.C.J.); (G.M.)
- Radiology Department, Centre Hospitalier Universitaire (CHU) Hautepierre, 67200 Strasbourg, France
| | - Benoit Romain
- Laboratory STREINTH (Stress Response and Innovative Therapies), Inserm IRFAC UMR_S1113, Université de Strasbourg, 3 av. Molière, 67200 Strasbourg, France; (M.-E.S.); (A.G.); (A.B.); (A.V.); (B.R.); (C.O.); (A.C.J.); (G.M.)
- Digestive Surgery department, CHU Hautepierre, 67200 Strasbourg, France
| | - Christophe Orvain
- Laboratory STREINTH (Stress Response and Innovative Therapies), Inserm IRFAC UMR_S1113, Université de Strasbourg, 3 av. Molière, 67200 Strasbourg, France; (M.-E.S.); (A.G.); (A.B.); (A.V.); (B.R.); (C.O.); (A.C.J.); (G.M.)
| | | | - Koji Okamoto
- National Cancer Research Center, Tokyo 104_0045, Japan; (K.Y.); (K.O.)
| | - Alain C. Jung
- Laboratory STREINTH (Stress Response and Innovative Therapies), Inserm IRFAC UMR_S1113, Université de Strasbourg, 3 av. Molière, 67200 Strasbourg, France; (M.-E.S.); (A.G.); (A.B.); (A.V.); (B.R.); (C.O.); (A.C.J.); (G.M.)
- Centre de Lutte contre le Cancer Paul Strauss (CLCC), 67065 Strasbourg, France
| | - Georg Mellitzer
- Laboratory STREINTH (Stress Response and Innovative Therapies), Inserm IRFAC UMR_S1113, Université de Strasbourg, 3 av. Molière, 67200 Strasbourg, France; (M.-E.S.); (A.G.); (A.B.); (A.V.); (B.R.); (C.O.); (A.C.J.); (G.M.)
- Centre de Lutte contre le Cancer Paul Strauss (CLCC), 67065 Strasbourg, France
| | - Sébastien Pfeffer
- Architecture and Reactivity of RNA, Institut de biologie moléculaire et cellulaire du CNRS, Université de Strasbourg, 15 rue René Descartes, 67084 Strasbourg, France;
| | - Christian Gaiddon
- Laboratory STREINTH (Stress Response and Innovative Therapies), Inserm IRFAC UMR_S1113, Université de Strasbourg, 3 av. Molière, 67200 Strasbourg, France; (M.-E.S.); (A.G.); (A.B.); (A.V.); (B.R.); (C.O.); (A.C.J.); (G.M.)
- Centre de Lutte contre le Cancer Paul Strauss (CLCC), 67065 Strasbourg, France
- Correspondence:
| |
Collapse
|
15
|
Mathur D, Medintz IL. The Growing Development of DNA Nanostructures for Potential Healthcare-Related Applications. Adv Healthc Mater 2019; 8:e1801546. [PMID: 30843670 PMCID: PMC9285959 DOI: 10.1002/adhm.201801546] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 01/17/2019] [Indexed: 12/21/2022]
Abstract
DNA self-assembly has proven to be a highly versatile tool for engineering complex and dynamic biocompatible nanostructures from the bottom up with a wide range of potential bioapplications currently being pursued. Primary among these is healthcare, with the goal of developing diagnostic, imaging, and drug delivery devices along with combinatorial theranostic devices. The path to understanding a role for DNA nanotechnology in biomedical sciences is being approached carefully and systematically, starting from analyzing the stability and immune-stimulatory properties of DNA nanostructures in physiological conditions, to estimating their accessibility and application inside cellular and model animal systems. Much remains to be uncovered but the field continues to show promising results toward developing useful biomedical devices. This review discusses some aspects of DNA nanotechnology that makes it a favorable ingredient for creating nanoscale research and biomedical devices and looks at experiments undertaken to determine its stability in vivo. This is presented in conjugation with examples of state-of-the-art developments in biomolecular sensing, imaging, and drug delivery. Finally, some of the major challenges that warrant the attention of the scientific community are highlighted, in order to advance the field into clinically relevant applications.
Collapse
Affiliation(s)
- Divita Mathur
- Center for Bio/Molecular Science and EngineeringU.S. Naval Research Laboratory Code 6910WashingtonDC20375USA
- College of ScienceGeorge Mason UniversityFairfaxVA22030USA
| | - Igor L. Medintz
- Center for Bio/Molecular Science and EngineeringU.S. Naval Research Laboratory Code 6907WashingtonDC20375USA
| |
Collapse
|
16
|
Briš A, Jašík J, Turel I, Roithová J. Anti-cancer organoruthenium(ii) complexes and their interactions with cysteine and its analogues. A mass-spectrometric study. Dalton Trans 2019; 48:2626-2634. [PMID: 30702097 PMCID: PMC8609305 DOI: 10.1039/c8dt04350g] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The ruthenium complexes [Ru(CYM)(p-Cl-dkt)(Cl)] (1), [Ru(CYM)(pta)(p-Cl-dkt)]PF6 (2), and [Ru(CYM)(pta)Cl2] (3, RAPTA-C) (CYM = para-cymene, p-Cl-dkt = 1-(4-chlorophenyl)-4,4,4-trifluorobutane-1,3-dione, pta = 1,3,5-triaza-7-phosphaadamantane) are biologically active and show anti-cancer activities, albeit with different mechanisms. To further understand these mechanisms, we compared their speciation in aqueous solutions with an amino acid (cysteine), with an amino acid derivative (N-acetylcysteine) and with a tripeptide (glutathione) by Mass Spectrometry (MS). Here, we show that all ruthenium complexes have high selectivity for cysteine and cysteine-derived molecules. On one hand, [Ru(CYM)(p-Cl-dkt)(Cl)] undergoes solvolysis in water and forms [Ru2(CYM)2(OH)3]+. Subsequently, all hydroxyl anions are exchanged by deprotonated cysteine. Infrared Photodissociation Spectroscopy (IRPD) showed that cysteine binds to the ruthenium atoms via the deprotonated thiol group and that sulfur bridges the ruthenium centers. On the other hand, the pta-bearing complexes remain monometallic and undergo only slow Cl or p-Cl-dkt exchange by deprotonated cysteine. Therefore, the pta ligand protects the ruthenium complexes from ligand exchange with water and from the formation of biruthenium clusters, possibly explaining why the mechanism of pta-bearing ruthenium complexes is not based on ROS production but on their reactivity as monometallic complexes. ESI-MS study of ruthenium complexes shows their high selectivity toward thiol containing molecules and formation of larger thiolate-bound clusters in absence of a protecting ligand such as pta.![]()
Collapse
Affiliation(s)
- Anamarija Briš
- Department of Organic Chemistry, Faculty of Science, Charles University, Hlavova 2030/8, 128 43 Prague, Czech Republic
| | | | | | | |
Collapse
|
17
|
Vidimar V, Licona C, Cerón-Camacho R, Guerin E, Coliat P, Venkatasamy A, Ali M, Guenot D, Le Lagadec R, Jung AC, Freund JN, Pfeffer M, Mellitzer G, Sava G, Gaiddon C. A redox ruthenium compound directly targets PHD2 and inhibits the HIF1 pathway to reduce tumor angiogenesis independently of p53. Cancer Lett 2019; 440-441:145-155. [DOI: 10.1016/j.canlet.2018.09.029] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 09/08/2018] [Accepted: 09/24/2018] [Indexed: 12/25/2022]
|
18
|
Chow MJ, Babak MV, Tan KW, Cheong MC, Pastorin G, Gaiddon C, Ang WH. Induction of the Endoplasmic Reticulum Stress Pathway by Highly Cytotoxic Organoruthenium Schiff-Base Complexes. Mol Pharm 2018; 15:3020-3031. [PMID: 29979603 DOI: 10.1021/acs.molpharmaceut.8b00003] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Current anticancer drug discovery efforts focus on the identification of first-in-class compounds with a mode-of-action distinct from conventional DNA-targeting agents for chemotherapy. An emerging trend is the identification of endoplasmic reticulum (ER) targeting compounds that induce ER stress in cancer cells, leading to cell death. However, a limited pool of such compounds has been identified to date, and there are limited studies done on such compounds to allow for the rational design of ER stress-inducing agents. In our present study, we present a series of highly cytotoxic, ER stress-inducing Ru(II)-arene Schiff-Base (RAS) complexes, bearing iminoquinoline chelate ligands. We demonstrate that by structural modification to the iminoquinoline ligand, we could tune its π-acidity and influence reactive oxygen species (ROS) induction, switching between a ROS-mediated ER stress pathway activation and one that is not mediated by ROS induction. Our current study adds to the available ER stress inducers and shows how structural tuning could be used as a means to modulate the mode-of-action of such compounds.
Collapse
Affiliation(s)
- Mun Juinn Chow
- Centre for Translational Medicine , Cancer Science Institute of Singapore, National University of Singapore , MD6 Centre for Translational Medicine, 14 Medical Drive , 117599 Singapore
| | - Maria V Babak
- Department of Chemistry , National University of Singapore , 3 Science Drive 3 , 117543 Singapore
| | - Kwan Wei Tan
- Department of Chemistry , National University of Singapore , 3 Science Drive 3 , 117543 Singapore
| | - Mei Chi Cheong
- Department of Chemistry , National University of Singapore , 3 Science Drive 3 , 117543 Singapore
| | - Giorgia Pastorin
- Department of Pharmacy , National University of Singapore , 3 Science Drive 3 , 117543 Singapore.,NUS Graduate School for Integrative Sciences and Engineering , 117456 Singapore
| | - Christian Gaiddon
- U1113 INSERM , 3 Avenue Molière , Strasbourg 67200 , France.,Oncology Section, FMTS , Université de Strasbourg , Strasbourg 67200 , France
| | - Wee Han Ang
- Department of Chemistry , National University of Singapore , 3 Science Drive 3 , 117543 Singapore.,NUS Graduate School for Integrative Sciences and Engineering , 117456 Singapore
| |
Collapse
|
19
|
Pal M, Nandi U, Mukherjee D. Detailed account on activation mechanisms of ruthenium coordination complexes and their role as antineoplastic agents. Eur J Med Chem 2018; 150:419-445. [DOI: 10.1016/j.ejmech.2018.03.015] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 03/02/2018] [Accepted: 03/03/2018] [Indexed: 10/17/2022]
|
20
|
Comparative studies on DNA-binding and in vitro antitumor activity of enantiomeric ruthenium(II) complexes. J Inorg Biochem 2018; 180:54-60. [DOI: 10.1016/j.jinorgbio.2017.11.024] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 10/31/2017] [Accepted: 11/26/2017] [Indexed: 02/07/2023]
|
21
|
Licona C, Spaety ME, Capuozzo A, Ali M, Santamaria R, Armant O, Delalande F, Van Dorsselaer A, Cianferani S, Spencer J, Pfeffer M, Mellitzer G, Gaiddon C. A ruthenium anticancer compound interacts with histones and impacts differently on epigenetic and death pathways compared to cisplatin. Oncotarget 2018; 8:2568-2584. [PMID: 27935863 PMCID: PMC5356824 DOI: 10.18632/oncotarget.13711] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 10/17/2016] [Indexed: 12/20/2022] Open
Abstract
Ruthenium complexes are considered as potential replacements for platinum compounds in oncotherapy. Their clinical development is handicapped by a lack of consensus on their mode of action. In this study, we identify three histones (H3.1, H2A, H2B) as possible targets for an anticancer redox organoruthenium compound (RDC11). Using purified histones, we confirmed an interaction between the ruthenium complex and histones that impacted on histone complex formation. A comparative study of the ruthenium complex versus cisplatin showed differential epigenetic modifications on histone H3 that correlated with differential expression of histone deacetylase (HDAC) genes. We then characterized the impact of these epigenetic modifications on signaling pathways employing a transcriptomic approach. Clustering analyses showed gene expression signatures specific for cisplatin (42%) and for the ruthenium complex (30%). Signaling pathway analyses pointed to specificities distinguishing the ruthenium complex from cisplatin. For instance, cisplatin triggered preferentially p53 and folate biosynthesis while the ruthenium complex induced endoplasmic reticulum stress and trans-sulfuration pathways. To further understand the role of HDACs in these regulations, we used suberanilohydroxamic acid (SAHA) and showed that it synergized with cisplatin cytotoxicity while antagonizing the ruthenium complex activity. This study provides critical information for the characterization of signaling pathways differentiating both compounds, in particular, by the identification of a non-DNA direct target for an organoruthenium complex.
Collapse
Affiliation(s)
- Cynthia Licona
- INSERM 1113, Molecular Signaling of the Cell Stress Response and Pathology, Université de Strasbourg, Section Oncologie FMTS, Strasbourg, France
| | - Marie-Elodie Spaety
- INSERM 1113, Molecular Signaling of the Cell Stress Response and Pathology, Université de Strasbourg, Section Oncologie FMTS, Strasbourg, France
| | - Antonelle Capuozzo
- INSERM 1113, Molecular Signaling of the Cell Stress Response and Pathology, Université de Strasbourg, Section Oncologie FMTS, Strasbourg, France.,Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Moussa Ali
- Institut of Chemistry, UMR7177 CNRS, Université de Strasbourg, Laboratory of Metal-Induced Synthesis, France
| | - Rita Santamaria
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Olivier Armant
- Karlsruhe Institute of Technology (KIT), Institute of Toxicology and Genetics (ITG), Germany
| | - Francois Delalande
- Institut Pluridisciplinaire Hubert Curien, Département Sciences Analytiques, Université de Strasbourg, France
| | - Alain Van Dorsselaer
- Institut Pluridisciplinaire Hubert Curien, Département Sciences Analytiques, Université de Strasbourg, France
| | - Sarah Cianferani
- Institut Pluridisciplinaire Hubert Curien, Département Sciences Analytiques, Université de Strasbourg, France
| | - John Spencer
- Department of Chemistry, School of Life Sciences, University of Sussex, Falmer, Brighton, East Sussex, UK
| | - Michel Pfeffer
- Institut of Chemistry, UMR7177 CNRS, Université de Strasbourg, Laboratory of Metal-Induced Synthesis, France
| | - Georg Mellitzer
- INSERM 1113, Molecular Signaling of the Cell Stress Response and Pathology, Université de Strasbourg, Section Oncologie FMTS, Strasbourg, France
| | - Christian Gaiddon
- INSERM 1113, Molecular Signaling of the Cell Stress Response and Pathology, Université de Strasbourg, Section Oncologie FMTS, Strasbourg, France
| |
Collapse
|
22
|
Novohradsky V, Yellol J, Stuchlikova O, Santana MD, Kostrhunova H, Yellol G, Kasparkova J, Bautista D, Ruiz J, Brabec V. Organoruthenium Complexes with C^N Ligands are Highly Potent Cytotoxic Agents that Act by a New Mechanism of Action. Chemistry 2017; 23:15294-15299. [DOI: 10.1002/chem.201703581] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Indexed: 01/19/2023]
Affiliation(s)
- Vojtech Novohradsky
- Institute of Biophysics; Czech Academy of Sciences; Kralovopolska 135 61265 Brno Czech Republic
| | - Jyoti Yellol
- Departamento de Química Inorgánica and Regional Campus of International Excellence “Campus Mare Nostrum”; Universidad de Murcia and Biomedical Research Institute of Murcia (IMIB-Arrixaca); 30071 Murcia Spain
| | - Olga Stuchlikova
- Institute of Biophysics; Czech Academy of Sciences; Kralovopolska 135 61265 Brno Czech Republic
- Department of Biophysics, Faculty of Science; Palacky University; 17. listopadu 12 77146 Olomouc Czech Republic
| | - María Dolores Santana
- Departamento de Química Inorgánica and Regional Campus of International Excellence “Campus Mare Nostrum”; Universidad de Murcia and Biomedical Research Institute of Murcia (IMIB-Arrixaca); 30071 Murcia Spain
| | - Hana Kostrhunova
- Institute of Biophysics; Czech Academy of Sciences; Kralovopolska 135 61265 Brno Czech Republic
| | - Gorakh Yellol
- Departamento de Química Inorgánica and Regional Campus of International Excellence “Campus Mare Nostrum”; Universidad de Murcia and Biomedical Research Institute of Murcia (IMIB-Arrixaca); 30071 Murcia Spain
| | - Jana Kasparkova
- Institute of Biophysics; Czech Academy of Sciences; Kralovopolska 135 61265 Brno Czech Republic
- Department of Biophysics, Faculty of Science; Palacky University; 17. listopadu 12 77146 Olomouc Czech Republic
| | | | - José Ruiz
- Departamento de Química Inorgánica and Regional Campus of International Excellence “Campus Mare Nostrum”; Universidad de Murcia and Biomedical Research Institute of Murcia (IMIB-Arrixaca); 30071 Murcia Spain
| | - Viktor Brabec
- Institute of Biophysics; Czech Academy of Sciences; Kralovopolska 135 61265 Brno Czech Republic
| |
Collapse
|
23
|
Ali M, Hamada A, Habbita H, Weckbach J, Orvain C, Gaiddon C, Pfeffer M. Trans-C versus Cis-C thermally induced isomerisation of a terpyridine adduct of cytotoxic cycloruthenated compound. J Organomet Chem 2017. [DOI: 10.1016/j.jorganchem.2017.05.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
24
|
Schoenhacker-Alte B, Mohr T, Pirker C, Kryeziu K, Kuhn PS, Buck A, Hofmann T, Gerner C, Hermann G, Koellensperger G, Keppler BK, Berger W, Heffeter P. Sensitivity towards the GRP78 inhibitor KP1339/IT-139 is characterized by apoptosis induction via caspase 8 upon disruption of ER homeostasis. Cancer Lett 2017; 404:79-88. [PMID: 28716523 DOI: 10.1016/j.canlet.2017.07.009] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 07/05/2017] [Accepted: 07/07/2017] [Indexed: 11/30/2022]
Abstract
The ruthenium drug and GRP78 inhibitor KP1339/IT-139 has already demonstrated promising anticancer activity in a phase I clinical trial. This study aimed to identify mechanisms underlying increased sensitivity to KP1339 treatment. Based on a screen utilizing 23 cell lines, a small panel was selected to compare KP1339-sensitive and low-responsive models. KP1339 sensitivity was neither based on differences in ruthenium accumulation, nor sensitivity to oxidative stress or constituents of KP1339 (ruthenium chloride and indazole). Subsequently, the biochemical response to KP1339 was analyzed using whole genome expression arrays indicating that, while sensitive cell lines were characterized by "response to chemical stimuli" and "regulation of cell death", low-responsive cells preferentially activated pathways controlling cell cycle, DNA repair, and metabolism. Cell culture experiments confirmed that, while low-responsive cells executed cell cycle arrest in G2 phase, pronounced apoptosis induction via activation of caspase 8 was found in sensitive cells. Cell death induction is based on a unique disruption of the ER homeostasis by depletion of key cellular chaperones including GRP78 in combination with enhanced KP1339-mediated protein damage.
Collapse
Affiliation(s)
- Beatrix Schoenhacker-Alte
- Institute of Inorganic Chemistry, University of Vienna, Waehringer Str. 42, A-1090, Vienna, Austria; Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center of the Medical University, Medical University of Vienna, Borschkegasse 8a, A-1090, Vienna, Austria
| | - Thomas Mohr
- Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center of the Medical University, Medical University of Vienna, Borschkegasse 8a, A-1090, Vienna, Austria
| | - Christine Pirker
- Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center of the Medical University, Medical University of Vienna, Borschkegasse 8a, A-1090, Vienna, Austria
| | - Kushtrim Kryeziu
- Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center of the Medical University, Medical University of Vienna, Borschkegasse 8a, A-1090, Vienna, Austria
| | - Paul-Steffen Kuhn
- Institute of Inorganic Chemistry, University of Vienna, Waehringer Str. 42, A-1090, Vienna, Austria
| | - Alicia Buck
- Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center of the Medical University, Medical University of Vienna, Borschkegasse 8a, A-1090, Vienna, Austria
| | - Thilo Hofmann
- Department of Environmental Geosciences, University of Vienna, Althanstraße 14, A-1090, Vienna, Austria
| | - Christopher Gerner
- Department of Analytical Chemistry, University of Vienna, Waehringer Str. 38, A-1090, Vienna, Austria
| | - Gerrit Hermann
- Department of Analytical Chemistry, University of Vienna, Waehringer Str. 38, A-1090, Vienna, Austria
| | - Gunda Koellensperger
- Department of Analytical Chemistry, University of Vienna, Waehringer Str. 38, A-1090, Vienna, Austria
| | - Bernhard K Keppler
- Institute of Inorganic Chemistry, University of Vienna, Waehringer Str. 42, A-1090, Vienna, Austria; Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center of the Medical University, Medical University of Vienna, Borschkegasse 8a, A-1090, Vienna, Austria
| | - Walter Berger
- Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center of the Medical University, Medical University of Vienna, Borschkegasse 8a, A-1090, Vienna, Austria; Research Cluster "Translational Cancer Therapy Research", Vienna, Austria
| | - Petra Heffeter
- Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center of the Medical University, Medical University of Vienna, Borschkegasse 8a, A-1090, Vienna, Austria; Research Cluster "Translational Cancer Therapy Research", Vienna, Austria.
| |
Collapse
|
25
|
Thangavel P, Viswanath B, Kim S. Recent developments in the nanostructured materials functionalized with ruthenium complexes for targeted drug delivery to tumors. Int J Nanomedicine 2017; 12:2749-2758. [PMID: 28435255 PMCID: PMC5388259 DOI: 10.2147/ijn.s131304] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
In recent years, the field of metal-based drugs has been dominated by other existing precious metal drugs, and many researchers have focused their attention on the synthesis of various ruthenium (Ru) complexes due to their potential medical and pharmaceutical applications. The beneficial properties of Ru, which make it a highly promising therapeutic agent, include its variable oxidation states, low toxicity, high selectivity for diseased cells, ligand exchange properties, and the ability to mimic iron binding to biomolecules. In addition, Ru complexes have favorable adsorption properties, along with excellent photochemical and photophysical properties, which make them promising tools for photodynamic therapy. At present, nanostructured materials functionalized with Ru complexes have become an efficient way to administer Ru-based anticancer drugs for cancer treatment. In this review, the recent developments in the nanostructured materials functionalized with Ru complexes for targeted drug delivery to tumors are discussed. In addition, information on "traditional" (ie, non-nanostructured) Ru-based cancer therapies is included in a precise manner.
Collapse
Affiliation(s)
- Prakash Thangavel
- Department of Bionanotechnology, Gachon University, Bokjeong-Dong, Sujeong-Gu, Seongnam-Si, Gyeonggi-Do
| | - Buddolla Viswanath
- Department of Bionanotechnology, Gachon University, Bokjeong-Dong, Sujeong-Gu, Seongnam-Si, Gyeonggi-Do
| | - Sanghyo Kim
- Department of Bionanotechnology, Gachon University, Bokjeong-Dong, Sujeong-Gu, Seongnam-Si, Gyeonggi-Do
- Graduate Gachon Medical Research Institute, Gil Medical Center, Incheon, Republic of Korea
| |
Collapse
|
26
|
Gaiddon C, Pfeffer M. The Fate of Cycloruthenated Compounds: From C-H Activation to Innovative Anticancer Therapy. Eur J Inorg Chem 2017. [DOI: 10.1002/ejic.201601216] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Christian Gaiddon
- University of Strasbourg; U1113 Inserm; 3 av. Molière 67200 Strasbourg France
| | - Michel Pfeffer
- University of Strasbourg; UMR 7177 CNRS; 4, rue Blaise Pascal 67000 Strasbourg France
| |
Collapse
|
27
|
Rico Bautista H, Saavedra Díaz RO, Shen LQ, Orvain C, Gaiddon C, Le Lagadec R, Ryabov AD. Impact of cyclometalated ruthenium(II) complexes on lactate dehydrogenase activity and cytotoxicity in gastric and colon cancer cells. J Inorg Biochem 2016; 163:28-38. [PMID: 27513948 DOI: 10.1016/j.jinorgbio.2016.07.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Revised: 07/11/2016] [Accepted: 07/25/2016] [Indexed: 11/15/2022]
Abstract
Lactate dehydrogenase (LDH) is a redox enzyme often overexpressed in cancer cells allowing their survival in stressful metabolic tumor environment. Ruthenium(II) complexes have been shown to impact on the activity of purified horseradish peroxidase and glucose oxidase but the physiological relevance remains unclear. In this study we investigated how ruthenium complexes impact on the activity of LDH in vitro and in cancer cells and performed a comparative study using polypyridine ruthenium(II) complex [Ru(bpy)3]2+ (1) and its structurally related cyclometalated 2-phenylpyridinato counterpart [Ru(phpy)(bpy)2]+ (2) (bpy=2,2'-bipyridine, phpyH=2-phenylpyridine). We show that the cytotoxicity in gastric and colon cancer cells induced by 2 is significantly higher compared to 1. The kinetic inhibition mechanisms on purified LDH and the corresponding inhibition constants Ki or i0.5 values were calculated. Though complexes 1 and 2 are structurally very similar (one Ru-C bond in 2 replaces one Ru-N bond in 1), their inhibition modes are different. Cyclometalated complex 2 behaves exclusively as a non-competitive inhibitor of LDH from rabbit muscle (LDHrm), strongly suggesting that 2 does not interact with LDH in the vicinities of either lactate/pyruvate or NAD+/NADH binding sites. Sites of interaction of 1 and 2 with LDHrm were revealed theoretically through computational molecular docking. Inhibition of LDH activity by 2 was confirmed in cancer cells. Altogether, these results revealed an inhibition of LDH activity by ruthenium complex through a direct interaction structurally tuned by a Ru-C bond.
Collapse
Affiliation(s)
- Hugo Rico Bautista
- Instituto de Química UNAM, Circuito Exterior s/n, Ciudad Universitaria, 04510 México D.F., Mexico
| | | | - Longzhu Q Shen
- Department of Chemistry, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, PA 15213, USA
| | - Christophe Orvain
- INSERM U1113, Strasbourg University, FMTS, 3 Avenue Molière, 67200 Strasbourg, France; Oncology Section, FMTS, Strasbourg University, Strasbourg, France
| | - Christian Gaiddon
- INSERM U1113, Strasbourg University, FMTS, 3 Avenue Molière, 67200 Strasbourg, France; Oncology Section, FMTS, Strasbourg University, Strasbourg, France
| | - Ronan Le Lagadec
- Instituto de Química UNAM, Circuito Exterior s/n, Ciudad Universitaria, 04510 México D.F., Mexico.
| | - Alexander D Ryabov
- Department of Chemistry, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, PA 15213, USA
| |
Collapse
|
28
|
Chow MJ, Licona C, Pastorin G, Mellitzer G, Ang WH, Gaiddon C. Structural tuning of organoruthenium compounds allows oxidative switch to control ER stress pathways and bypass multidrug resistance. Chem Sci 2016; 7:4117-4124. [PMID: 30155055 PMCID: PMC6013925 DOI: 10.1039/c6sc00268d] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 02/25/2016] [Indexed: 12/12/2022] Open
Abstract
Multidrug resistance (MDR) is a major impediment to the success of chemotherapy in many cancer types. One particular MDR mechanism is the inherent or acquired adaptation of the cellular survival pathways that render malignant cells resistant to apoptotic cell death. Since most drugs act through apoptosis, compounds capable of inducing alternative forms of programmed cell death (PCD) can potentially be harnessed to bypass MDR. We investigated two organoruthenium complexes, RAS-1H and RAS-1T, and demonstrated that although they both induced non-apoptotic PCD through ER stress pathways, their modes-of-action were drastically different despite modest structural variations. RAS-1T acted through ROS-mediated ER stress while RAS-1H was ROS-independent. We further showed that they were more efficacious against apoptosis-resistant cells compared to clinical drugs including oxaliplatin. This work provides the basis for underpinning ER stress modulation using metal complexes to bypass apoptosis resistance.
Collapse
Affiliation(s)
- Mun Juinn Chow
- Department of Chemistry , National University of Singapore , 3 Science Drive 3 , 117543 Singapore . ; Tel: +65 6516 5131
- NUS Graduate School for Integrative Sciences and Engineering , Singapore
| | - Cynthia Licona
- U1113 INSERM , 3 Avenue Molière , Strasbourg 67200 , France . ; Tel: +33 68 52 53 56
- Section Oncology , FMTS , Strasbourg University , Strasbourg , France
| | - Giorgia Pastorin
- NUS Graduate School for Integrative Sciences and Engineering , Singapore
- Department of Pharmacy , National University of Singapore , 18 Science Drive 4 , 117543 Singapore
| | - Georg Mellitzer
- U1113 INSERM , 3 Avenue Molière , Strasbourg 67200 , France . ; Tel: +33 68 52 53 56
- Section Oncology , FMTS , Strasbourg University , Strasbourg , France
| | - Wee Han Ang
- Department of Chemistry , National University of Singapore , 3 Science Drive 3 , 117543 Singapore . ; Tel: +65 6516 5131
- NUS Graduate School for Integrative Sciences and Engineering , Singapore
| | - Christian Gaiddon
- U1113 INSERM , 3 Avenue Molière , Strasbourg 67200 , France . ; Tel: +33 68 52 53 56
- Section Oncology , FMTS , Strasbourg University , Strasbourg , France
| |
Collapse
|
29
|
Huang Y, Huang W, Chan L, Zhou B, Chen T. A multifunctional DNA origami as carrier of metal complexes to achieve enhanced tumoral delivery and nullified systemic toxicity. Biomaterials 2016; 103:183-196. [PMID: 27388944 DOI: 10.1016/j.biomaterials.2016.06.053] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 06/17/2016] [Accepted: 06/21/2016] [Indexed: 12/20/2022]
Abstract
The use of metal complexes in cancer treatment is hampered by the insufficient accumulation in tumor regions and observable systemic toxicity due to their nonspecificity in vivo. Herein we present a cancer-targeted DNA origami as biocompatible nanocarrier of metal complexes to achieve advanced antitumor effect. The formation of unique tetrahedral nanostructure of DNA cages effectively enhances the interaction between ruthenium polypyridyl complexes (RuPOP) and the cages, thus increasing the drug loading efficacy. Conjugation of biotin to the DNA-based nanosystem (Bio-cage@Ru) enhances its specific cellular uptake, drug retention and cytotoxicity against HepG2 cells. Different from free RuPOP and the cage itself, Bio-cage@Ru translocates to cell nucleus after internalization, where it undergoes self-immolative cleavage in response to DNases, leading to triggered drug release and induction of ROS-mediated cell apoptosis. Moreover, in the nude mice model, the nanosystem specifically accumulates in tumor sites, thus exhibits satisfactory in vivo antitumor efficacy, and alleviates the damage of liver, kidney, lung and heart function of nude mice induced by RuPOP and tumor xenografts. Collectively, this study demonstrates a strategy for construction of biocompatible and cancer-targeted DNA origami with enhanced anticancer efficacy and reduced toxicity for next-generation cancer therapy.
Collapse
Affiliation(s)
- Yanyu Huang
- Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Wei Huang
- Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Leung Chan
- Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Binwei Zhou
- Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Tianfeng Chen
- Department of Chemistry, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
30
|
Chow MJ, Babak MV, Wong DYQ, Pastorin G, Gaiddon C, Ang WH. Structural Determinants of p53-Independence in Anticancer Ruthenium-Arene Schiff-Base Complexes. Mol Pharm 2016; 13:2543-54. [PMID: 27174050 DOI: 10.1021/acs.molpharmaceut.6b00348] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
p53 is a key tumor suppressor gene involved in key cellular processes and implicated in cancer therapy. However, it is inactivated in more than 50% of all cancers due to mutation or overexpression of its negative regulators. This leads to drug resistance and poor chemotherapeutic outcome as most clinical drugs act via a p53-dependent mechanism of action. An attractive strategy to circumvent this resistance would be to identify new anticancer drugs that act via p53-independent mode of action. In the present study, we identified 9 Ru (II)-Arene Schiff-base (RAS) complexes able to induce p53-independent cytotoxicity and discuss structural features that are required for their p53-independent activity. Increasing hydrophobicity led to an increase in cellular accumulation in cells with a corresponding increase in efficacy. We further showed that all nine complexes demonstrated p53-independent activity. This was despite significant differences in their physicochemical properties, suggesting that the iminoquinoline ligand, a common structural feature for all the complexes, is required for the p53-independent activity.
Collapse
Affiliation(s)
- Mun Juinn Chow
- Department of Chemistry, National University of Singapore , 3 Science Drive 3, 117543 Singapore.,NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore , 28 Medical Drive, 117456 Singapore
| | - Maria V Babak
- Department of Chemistry, National University of Singapore , 3 Science Drive 3, 117543 Singapore
| | - Daniel Yuan Qiang Wong
- Department of Chemistry, National University of Singapore , 3 Science Drive 3, 117543 Singapore
| | - Giorgia Pastorin
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore , 28 Medical Drive, 117456 Singapore.,Department of Pharmacy, National University of Singapore , 18 Science Drive 4, 117543 Singapore
| | - Christian Gaiddon
- U1113 INSERM, 3 Avenue Molière, Strasbourg 67200, France.,Oncology Section, FMTS, Université de Strasbourg , F-67081 Strasbourg, France
| | - Wee Han Ang
- Department of Chemistry, National University of Singapore , 3 Science Drive 3, 117543 Singapore.,NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore , 28 Medical Drive, 117456 Singapore
| |
Collapse
|
31
|
Tracking antitumor metallodrugs: promising agents with the Ru(II)- and Fe(II)-cyclopentadienyl scaffolds. Future Med Chem 2016; 8:527-44. [PMID: 27096164 DOI: 10.4155/fmc.16.7] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Research on the field of metal complexes for the treatment of cancer diseases has attracted increasing interest due to the urgency in finding more efficient and selective treatments. Owing to their wide structural diversity, organometallic complexes appear as potential alternatives to the design of new anticancer candidates. Herein, we review recent progress in our work toward the development of new drugs based on Ru(II)- and Fe(II)-cyclopentadienyl scaffolds. Their design and chemical properties are reviewed and correlated with their biological effects, in particular the key role that coligands play in the overall behavior of the complex.
Collapse
|
32
|
von Grabowiecki Y, Abreu P, Blanchard O, Palamiuc L, Benosman S, Mériaux S, Devignot V, Gross I, Mellitzer G, Gonzalez de Aguilar JL, Gaiddon C. Transcriptional activator TAp63 is upregulated in muscular atrophy during ALS and induces the pro-atrophic ubiquitin ligase Trim63. eLife 2016; 5. [PMID: 26919175 PMCID: PMC4786414 DOI: 10.7554/elife.10528] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 01/08/2016] [Indexed: 12/14/2022] Open
Abstract
Mechanisms of muscle atrophy are complex and their understanding might help finding therapeutic solutions for pathologies such as amyotrophic lateral sclerosis (ALS). We meta-analyzed transcriptomic experiments of muscles of ALS patients and mouse models, uncovering a p53 deregulation as common denominator. We then characterized the induction of several p53 family members (p53, p63, p73) and a correlation between the levels of p53 family target genes and the severity of muscle atrophy in ALS patients and mice. In particular, we observed increased p63 protein levels in the fibers of atrophic muscles via denervation-dependent and -independent mechanisms. At a functional level, we demonstrated that TAp63 and p53 transactivate the promoter and increased the expression of Trim63 (MuRF1), an effector of muscle atrophy. Altogether, these results suggest a novel function for p63 as a contributor to muscular atrophic processes via the regulation of multiple genes, including the muscle atrophy gene Trim63.
Collapse
Affiliation(s)
- Yannick von Grabowiecki
- UMR_S 1113, Molecular mechanisms of stress response and pathologies, Institut national de la santé et de la recherche médicale, Strasbourg, France.,Fédération de Recherche Translationnelle, Strasbourg University, Strasbourg, France
| | - Paula Abreu
- UMR_S 1113, Molecular mechanisms of stress response and pathologies, Institut national de la santé et de la recherche médicale, Strasbourg, France.,Fédération de Recherche Translationnelle, Strasbourg University, Strasbourg, France
| | - Orphee Blanchard
- UMR_S 1113, Molecular mechanisms of stress response and pathologies, Institut national de la santé et de la recherche médicale, Strasbourg, France.,Fédération de Recherche Translationnelle, Strasbourg University, Strasbourg, France
| | - Lavinia Palamiuc
- Fédération de Recherche Translationnelle, Strasbourg University, Strasbourg, France.,Sanford Burnham Medical Research Institute, San Diego, United States
| | - Samir Benosman
- Sanford Burnham Medical Research Institute, San Diego, United States
| | - Sophie Mériaux
- Fédération de Recherche Translationnelle, Strasbourg University, Strasbourg, France.,Sanford Burnham Medical Research Institute, San Diego, United States
| | - Véronique Devignot
- UMR_S 1113, Molecular mechanisms of stress response and pathologies, Institut national de la santé et de la recherche médicale, Strasbourg, France.,Fédération de Recherche Translationnelle, Strasbourg University, Strasbourg, France
| | - Isabelle Gross
- UMR_S 1113, Molecular mechanisms of stress response and pathologies, Institut national de la santé et de la recherche médicale, Strasbourg, France.,Fédération de Recherche Translationnelle, Strasbourg University, Strasbourg, France
| | - Georg Mellitzer
- UMR_S 1113, Molecular mechanisms of stress response and pathologies, Institut national de la santé et de la recherche médicale, Strasbourg, France.,Fédération de Recherche Translationnelle, Strasbourg University, Strasbourg, France
| | - José L Gonzalez de Aguilar
- Fédération de Recherche Translationnelle, Strasbourg University, Strasbourg, France.,Institut national de la santé et de la recherche médicale, Laboratoire SMN, Strasbourg, France
| | - Christian Gaiddon
- UMR_S 1113, Molecular mechanisms of stress response and pathologies, Institut national de la santé et de la recherche médicale, Strasbourg, France.,Fédération de Recherche Translationnelle, Strasbourg University, Strasbourg, France
| |
Collapse
|
33
|
Nikolić S, Rangasamy L, Gligorijević N, Aranđelović S, Radulović S, Gasser G, Grgurić-Šipka S. Synthesis, characterization and biological evaluation of novel Ru(II)-arene complexes containing intercalating ligands. J Inorg Biochem 2016; 160:156-65. [PMID: 26818702 DOI: 10.1016/j.jinorgbio.2016.01.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 12/30/2015] [Accepted: 01/07/2016] [Indexed: 01/20/2023]
Abstract
Three new ruthenium(II)-arene complexes, namely [(η(6)-p-cymene)Ru(Me2dppz)Cl]PF6 (1), [(η(6)-benzene)Ru(Me2dppz)Cl]PF6 (2) and [(η(6)-p-cymene)Ru(aip)Cl]PF6 (3) (Me2dppz=11,12-dimethyldipyrido[3,2-a:2',3'-c]phenazine; aip=2-(9-anthryl)-1H-imidazo[4,5-f] [1,10] phenanthroline) have been synthesized and characterized using different spectroscopic techniques including elemental analysis. The complexes were found to be well soluble and stable in DMSO. The biological activity of the three complexes was tested in three different human cancer cell lines (A549, MDA-MB-231 and HeLa) and in one human non-cancerous cell line (MRC-5). Complexes 1 and 3, carrying η(6)-p-cymene as the arene ligand, were shown to be toxic in all cell lines in the low micromolar/subnanomolar range, with complex 1 being the most cytotoxic complex of the series. Flow cytometry analysis revealed that complex 1 caused concentration- and time-dependent arrest of the cell cycle in G2-M and S phases in HeLa cells. This event is followed by the accumulation of the sub-G1 DNA content after 48h, in levels higher than cisplatin and in the absence of phosphatidylserine externalization. Fluorescent microscopy and acridine orange/ethidium bromide staining revealed that complex 1 induced both apoptotic and necrotic cell morphology characteristics. Drug-accumulation and DNA-binding studies performed by inductively coupled plasma mass spectrometry in HeLa cells showed that the total ruthenium uptake increased in a time- and concentration-dependent manner, and that complex 1 accumulated more efficiently than cisplatin at equimolar concentrations. The introduction of a Me2dppz ligand into the ruthenium(II)-p-cymene scaffold was found to allow the discovery of a strongly cytotoxic complex with significantly higher cellular uptake and DNA-binding properties than cisplatin.
Collapse
Affiliation(s)
- Stefan Nikolić
- Faculty of Chemistry, University of Belgrade, Studentski trg 12-16, 11000 Belgrade, Serbia
| | - Loganathan Rangasamy
- Department of Chemistry, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | - Nevenka Gligorijević
- Institute for Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia
| | - Sandra Aranđelović
- Institute for Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia
| | - Siniša Radulović
- Institute for Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia
| | - Gilles Gasser
- Department of Chemistry, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland.
| | - Sanja Grgurić-Šipka
- Faculty of Chemistry, University of Belgrade, Studentski trg 12-16, 11000 Belgrade, Serbia.
| |
Collapse
|
34
|
Barbosa ASL, Werlé C, Colunga COO, Rodríguez CF, Toscano RA, Le Lagadec R, Pfeffer M. Further Insight into the Lability of MeCN Ligands of Cytotoxic Cycloruthenated Compounds: Evidence for the Antisymbiotic Effect Trans to the Carbon Atom at the Ru Center. Inorg Chem 2015; 54:7617-26. [DOI: 10.1021/acs.inorgchem.5b01236] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Ana Soraya Lima Barbosa
- Laboratoire de Chimie & Systémique Organo-Métallique of the Institut de Chimie, UMR 7177, CNRS, Université de Strasbourg, 4, rue Blaise Pascal, 67000 Strasbourg, France
| | - Christophe Werlé
- Laboratoire de Chimie & Systémique Organo-Métallique of the Institut de Chimie, UMR 7177, CNRS, Université de Strasbourg, 4, rue Blaise Pascal, 67000 Strasbourg, France
| | | | - Cecilia Franco Rodríguez
- Instituto de Química, UNAM, Circuito Exterior s/n, Ciudad Universitaria, Mexico, D. F. 04510, Mexico
| | - Ruben Alfredo Toscano
- Instituto de Química, UNAM, Circuito Exterior s/n, Ciudad Universitaria, Mexico, D. F. 04510, Mexico
| | - Ronan Le Lagadec
- Instituto de Química, UNAM, Circuito Exterior s/n, Ciudad Universitaria, Mexico, D. F. 04510, Mexico
| | - Michel Pfeffer
- Laboratoire de Chimie & Systémique Organo-Métallique of the Institut de Chimie, UMR 7177, CNRS, Université de Strasbourg, 4, rue Blaise Pascal, 67000 Strasbourg, France
| |
Collapse
|
35
|
Ali M, Dondaine L, Adolle A, Sampaio C, Chotard F, Richard P, Denat F, Bettaieb A, Le Gendre P, Laurens V, Goze C, Paul C, Bodio E. Anticancer Agents: Does a Phosphonium Behave Like a Gold(I) Phosphine Complex? Let a “Smart” Probe Answer! J Med Chem 2015; 58:4521-8. [DOI: 10.1021/acs.jmedchem.5b00480] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Moussa Ali
- Institut de Chimie Moléculaire de l’Université de Bourgogne, ICMUB UMR CNRS 6302, 9 Avenue Alain Savary, BP 47870, Dijon Cedex, 21078, France
| | - Lucile Dondaine
- École Pratique des Hautes Études, Paris, F-75014, France
- EA7269 EPHE-University of Burgundy, University of Burgundy, Dijon, F-21000, France
| | - Anais Adolle
- École Pratique des Hautes Études, Paris, F-75014, France
- EA7269 EPHE-University of Burgundy, University of Burgundy, Dijon, F-21000, France
| | - Carla Sampaio
- École Pratique des Hautes Études, Paris, F-75014, France
- EA7269 EPHE-University of Burgundy, University of Burgundy, Dijon, F-21000, France
| | - Florian Chotard
- Institut de Chimie Moléculaire de l’Université de Bourgogne, ICMUB UMR CNRS 6302, 9 Avenue Alain Savary, BP 47870, Dijon Cedex, 21078, France
| | - Philippe Richard
- Institut de Chimie Moléculaire de l’Université de Bourgogne, ICMUB UMR CNRS 6302, 9 Avenue Alain Savary, BP 47870, Dijon Cedex, 21078, France
| | - Franck Denat
- Institut de Chimie Moléculaire de l’Université de Bourgogne, ICMUB UMR CNRS 6302, 9 Avenue Alain Savary, BP 47870, Dijon Cedex, 21078, France
| | - Ali Bettaieb
- École Pratique des Hautes Études, Paris, F-75014, France
- EA7269 EPHE-University of Burgundy, University of Burgundy, Dijon, F-21000, France
| | - Pierre Le Gendre
- Institut de Chimie Moléculaire de l’Université de Bourgogne, ICMUB UMR CNRS 6302, 9 Avenue Alain Savary, BP 47870, Dijon Cedex, 21078, France
| | - Véronique Laurens
- École Pratique des Hautes Études, Paris, F-75014, France
- EA7269 EPHE-University of Burgundy, University of Burgundy, Dijon, F-21000, France
| | - Christine Goze
- Institut de Chimie Moléculaire de l’Université de Bourgogne, ICMUB UMR CNRS 6302, 9 Avenue Alain Savary, BP 47870, Dijon Cedex, 21078, France
| | - Catherine Paul
- École Pratique des Hautes Études, Paris, F-75014, France
- EA7269 EPHE-University of Burgundy, University of Burgundy, Dijon, F-21000, France
| | - Ewen Bodio
- Institut de Chimie Moléculaire de l’Université de Bourgogne, ICMUB UMR CNRS 6302, 9 Avenue Alain Savary, BP 47870, Dijon Cedex, 21078, France
| |
Collapse
|
36
|
Jia F, Despax S, Münch JP, Hébraud P. Flexibility of short ds-DNA intercalated by a dipyridophenazine ligand. Front Chem 2015; 3:25. [PMID: 25932461 PMCID: PMC4399336 DOI: 10.3389/fchem.2015.00025] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 03/20/2015] [Indexed: 11/13/2022] Open
Abstract
We use Förster Resonant Energy Transfer (FRET) in order to measure the increase of flexibility of short ds-DNA induced by the intercalation of dipyridophenazine (dppz) ligand in between DNA base pairs. By using a DNA double strand fluorescently labeled at its extremities, it is shown that the end-to-end length increase of DNA due to the intercalation of one dppz ligand is smaller than the DNA base pair interdistance. This may be explained either by a local bending of the DNA or by an increase of its flexibility. The persistence length of the formed DNA/ligand is evaluated. The described structure may have implications in the photophysical damages induced by the complexation of DNA by organometallic molecules.
Collapse
Affiliation(s)
- Fuchao Jia
- Institut de Physique et Chimie des Matériaux de Strasbourg/Centre National de la Recherche Scientifique, University of Strasbourg Strasbourg, France ; Department of Physics, School of Science, Shangdong University Zibo, China
| | - Stéphane Despax
- Institut de Physique et Chimie des Matériaux de Strasbourg/Centre National de la Recherche Scientifique, University of Strasbourg Strasbourg, France
| | - Jean-Pierre Münch
- Institut de Physique et Chimie des Matériaux de Strasbourg/Centre National de la Recherche Scientifique, University of Strasbourg Strasbourg, France
| | - Pascal Hébraud
- Institut de Physique et Chimie des Matériaux de Strasbourg/Centre National de la Recherche Scientifique, University of Strasbourg Strasbourg, France
| |
Collapse
|
37
|
Abstract
Ruthenium compounds are highly regarded as potential drug candidates. The compounds offer the potential of reduced toxicity and can be tolerated in vivo. The various oxidation states, different mechanism of action, and the ligand substitution kinetics of ruthenium compounds give them advantages over platinum-based complexes, thereby making them suitable for use in biological applications. Several studies have focused attention on the interaction between active ruthenium complexes and their possible biological targets. In this paper, we review several ruthenium compounds which reportedly possess promising cytotoxic profiles: from the discovery of highly active compounds imidazolium [trans-tetrachloro(dmso)(imidazole)ruthenate(III)] (NAMI-A), indazolium [trans-tetrachlorobis(1H-indazole)ruthenate(III)](KP1019), and sodium trans-[tetrachloridobis(1H-indazole)ruthenate(III)] (NKP-1339) to the recent work based on both inorganic and organometallic ruthenium(II) compounds. Half-sandwich organometallic ruthenium complexes offer the opportunity of derivatization at the arene moiety, while the three remaining coordination sites on the metal centre can be functionalised with various coordination groups of various monoligands. It is clear from the review that these mononuclear ruthenium(II) compounds represent a strongly emerging field of research that will soon culminate into several ruthenium based antitumor agents.
Collapse
|
38
|
Gold-phosphine-porphyrin as potential metal-based theranostics. J Biol Inorg Chem 2015; 20:143-154. [PMID: 25476859 DOI: 10.1007/s00775-014-1220-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 11/18/2014] [Indexed: 01/12/2023]
Abstract
Two new gold-phosphine-porphyrin derivatives were synthesized and fully characterized, and their photophysical properties investigated along a water-soluble analog. The cytotoxicity of the compounds was tested on cancer cells (HCT116 and SW480), and their cell uptake was followed by fluorescence microscopy in vitro (on SW480). The proof that the water-soluble gold-phosphine-porphyrin is a biologically active compound that can be tracked in vitro was clearly established, especially concerning the water-soluble analog. Some preliminary photodynamic therapy (PDT) experiments were also performed. They highlight a dramatic increase of the cytotoxicity when the cells were illuminated for 30 min with white light.
Collapse
|
39
|
Huang H, Zhang P, Chen Y, Ji L, Chao H. Labile ruthenium(ii) complexes with extended phenyl-substituted terpyridyl ligands: synthesis, aquation and anticancer evaluation. Dalton Trans 2015; 44:15602-10. [DOI: 10.1039/c5dt02446c] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The present study demonstrated that the anticancer activities of labile Ru(ii) complexes can be efficiently tuned by chelating with different phenyl-substituted terpyridyl ligands.
Collapse
Affiliation(s)
- Huaiyi Huang
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry
- School of Chemistry and Chemical Engineering
- Sun Yat-Sen University
- Guangzhou
- P. R. China
| | - Pingyu Zhang
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry
- School of Chemistry and Chemical Engineering
- Sun Yat-Sen University
- Guangzhou
- P. R. China
| | - Yu Chen
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry
- School of Chemistry and Chemical Engineering
- Sun Yat-Sen University
- Guangzhou
- P. R. China
| | - Liangnian Ji
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry
- School of Chemistry and Chemical Engineering
- Sun Yat-Sen University
- Guangzhou
- P. R. China
| | - Hui Chao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry
- School of Chemistry and Chemical Engineering
- Sun Yat-Sen University
- Guangzhou
- P. R. China
| |
Collapse
|
40
|
Huang H, Zhang P, Chen H, Ji L, Chao H. Comparison between polypyridyl and cyclometalated ruthenium(II) complexes: anticancer activities against 2D and 3D cancer models. Chemistry 2014; 21:715-25. [PMID: 25388328 DOI: 10.1002/chem.201404922] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Indexed: 12/17/2022]
Abstract
The aim of this study was to illustrate the dramatically different anticancer activities between coordinatively saturated polypyridyl (1 a-4 a) and cyclometalated (1 b-4 b) ruthenium(II) complexes. The cyclometalated complexes 1 b-4 b function as DNA transcription inhibitors, exhibiting switch-on cytotoxicity against a 2D cancer cell monolayer, whereas the polypyridyl complexes 1 a-4 a are relatively inactive. Moreover, complexes 1 b-4 b exhibit excellent cytotoxicity against 3D multicellular tumor spheroids (MCTSs), which serve as an intermediate model between in vitro 2D cell monolayers and in vivo 3D solid tumors. The hydrophobicity, efficient cell uptake, and nucleus targeting ability, as well as the high DNA binding affinity of complexes 1 b-4 b, likely contribute to their enhanced anticancer activity. We surmise that cyclometalation could be a universal approach to significantly enhance the anticancer activity of substituted polypyridyl Ru(II) complexes. We also suggest that 3D MCTSs may be a more practical platform for anticancer drug screening than 2D cancer monolayer approaches.
Collapse
Affiliation(s)
- Huaiyi Huang
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, State Key Laboratory of Optoelectronic Materials and Technologies, School of Chemistry and Chemical Engineering, Sun Yat-Sen University, Guangzhou 510275 (P. R. China)
| | | | | | | | | |
Collapse
|
41
|
Huang H, Zhang P, Yu B, Chen Y, Wang J, Ji L, Chao H. Targeting Nucleus DNA with a Cyclometalated Dipyridophenazineruthenium(II) Complex. J Med Chem 2014; 57:8971-83. [DOI: 10.1021/jm501095r] [Citation(s) in RCA: 187] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Huaiyi Huang
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, State Key Laboratory of Optoelectronic Materials and Technologies, School of Chemistry and Chemical Engineering, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Pingyu Zhang
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, State Key Laboratory of Optoelectronic Materials and Technologies, School of Chemistry and Chemical Engineering, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Bole Yu
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, State Key Laboratory of Optoelectronic Materials and Technologies, School of Chemistry and Chemical Engineering, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Yu Chen
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, State Key Laboratory of Optoelectronic Materials and Technologies, School of Chemistry and Chemical Engineering, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Jinquan Wang
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, State Key Laboratory of Optoelectronic Materials and Technologies, School of Chemistry and Chemical Engineering, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Liangnian Ji
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, State Key Laboratory of Optoelectronic Materials and Technologies, School of Chemistry and Chemical Engineering, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Hui Chao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, State Key Laboratory of Optoelectronic Materials and Technologies, School of Chemistry and Chemical Engineering, Sun Yat-Sen University, Guangzhou, 510275, China
| |
Collapse
|
42
|
von Grabowiecki Y, Licona C, Palamiuc L, Abreu P, Vidimar V, Coowar D, Mellitzer G, Gaiddon C. Regulation of a Notch3-Hes1 pathway and protective effect by a tocopherol-omega alkanol chain derivative in muscle atrophy. J Pharmacol Exp Ther 2014; 352:23-32. [PMID: 25326132 DOI: 10.1124/jpet.114.216879] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Muscular atrophy, a physiopathologic process associated with severe human diseases such as amyotrophic lateral sclerosis (ALS) or cancer, has been linked to reactive oxygen species (ROS) production. The Notch pathway plays a role in muscle development and in muscle regeneration upon physical injury. In this study, we explored the possibility that the Notch pathway participates in the ROS-related muscular atrophy occurring in cancer-associated cachexia and ALS. We also tested whether hybrid compounds of tocopherol, harboring antioxidant activity, and the omega-alkanol chain, presenting cytoprotective activity, might reduce muscle atrophy and impact the Notch pathway. We identified one tocopherol-omega alkanol chain derivative, AGT251, protecting myoblastic cells against known cytotoxic agents. We showed that this compound presenting antioxidant activity counteracts the induction of the Notch pathway by cytotoxic stress, leading to a decrease of Notch1 and Notch3 expression. At the functional level, these regulations correlated with a repression of the Notch target gene Hes1 and the atrophy/remodeling gene MuRF1. Importantly, we also observed an induction of Notch3 and Hes1 expression in two murine models of muscle atrophy: a doxorubicin-induced cachexia model and an ALS murine model expressing mutated superoxide dismutase 1. In both models, the induction of Notch3 and Hes1 were partially opposed by AGT251, which correlated with ameliorations in body and muscle weight, reduction of muscular atrophy markers, and improved survival. Altogether, we identified a compound of the tocopherol family that protects against muscle atrophy in various models, possibly through the regulation of the Notch pathway.
Collapse
Affiliation(s)
- Yannick von Grabowiecki
- INSERM U1113, Molecular Mechanisms of Stress Response and Pathologies, Strasbourg, France (Y.v.G., C.L., P.A., V.V., G.M., C.G.); Faculté de Médecine de Strasbourg, Strasbourg University, Strasbourg, France (Y.v.G., C.L., L.P., P.A., V.V., G.M., C.G.); and AxoGlia Therapeutics, Fentange, Luxembourg (D.C.)
| | - Cynthia Licona
- INSERM U1113, Molecular Mechanisms of Stress Response and Pathologies, Strasbourg, France (Y.v.G., C.L., P.A., V.V., G.M., C.G.); Faculté de Médecine de Strasbourg, Strasbourg University, Strasbourg, France (Y.v.G., C.L., L.P., P.A., V.V., G.M., C.G.); and AxoGlia Therapeutics, Fentange, Luxembourg (D.C.)
| | - Lavinia Palamiuc
- INSERM U1113, Molecular Mechanisms of Stress Response and Pathologies, Strasbourg, France (Y.v.G., C.L., P.A., V.V., G.M., C.G.); Faculté de Médecine de Strasbourg, Strasbourg University, Strasbourg, France (Y.v.G., C.L., L.P., P.A., V.V., G.M., C.G.); and AxoGlia Therapeutics, Fentange, Luxembourg (D.C.)
| | - Paula Abreu
- INSERM U1113, Molecular Mechanisms of Stress Response and Pathologies, Strasbourg, France (Y.v.G., C.L., P.A., V.V., G.M., C.G.); Faculté de Médecine de Strasbourg, Strasbourg University, Strasbourg, France (Y.v.G., C.L., L.P., P.A., V.V., G.M., C.G.); and AxoGlia Therapeutics, Fentange, Luxembourg (D.C.)
| | - Vania Vidimar
- INSERM U1113, Molecular Mechanisms of Stress Response and Pathologies, Strasbourg, France (Y.v.G., C.L., P.A., V.V., G.M., C.G.); Faculté de Médecine de Strasbourg, Strasbourg University, Strasbourg, France (Y.v.G., C.L., L.P., P.A., V.V., G.M., C.G.); and AxoGlia Therapeutics, Fentange, Luxembourg (D.C.)
| | - Djalil Coowar
- INSERM U1113, Molecular Mechanisms of Stress Response and Pathologies, Strasbourg, France (Y.v.G., C.L., P.A., V.V., G.M., C.G.); Faculté de Médecine de Strasbourg, Strasbourg University, Strasbourg, France (Y.v.G., C.L., L.P., P.A., V.V., G.M., C.G.); and AxoGlia Therapeutics, Fentange, Luxembourg (D.C.)
| | - Georg Mellitzer
- INSERM U1113, Molecular Mechanisms of Stress Response and Pathologies, Strasbourg, France (Y.v.G., C.L., P.A., V.V., G.M., C.G.); Faculté de Médecine de Strasbourg, Strasbourg University, Strasbourg, France (Y.v.G., C.L., L.P., P.A., V.V., G.M., C.G.); and AxoGlia Therapeutics, Fentange, Luxembourg (D.C.)
| | - Christian Gaiddon
- INSERM U1113, Molecular Mechanisms of Stress Response and Pathologies, Strasbourg, France (Y.v.G., C.L., P.A., V.V., G.M., C.G.); Faculté de Médecine de Strasbourg, Strasbourg University, Strasbourg, France (Y.v.G., C.L., L.P., P.A., V.V., G.M., C.G.); and AxoGlia Therapeutics, Fentange, Luxembourg (D.C.)
| |
Collapse
|
43
|
Chow MJ, Licona C, Yuan Qiang Wong D, Pastorin G, Gaiddon C, Ang WH. Discovery and investigation of anticancer ruthenium-arene Schiff-base complexes via water-promoted combinatorial three-component assembly. J Med Chem 2014; 57:6043-59. [PMID: 25023617 DOI: 10.1021/jm500455p] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The structural diversity of metal scaffolds makes them a viable alternative to traditional organic scaffolds for drug design. Combinatorial chemistry and multicomponent reactions, coupled with high-throughput screening, are useful techniques in drug discovery, but they are rarely used in metal-based drug design. We report the optimization and validation of a new combinatorial, metal-based, three-component assembly reaction for the synthesis of a library of 442 Ru-arene Schiff-base (RAS) complexes. These RAS complexes were synthesized in a one-pot, on-a-plate format using commercially available starting materials under aqueous conditions. The library was screened for their anticancer activity, and several cytotoxic lead compounds were identified. In particular, [(η6-1,3,5-triisopropylbenzene)RuCl(4-methoxy-N-(2-quinolinylmethylene)aniline)]Cl (4) displayed low micromolar IC50 values in ovarian cancers (A2780, A2780cisR), breast cancer (MCF7), and colorectal cancer (HCT116, SW480). The absence of p53 activation or changes in IC50 value between p53+/+ and p53-/- cells suggests that 4 and possibly the other lead compounds may act independently of the p53 tumor suppressor gene frequently mutated in cancer.
Collapse
Affiliation(s)
- Mun Juinn Chow
- Department of Chemistry, National University of Singapore , 3 Science Drive 3, 117543 Singapore
| | | | | | | | | | | |
Collapse
|
44
|
Hanif M, Babak MV, Hartinger CG. Development of anticancer agents: wizardry with osmium. Drug Discov Today 2014; 19:1640-8. [PMID: 24955838 DOI: 10.1016/j.drudis.2014.06.016] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 04/22/2014] [Accepted: 06/16/2014] [Indexed: 12/20/2022]
Abstract
Platinum compounds are one of the pillars of modern cancer chemotherapy. The apparent disadvantages of existing chemotherapeutics have led to the development of novel anticancer agents with alternative modes of action. Many complexes of the heavy metal osmium (Os) are potent growth inhibitors of human cancer cells and are active in vivo, often superior or comparable to cisplatin, as the benchmark metal-based anticancer agent, or clinically tested ruthenium (Ru) drug candidates. Depending on the choice of ligand system, osmium compounds exhibit diverse modes of action, including redox activation, DNA targeting or inhibition of protein kinases. In this review, we highlight recent advances in the development of osmium anticancer drug candidates and discuss their cellular mechanisms of action.
Collapse
Affiliation(s)
- Muhammad Hanif
- School of Chemical Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.
| | - Maria V Babak
- School of Chemical Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Christian G Hartinger
- School of Chemical Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.
| |
Collapse
|
45
|
Finck S, Issenhuth JT, Despax S, Sirlin C, Pfeffer M, Poidevin C, Gourlaouen C, Boeglin A, Daniel C. Structural and optical properties of new cyclometalated Ru(II) derived compounds. J Organomet Chem 2014. [DOI: 10.1016/j.jorganchem.2013.08.032] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
46
|
Lainé AL, Clavreul A, Rousseau A, Tétaud C, Vessieres A, Garcion E, Jaouen G, Aubert L, Guilbert M, Benoit JP, Toillon RA, Passirani C. Inhibition of ectopic glioma tumor growth by a potent ferrocenyl drug loaded into stealth lipid nanocapsules. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2014; 10:1667-77. [PMID: 24842766 DOI: 10.1016/j.nano.2014.05.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Revised: 03/28/2014] [Accepted: 05/03/2014] [Indexed: 10/25/2022]
Abstract
UNLABELLED In this work, a novel ferrocenyl complex (ansa-FcdiOH) was assessed for brain tumor therapy through stealth lipid nanocapsules (LNCs). Stealth LNCs, prepared according to a one-step process, showed rapid uptake by cancer cells and extended blood circulation time. The ferrocenyl complex was successfully encapsulated into these LNCs measuring 40 nm with a high loading capacity (6.4%). In vitro studies showed a potent anticancer effect of ansa-FcdiOH on 9L cells with a low IC50 value (0.1 μM) associated with an oxidative stress and a dose-dependent alteration of the cell cycle. Repeated intravenous injections of stealth ansa-FcdiOH LNCs in ectopic glioma bearing rats induced a significant tumor growth inhibition, supported by a reduced number of proliferative cells in tumors compared to control group. Additionally, no liver damage was observed in treated animals. These results indicated that stealth ansa-FcdiOH LNCs might be considered as a potential new approach for cancer chemotherapy. FROM THE CLINICAL EDITOR In this study, a novel ferrocenyl complex was assessed for brain tumor therapy through stealth lipid nanocapsules, demonstrating no liver damage, and superior tumor volume reduction compared to saline and stealth lipid nanocapsules alone in an ectopic glioma model.
Collapse
Affiliation(s)
- Anne-Laure Lainé
- LUNAM Université - Micro et Nanomédecines Biomimétiques, Angers, France; Inserm U1066, IBS-CHU, Angers, France
| | - Anne Clavreul
- Inserm U1066, IBS-CHU, Angers, France; Département de Neurochirurgie - CHU, Angers, France
| | - Audrey Rousseau
- Département de Pathologie Cellulaire et Tissulaire - CHU, Angers, France
| | - Clément Tétaud
- Inserm U1066, IBS-CHU, Angers, France; Département de Neurochirurgie - CHU, Angers, France
| | | | - Emmanuel Garcion
- LUNAM Université - Micro et Nanomédecines Biomimétiques, Angers, France; Inserm U1066, IBS-CHU, Angers, France
| | | | - Léo Aubert
- Inserm U908, Université Lille 1, Villeneuve d'Ascq, France
| | | | - Jean-Pierre Benoit
- LUNAM Université - Micro et Nanomédecines Biomimétiques, Angers, France; Inserm U1066, IBS-CHU, Angers, France
| | | | - Catherine Passirani
- LUNAM Université - Micro et Nanomédecines Biomimétiques, Angers, France; Inserm U1066, IBS-CHU, Angers, France.
| |
Collapse
|
47
|
Klajner M, Licona C, Fetzer L, Hebraud P, Mellitzer G, Pfeffer M, Harlepp S, Gaiddon C. Subcellular localization and transport kinetics of ruthenium organometallic anticancer compounds in living cells: a dose-dependent role for amino acid and iron transporters. Inorg Chem 2014; 53:5150-8. [PMID: 24786362 DOI: 10.1021/ic500250e] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Ruthenium-based compounds are developed for anticancer treatment, but their mode of action including their import mechanism and subcellular localization remains elusive. Here, we used the intrinsic luminescent properties of cytotoxic organoruthenium (Ru(II)) compounds obtained with an anionic cyclometalated 2-phenylpyridine chelate and neutral aromatic chelating ligands (e.g., phenanthrolines) to follow their behavior in living cells. We established that the difference in sensitivity between cancer cells and noncancerous cells toward one of the compounds correlates with its import kinetics and follows a balance between active and passive transport. The active-transport mechanism involves iron and amino-acid transporters, which are transcriptionally regulated by the drug. We also demonstrated a correlation between the accumulation of these compounds in specific compartments (endoplasmic reticulum, nucleus, mitochondria) and the activation of specific cytotoxic mechanisms such as the mitochondrial stress pathway. Our study pinpoints a novel and complex mechanism of accumulation of ruthenium drugs in cancer cells.
Collapse
Affiliation(s)
- M Klajner
- UMR7504, I.P.C.M.S. , 23 rue du Loess, 67200, Strasbourg, France
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Belsa L, López C, González A, Font-Bardı́a M, Calvet T, Calvis C, Messeguer R. Neutral and Ionic Cycloruthenated 2-Phenylindoles as Cytotoxic Agents. Organometallics 2013. [DOI: 10.1021/om400941b] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Lluís Belsa
- Departament de Quı́mica
Inorgànica,
Facultat de Quı́mica, Universitat de Barcelona, Martí
i Franquès 1-11, E-08028 Barcelona, Spain
| | - Concepción López
- Departament de Quı́mica
Inorgànica,
Facultat de Quı́mica, Universitat de Barcelona, Martí
i Franquès 1-11, E-08028 Barcelona, Spain
| | - Asensio González
- Laboratori de Quı́mica Orgànica,
Facultat de Farmàcia, Universitat de Barcelona, Pl. Pius
XII s/n, E-08028 Barcelona, Spain
| | - Mercè Font-Bardı́a
- Unitat
de Difracció de Raig-X, Centre Científic i Tecnològic de la Universitat de Barcelona, Solé i Sabarı́s 1-3, E-08028 Barcelona, Spain
| | - Teresa Calvet
- Departament de Crystal·lografı́a,
Mineralogia i Dipòsits Minerals, Facultat de Geologı́a, Universitat de Barcelona, Martı́ i Franquès s/n, E-08028 Barcelona, Spain
| | - Carmen Calvis
- Biomed Division, LEITAT Tecnological Center, Parc Científic de Barcelona, Edifici Hèlix, Baldiri Reixach
15-21, E-08028 Barcelona, Spain
| | - Ramon Messeguer
- Biomed Division, LEITAT Tecnological Center, Parc Científic de Barcelona, Edifici Hèlix, Baldiri Reixach
15-21, E-08028 Barcelona, Spain
| |
Collapse
|
49
|
A ruthenium(II) β-carboline complex induced p53-mediated apoptosis in cancer cells. Biochimie 2013; 95:2050-9. [DOI: 10.1016/j.biochi.2013.07.016] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2013] [Accepted: 07/16/2013] [Indexed: 01/15/2023]
|
50
|
Synthesis, characterization, and anticancer activity of ruthenium(II)-β-carboline complex. Eur J Med Chem 2013; 70:120-9. [PMID: 24141202 DOI: 10.1016/j.ejmech.2013.09.051] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Revised: 09/09/2013] [Accepted: 09/29/2013] [Indexed: 01/19/2023]
Abstract
Four [Ru(tpy)(N-N)(L)] type complexes: [Ru(tpy)(bpy)(Nh)](2+) (Ru1, tpy = 2,2';6',2″-terpyridine, bpy = 2'2-bipyridine, Nh = Norharman), [Ru(tpy)(phen)(Nh)](2+) (Ru2, phen = 1,10-phenanthroline), [Ru(tpy)(dpa)(Nh)](2+) (Ru3, dpa = 2,2'-dipyridylamine) and [Ru(tpy)(dip)(Nh)](2+) (Ru4, dip = 4,7-diphenyl-1,10-phenanthroline) were presented as anticancer drugs. In vitro cytotoxicity assays indicated that these complexes showed anticancer activity against various cancer cells. Flow cytometry and signaling pathways analysis demonstrated that these complexes induced apoptosis via the mitochondrial pathway, as evidenced by the loss of mitochondrial membrane potential and the release of cytochrome c. The resulting accumulation of p53 proteins from phosphorylation at serine-15 and serine-392 was correlated with an increase in p21 and caspase activation. Taken together, these findings suggested that Ru1-Ru4 may contribute to the future development of improved chemotherapeutics against human cancers.
Collapse
Key Words
- 1,10-phenanthroline
- 2,2′-bipyridine
- 2,2′-dipyridylamine
- 2,2′:6′,2″-terpyridine
- 2′,7′-dichlorofluorescein-diacetate
- 4,5-dihydroxy-1,3-benzenedisulfonic acid disodium salt
- 4,7-dibenzyl-1,10-phenanthroline
- Apoptosis
- CCCP
- DCFH-DA
- Mitochondria
- N-acetylcysteine
- NAC
- Nh
- Norharman
- PI
- Ruthenium(II) complex
- bpy
- carbonyl cyanide m-chlorophenylhydrazone
- dip
- dpa
- p53
- phen
- propidium iodide
- tiron
- tpy
- β-Carboline
Collapse
|