1
|
Ma CI, Tirtorahardjo JA, Schweizer SS, Zhang J, Fang Z, Xing L, Xu M, Herman DA, Kleinman MT, McCullough BS, Barrios AM, Andrade RM. Gold(I) ion and the phosphine ligand are necessary for the anti- Toxoplasma gondii activity of auranofin. Microbiol Spectr 2024; 12:e0296823. [PMID: 38206030 PMCID: PMC10845965 DOI: 10.1128/spectrum.02968-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 12/08/2023] [Indexed: 01/12/2024] Open
Abstract
Auranofin, an FDA-approved drug for rheumatoid arthritis, has emerged as a promising antiparasitic medication in recent years. The gold(I) ion in auranofin is postulated to be responsible for its antiparasitic activity. Notably, aurothiomalate and aurothioglucose also contain gold(I), and, like auranofin, they were previously used to treat rheumatoid arthritis. Whether they have antiparasitic activity remains to be elucidated. Herein, we demonstrated that auranofin and similar derivatives, but not aurothiomalate and aurothioglucose, inhibited the growth of Toxoplasma gondii in vitro. We found that auranofin affected the T. gondii biological cycle (lytic cycle) by inhibiting T. gondii's invasion and triggering its egress from the host cell. However, auranofin could not prevent parasite replication once T. gondii resided within the host. Auranofin treatment induced apoptosis in T. gondii parasites, as demonstrated by its reduced size and elevated phosphatidylserine externalization (PS). Notably, the gold from auranofin enters the cytoplasm of T. gondii, as demonstrated by scanning transmission electron microscopy-energy dispersive X-ray spectroscopy (STEM-EDS) and Inductively Coupled Plasma-Mass Spectrometry (ICP-MS).IMPORTANCEToxoplasmosis, caused by Toxoplasma gondii, is a devastating disease affecting the brain and the eyes, frequently affecting immunocompromised individuals. Approximately 60 million people in the United States are already infected with T. gondii, representing a population at-risk of developing toxoplasmosis. Recent advances in treating cancer, autoimmune diseases, and organ transplants have contributed to this at-risk population's exponential growth. Paradoxically, treatments for toxoplasmosis have remained the same for more than 60 years, relying on medications well-known for their bone marrow toxicity and allergic reactions. Discovering new therapies is a priority, and repurposing FDA-approved drugs is an alternative approach to speed up drug discovery. Herein, we report the effect of auranofin, an FDA-approved drug, on the biological cycle of T. gondii and how both the phosphine ligand and the gold molecule determine the anti-parasitic activity of auranofin and other gold compounds. Our studies would contribute to the pipeline of candidate anti-T. gondii agents.
Collapse
Affiliation(s)
- C. I. Ma
- Department of Medicine, Division of Infectious Diseases, University of California at Irvine, Irvine, California, USA
| | - J. A. Tirtorahardjo
- Department of Microbiology and Molecular Genetics, University of California at Irvine, Irvine, California, USA
| | - S. S. Schweizer
- School of Biological Sciences; University of California at Irvine, Irvine, California, USA
| | - J. Zhang
- School of Biological Sciences; University of California at Irvine, Irvine, California, USA
| | - Z. Fang
- School of Biological Sciences; University of California at Irvine, Irvine, California, USA
| | - L. Xing
- Irvine Materials Research Institute; University of California at Irvine, Irvine, California, USA
| | - M. Xu
- Irvine Materials Research Institute; University of California at Irvine, Irvine, California, USA
| | - D. A. Herman
- Department of Medicine, Occupational and Environmental Medicine, University of California at Irvine, Irvine, California, USA
| | - M. T. Kleinman
- Department of Medicine, Occupational and Environmental Medicine, University of California at Irvine, Irvine, California, USA
| | - B. S. McCullough
- Department of Medicinal Chemistry, University of Utah College of Pharmacy, Salt Lake City, Utah, USA
| | - A. M. Barrios
- Department of Medicinal Chemistry, University of Utah College of Pharmacy, Salt Lake City, Utah, USA
| | - R. M. Andrade
- Department of Medicine, Division of Infectious Diseases, University of California at Irvine, Irvine, California, USA
- Department of Microbiology and Molecular Genetics, University of California at Irvine, Irvine, California, USA
| |
Collapse
|
2
|
Qu Z, Krabill AD, Zhang ZY. High-Throughput Discovery and Characterization of Covalent Inhibitors for Protein Tyrosine Phosphatases. Methods Mol Biol 2024; 2743:301-316. [PMID: 38147223 DOI: 10.1007/978-1-0716-3569-8_19] [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] [Indexed: 12/27/2023]
Abstract
Covalent inhibition has gained increasing interest in targeting the undruggable protein tyrosine phosphatases (PTPs). However, a systematic method for discovering and characterizing covalent PTP inhibitors has yet to be established. Here, we describe a workflow involving high-throughput screening of covalent fragment libraries and a novel biochemical assay that enables the acquisition of kinetics parameters of PTP inhibition by covalent inhibitors with higher throughput.
Collapse
Affiliation(s)
- Zihan Qu
- Department of Chemistry, Purdue University, West Lafayette, IN, USA
| | - Aaron D Krabill
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, USA
| | - Zhong-Yin Zhang
- Department of Chemistry, Purdue University, West Lafayette, IN, USA.
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, USA.
- Institute for Cancer Research, Purdue University, West Lafayette, IN, USA.
- Institute for Drug Discovery, Purdue University, West Lafayette, IN, USA.
| |
Collapse
|
3
|
Hu L, Li H, Qin J, Yang D, Liu J, Luo X, Ma J, Luo C, Ye F, Zhou Y, Li J, Wang M. Discovery of PVD-06 as a Subtype-Selective and Efficient PTPN2 Degrader. J Med Chem 2023; 66:15269-15287. [PMID: 37966047 DOI: 10.1021/acs.jmedchem.3c01348] [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: 11/16/2023]
Abstract
Protein tyrosine phosphatase nonreceptor Type 2 (PTPN2) is an attractive target for cancer immunotherapy. PTPN2 and another subtype of PTP1B are highly similar in structure, but their biological functions are distinct. Therefore, subtype-selective targeting of PTPN2 remains a challenge for researchers. Herein, the development of small molecular PTPN2 degraders based on a thiadiazolidinone dioxide-naphthalene scaffold and a VHL E3 ligase ligand is described, and the PTPN2/PTP1B subtype-selective degradation is achieved for the first time. The linker structure modifications led to the discovery of the subtype-selective PTPN2 degrader PVD-06 (PTPN2/PTP1B selective index > 60-fold), which also exhibits excellent proteome-wide degradation selectivity. PVD-06 induces PTPN2 degradation in a ubiquitination- and proteasome-dependent manner. It efficiently promotes T cell activation and amplifies IFN-γ-mediated B16F10 cell growth inhibition. This study provides a convenient chemical knockdown tool for PTPN2-related research and a paradigm for subtype-selective PTP degradation through nonspecific substrate-mimicking ligands, demonstrating the therapeutic potential of PTPN2 subtype-selective degradation.
Collapse
Affiliation(s)
- Linghao Hu
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan Tsuihang New District, Guangdong 528400, China
| | - Huiyun Li
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan Tsuihang New District, Guangdong 528400, China
- School of Pharmacy, Zunyi Medical University, Zunyi 563000, Guizhou China
| | - Junlin Qin
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Dan Yang
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan Tsuihang New District, Guangdong 528400, China
- School of Pharmaceutical Sciences, Southern Medical University, No.1023, South Shatai Road, Baiyun District, Guangzhou 510515, Guangdong, China
| | - Jieming Liu
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan Tsuihang New District, Guangdong 528400, China
| | - Xiaomin Luo
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan Tsuihang New District, Guangdong 528400, China
| | | | - Cheng Luo
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan Tsuihang New District, Guangdong 528400, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Fei Ye
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Yubo Zhou
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan Tsuihang New District, Guangdong 528400, China
| | - Jia Li
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan Tsuihang New District, Guangdong 528400, China
- School of Pharmacy, Zunyi Medical University, Zunyi 563000, Guizhou China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Mingliang Wang
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan Tsuihang New District, Guangdong 528400, China
- School of Pharmaceutical Sciences, Southern Medical University, No.1023, South Shatai Road, Baiyun District, Guangzhou 510515, Guangdong, China
| |
Collapse
|
4
|
Tialiou A, Chin J, Keppler BK, Reithofer MR. Current Developments of N-Heterocyclic Carbene Au(I)/Au(III) Complexes toward Cancer Treatment. Biomedicines 2022; 10:biomedicines10061417. [PMID: 35740438 PMCID: PMC9219884 DOI: 10.3390/biomedicines10061417] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/07/2022] [Accepted: 06/08/2022] [Indexed: 11/29/2022] Open
Abstract
Since their first discovery, N-heterocyclic carbenes have had a significant impact on organometallic chemistry. Due to their nature as strong σ-donor and π-acceptor ligands, they are exceptionally well suited to stabilize Au(I) and Au(III) complexes in biological environments. Over the last decade, the development of rationally designed NHCAu(I/III) complexes to specifically target DNA has led to a new “gold rush” in bioinorganic chemistry. This review aims to summarize the latest advances of NHCAu(I/III) complexes that are able to interact with DNA. Furthermore, the latest advancements on acyclic diamino carbene gold complexes with anticancer activity are presented as these typically overlooked NHC alternatives offer great additional design possibilities in the toolbox of carbene-stabilized gold complexes for targeted therapy.
Collapse
Affiliation(s)
- Alexia Tialiou
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Währinger Str. 42, 1090 Vienna, Austria; (A.T.); (B.K.K.)
- Vienna Doctoral School in Chemistry (DoSChem), University of Vienna, Währinger Str. 42, 1090 Vienna, Austria
| | - Jiamin Chin
- Institute of Inorganic Chemistry—Functional Materials, Faculty of Chemistry, University of Vienna, Währinger Str. 42, 1090 Vienna, Austria
- Correspondence: (J.C.); (M.R.R.)
| | - Bernhard K. Keppler
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Währinger Str. 42, 1090 Vienna, Austria; (A.T.); (B.K.K.)
- Research Cluster “Translational Cancer Therapy Research”, University of Vienna and Medical University of Vienna, Währinger Str. 42, 1090 Vienna, Austria
| | - Michael R. Reithofer
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Währinger Str. 42, 1090 Vienna, Austria; (A.T.); (B.K.K.)
- Correspondence: (J.C.); (M.R.R.)
| |
Collapse
|
5
|
Jassim BA, Lin J, Zhang ZY. PTPN22: Structure, Function, and Developments in Inhibitor Discovery with Applications for Immunotherapy. Expert Opin Drug Discov 2022; 17:825-837. [PMID: 35637605 PMCID: PMC9378720 DOI: 10.1080/17460441.2022.2084607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION While immunotherapy strategies such as immune checkpoint inhibition and adoptive T cell therapy have become commonplace in cancer therapy, they suffer from limitations, including lack of patient response and toxicity. To wield the maximum potential of the immune system, cancer immunotherapy must integrate novel targets and therapeutic strategies with potential to augment clinical efficacy of currently utilized immunotherapies. PTPN22, a member of the protein tyrosine phosphatase (PTP) superfamily that downregulates T cell signaling and proliferation, has recently emerged as a systemically druggable and novel immunotherapy target. AREAS COVERED This review describes the basics of PTPN22 structure and function and provides comprehensive insight into recent advances in small molecule PTPN22 inhibitor development and the immense potential of PTPN22 inhibition to synergize with current immunotherapies. EXPERT OPINION It is apparent that small molecule PTPN22 inhibitors have enormous potential to augment efficacy of current immunotherapy strategies such as checkpoint inhibition and adoptive cell transfer. Nevertheless, several constraints must be overcome before these inhibitors can be applied as useful therapeutics, namely selectivity, potency, and in vivo efficacy.
Collapse
Affiliation(s)
- Brenson A Jassim
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue Institute for Drug Discovery, West Lafayette
| | - Jianping Lin
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue Institute for Drug Discovery, West Lafayette
| | - Zhong-Yin Zhang
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue Institute for Drug Discovery, West Lafayette
| |
Collapse
|
6
|
Rachor SG, Müller R, Wittwer P, Kaupp M, Braun T. Synthesis, Reactivity, and Bonding of Gold(I) Fluorido-Phosphine Complexes. Inorg Chem 2021; 61:357-367. [PMID: 34913690 DOI: 10.1021/acs.inorgchem.1c02959] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Gold(I) fluorido complexes with phosphine ligands have been synthesized from their respective iodido precursors. The bonding situation in comparison between complexes bearing phosphines and N-heterocyclic carbenes (NHCs) was explored quantum-chemically, obtaining similar results for both. Calculations of the 19F NMR chemical shifts match the experimental values well, including the approximately 40 ppm low-field shifts for the phosphine complexes compared to the NHC complexes, in spite of similar negative charges on fluorine. The reactivity of the highly water-sensitive gold(I) fluorido complexes was studied, resulting in substitution at the metal using trimethylsilyl reagents. The compounds studied were characterized using NMR as well as X-ray diffraction methods.
Collapse
Affiliation(s)
- Simon G Rachor
- Department of Chemistry, Humboldt-Universität zu Berlin, Brook-Taylor-Straße 2, 12489 Berlin, Germany
| | - Robert Müller
- Institut für Chemie, Theoretische Chemie/Quantenchemie, Technische Universität Berlin, Sekr. C7, Strasse des 17 Juni 135, 10623 Berlin, Germany
| | - Philipp Wittwer
- Department of Chemistry, Humboldt-Universität zu Berlin, Brook-Taylor-Straße 2, 12489 Berlin, Germany
| | - Martin Kaupp
- Institut für Chemie, Theoretische Chemie/Quantenchemie, Technische Universität Berlin, Sekr. C7, Strasse des 17 Juni 135, 10623 Berlin, Germany
| | - Thomas Braun
- Department of Chemistry, Humboldt-Universität zu Berlin, Brook-Taylor-Straße 2, 12489 Berlin, Germany
| |
Collapse
|
7
|
Rashmi, More SK, Wang Q, Vomhof-DeKrey EE, Porter JE, Basson MD. ZINC40099027 activates human focal adhesion kinase by accelerating the enzymatic activity of the FAK kinase domain. Pharmacol Res Perspect 2021; 9:e00737. [PMID: 33715263 PMCID: PMC7955952 DOI: 10.1002/prp2.737] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 01/28/2021] [Accepted: 01/29/2021] [Indexed: 12/16/2022] Open
Abstract
Focal adhesion kinase (FAK) regulates gastrointestinal epithelial restitution and healing. ZINC40099027 (Zn27) activates cellular FAK and promotes intestinal epithelial wound closure in vitro and in mice. However, whether Zn27 activates FAK directly or indirectly remains unknown. We evaluated Zn27 potential modulation of the key phosphatases, PTP-PEST, PTP1B, and SHP2, that inactivate FAK, and performed in vitro kinase assays with purified FAK to assess direct Zn27-FAK interaction. In human Caco-2 cells, Zn27-stimulated FAK-Tyr-397 phosphorylation despite PTP-PEST inhibition and did not affect PTP1B-FAK interaction or SHP2 activity. Conversely, in vitro kinase assays demonstrated that Zn27 directly activates both full-length 125 kDa FAK and its 35 kDa kinase domain. The ATP-competitive FAK inhibitor PF573228 reduced basal and ZN27-stimulated FAK phosphorylation in Caco-2 cells, but Zn27 increased FAK phosphorylation even in cells treated with PF573228. Increasing PF573228 concentrations completely prevented activation of 35 kDa FAK in vitro by a normally effective Zn27 concentration. Conversely, increasing Zn27 concentrations dose-dependently activated kinase activity and overcame PF573228 inhibition of FAK, suggesting the direct interactions of Zn27 with FAK may be competitive. Zn27 increased the maximal activity (Vmax ) of FAK. The apparent Km of the substrate also increased under laboratory conditions less relevant to intracellular ATP concentrations. These results suggest that Zn27 is highly potent and enhances FAK activity via allosteric interaction with the FAK kinase domain to increase the Vmax of FAK for ATP. Understanding Zn27 enhancement of FAK activity will be important to redesign and develop a clinical drug that can promote mucosal wound healing.
Collapse
Affiliation(s)
- Rashmi
- Department of Surgery, University of North Dakota School of Medicine & Health Sciences, Grand Forks, ND, USA
| | - Shyam K More
- Department of Surgery, University of North Dakota School of Medicine & Health Sciences, Grand Forks, ND, USA
| | - Qinggang Wang
- Department of Surgery, University of North Dakota School of Medicine & Health Sciences, Grand Forks, ND, USA
| | - Emilie E Vomhof-DeKrey
- Department of Surgery, University of North Dakota School of Medicine & Health Sciences, Grand Forks, ND, USA
- Department of Biomedical Sciences, University of North Dakota School of Medicine & Health Sciences, Grand Forks, ND, USA
| | - James E Porter
- Department of Biomedical Sciences, University of North Dakota School of Medicine & Health Sciences, Grand Forks, ND, USA
| | - Marc D Basson
- Department of Surgery, University of North Dakota School of Medicine & Health Sciences, Grand Forks, ND, USA
- Department of Biomedical Sciences, University of North Dakota School of Medicine & Health Sciences, Grand Forks, ND, USA
- Department of Pathology, University of North Dakota School of Medicine & Health Sciences, Grand Forks, ND, USA
| |
Collapse
|
8
|
Synthesis and Structure of (Nitronyl Nitroxide-2-ido)(tert-butyldiphenylphosphine)gold(I) and -(Di(tert-butyl)phenylphosphine)gold(I) Derivatives; Their Comparative Study in the Cross-Coupling Reaction. CRYSTALS 2020. [DOI: 10.3390/cryst10090770] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We synthesized two new organogold derivatives of a nitronyl nitroxide (NN) with phosphine ligands, namely NN-Au-PtBuPh2 and NN-Au-PtBu2Ph. They were characterized by X-ray diffraction analysis, cyclic voltammetry, and ESR, IR, and UV/Vis spectroscopy. The X-ray structural analysis revealed a tendency of the NN moiety to form a large number of short intermolecular contacts. This phenomenon is related to the anionic nature of the paramagnetic group NN, as evidenced by a significantly lower oxidation potential in comparison with purely organic derivatives of NN radicals. The cross-coupling reaction of NN-Au-PPh3, NN-Au-PtBuPh2, or NN-Au-PtBu2Ph with an activated bromoarene, namely, p-BrC6H4NO2, was investigated. It was shown that regardless of the presence of the bulky tert-butyl substituent, all gold derivatives have similar activities in the cross-coupling reaction and give a cross-coupling product, NN-C6H4NO2, with comparable yields.
Collapse
|
9
|
|
10
|
Alsaeedi MS, Babgi BA, Hussien MA, Abdellattif MH, Humphrey MG. DNA-Binding and Anticancer Activity of Binuclear Gold(I) Alkynyl Complexes with a Phenanthrenyl Bridging Ligand. Molecules 2020; 25:E1033. [PMID: 32106590 PMCID: PMC7179095 DOI: 10.3390/molecules25051033] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 02/17/2020] [Accepted: 02/21/2020] [Indexed: 12/16/2022] Open
Abstract
3,6-Diethynyl-9,10-diethoxyphenanthrene (4) was synthesized from phenanthrene and employed in the synthesis of the binuclear gold(I) alkynyl complexes (R3P)Au(C≡C-3-[C14H6-9,10-diethoxy]-6-C≡C)Au(PR3) (R = Ph (5a), Cy (5b)). The diyne 4 and complexes 5a and 5b were characterized by NMR spectroscopy, mass spectrometry, and elemental analysis. UV-Vis spectroscopy studies of the metal complexes and precursor diyne show strong p à p* transitions in the near UV region that red shift by ca. 50 nm upon coordination at the gold centers. The emission spectrum of 4 shows an intense fluorescence band centered at 420 nm which red shifts, slightly upon coordination of 4 to gold. Binding studies of 4, 5a, and 5b against calf thymus DNA were carried out, revealing that 4, 5a, and 5b have >40% stronger binding affinities than the commonly used intercalating agent ethidium bromide. The molecular docking scores of 4, 5a, and 5b with B-DNA suggest a similar trend in behavior to that observed in the DNA-binding study. Unlike the ligand 4, promising anticancer properties for 5a and 5b were observed against several cell lines; the DNA binding capability of the precursor alkyne was maintained, and its anticancer efficacy enhanced by the gold centers. Such phenanthrenyl complexes could be promising candidates in certain biological applications because the two components (phenanthrenyl bridge and metal centers) can be altered independently to improve the targeting of the complex, as well as the biological and physicochemical properties.
Collapse
Affiliation(s)
- Mona S. Alsaeedi
- Department of Chemistry, Faculty of Science, King Abdulaziz University, P.O. Box 80203 Jeddah 21589, Saudi Arabia; (M.S.A.); (M.A.H.)
- Department of Chemistry, Faculty of Science, Taif University, Al-Haweiah, P.O. Box 888, Taif 21974, Saudi Arabia;
| | - Bandar A. Babgi
- Department of Chemistry, Faculty of Science, King Abdulaziz University, P.O. Box 80203 Jeddah 21589, Saudi Arabia; (M.S.A.); (M.A.H.)
- Department of Chemistry, College of Science and Arts, King Abdulaziz University, P.O. Box 344 Rabigh 21911, Saudi Arabia
| | - Mostafa A. Hussien
- Department of Chemistry, Faculty of Science, King Abdulaziz University, P.O. Box 80203 Jeddah 21589, Saudi Arabia; (M.S.A.); (M.A.H.)
| | - Magda H. Abdellattif
- Department of Chemistry, Faculty of Science, Taif University, Al-Haweiah, P.O. Box 888, Taif 21974, Saudi Arabia;
| | - Mark G. Humphrey
- Research School of Chemistry, Australian National University, Canberra, ACT 2601, Australia
| |
Collapse
|
11
|
Stratton M, Ramachandran A, Camacho EJM, Patil S, Waris G, Grice KA. Anti-fibrotic activity of gold and platinum complexes - Au(I) compounds as a new class of anti-fibrotic agents. J Inorg Biochem 2020; 206:111023. [PMID: 32163811 DOI: 10.1016/j.jinorgbio.2020.111023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 02/06/2020] [Accepted: 02/09/2020] [Indexed: 12/11/2022]
Abstract
Molecular gold(I) and platinum(II) species were examined for the inhibition of liver fibrosis and the hepatitis C virus (HCV). Determination of inhibition efficiency was conducted via morphological analysis, cell viability, western blot analysis, and quantitative reverse transcription polymerase chain reaction (RT-PCR). Auranofin and Ph3PAuCl demonstrated the greatest inhibition of liver fibrosis amongst the tested gold species in human hepatic stellate LX-2 cells. Western blot analysis indicated that auranofin and Ph3PAuCl prevent signal transducer and activator of transcription 3 (STAT3) phosphorylation, which may be a key connection to fibrosis and inflammation. Auranofin and Ph3PAuCl also reduced expression of HCV-nonstructural protein 3 (NS3) and HCV-NS5a proteins in a HCV subgenomic replicon system. These results demonstrate significant promise for the use of gold compounds in treating liver diseases such as HCV.
Collapse
Affiliation(s)
- Matthew Stratton
- Department of Microbiology and Immunology, Center for Cancer Cell Biology, Immunology and Infection, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| | - Akshaya Ramachandran
- Department of Microbiology and Immunology, Center for Cancer Cell Biology, Immunology and Infection, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| | | | - Shivaputra Patil
- Pharmaceutical Sciences Department, College of Pharmacy, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| | - Gulam Waris
- Department of Microbiology and Immunology, Center for Cancer Cell Biology, Immunology and Infection, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| | - Kyle A Grice
- Department of Chemistry and Biochemistry, College of Science and Health, DePaul University, Chicago, IL 60614, USA.
| |
Collapse
|
12
|
Sánchez Delgado GY, Paschoal D, de Oliveira MA, Dos Santos HF. Structure and redox stability of [Au(III)(X^N^X)PR3] complexes (X = C or N) in aqueous solution: The role of phosphine auxiliary ligand. J Inorg Biochem 2019; 200:110804. [DOI: 10.1016/j.jinorgbio.2019.110804] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 08/08/2019] [Accepted: 08/18/2019] [Indexed: 11/29/2022]
|
13
|
Abstract
Background:
Since the serendipitous discovery of the antitumor activity of cisplatin
there has been a continuous surge in studies aimed at the development of new cytotoxic
metal complexes. While the majority of these complexes have been designed to interact with
nuclear DNA, other targets for anticancer metallodrugs attract increasing interest. In cancer
cells the mitochondrial metabolism is deregulated. Impaired apoptosis, insensitivity to antigrowth
signals and unlimited proliferation have been linked to mitochondrial dysfunction. It
is therefore not surprising that mitochondria have emerged as a major target for cancer therapy.
Mitochondria-targeting agents are able to bypass resistance mechanisms and to (re-) activate
cell-death programs.
Methods:
Web-based literature searching tools such as SciFinder were used to search for reports
on cytotoxic metal complexes that are taken up by the mitochondria and interact with
mitochondrial DNA or mitochondrial proteins, disrupt the mitochondrial membrane potential,
facilitate mitochondrial membrane permeabilization or activate mitochondria-dependent celldeath
signaling by unbalancing the cellular redox state. Included in the search were publications
investigating strategies to selectively accumulate metallodrugs in the mitochondria.
Results:
This review includes 241 references on antimitochondrial metal complexes, the use
of mitochondria-targeting carrier ligands and the formation of lipophilic cationic complexes.
Conclusion:
Recent developments in the design, cytotoxic potency, and mechanistic understanding
of antimitochondrial metal complexes, in particular of cyclometalated Au, Ru, Ir and
Pt complexes, Ru polypyridine complexes and Au-N-heterocyclic carbene and phosphine
complexes are summarized and discussed.
Collapse
Affiliation(s)
- Andrea Erxleben
- School of Chemistry, National University of Ireland, Galway, Ireland
| |
Collapse
|
14
|
Casey GR, Stains CI. Interrogating Protein Phosphatases with Chemical Activity Probes. Chemistry 2018; 24:7810-7824. [PMID: 29338103 PMCID: PMC5986605 DOI: 10.1002/chem.201705194] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Indexed: 12/30/2022]
Abstract
Protein phosphatases, while long overlooked, have recently become appreciated as drivers of both normal- and disease-associated signaling events. As a result, the spotlight is now turning torwards this enzyme family and efforts geared towards the development of modern chemical tools for studying these enzymes are well underway. This Minireview focuses on the evolution of chemical activity probes, both optical and covalent, for the study of protein phosphatases. Small-molecule probes, global monitoring of phosphatase activity through the use of covalent modifiers, and targeted fluorescence-based activity probes are discussed. We conclude with an overview of open questions in the field and highlight the potential impact of chemical tools for studying protein phosphatases.
Collapse
Affiliation(s)
- Garrett R Casey
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Cliff I Stains
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| |
Collapse
|
15
|
McCullough BS, Barrios AM. Facile, Fluorogenic Assay for Protein Histidine Phosphatase Activity. Biochemistry 2018; 57:2584-2589. [DOI: 10.1021/acs.biochem.8b00278] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
- Brandon S. McCullough
- Department of Medicinal Chemistry, University of Utah, Salt Lake City, Utah 84112, United States
| | - Amy M. Barrios
- Department of Medicinal Chemistry, University of Utah, Salt Lake City, Utah 84112, United States
| |
Collapse
|
16
|
Ruddraraju KV, Zhang ZY. Covalent inhibition of protein tyrosine phosphatases. MOLECULAR BIOSYSTEMS 2018; 13:1257-1279. [PMID: 28534914 DOI: 10.1039/c7mb00151g] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Protein tyrosine phosphatases (PTPs) are a large family of 107 signaling enzymes that catalyze the hydrolytic removal of phosphate groups from tyrosine residues in a target protein. The phosphorylation status of tyrosine residues on proteins serve as a ubiquitous mechanism for cellular signal transduction. Aberrant function of PTPs can lead to many human diseases, such as diabetes, obesity, cancer, and autoimmune diseases. As the number of disease relevant PTPs increases, there is urgency in developing highly potent inhibitors that are selective towards specific PTPs. Most current efforts have been devoted to the development of active site-directed and reversible inhibitors for PTPs. This review summarizes recent progress made in the field of covalent inhibitors to target PTPs. Here, we discuss the in vivo and in vitro inactivation of various PTPs by small molecule-containing electrophiles, such as Michael acceptors, α-halo ketones, epoxides, and isothiocyanates, etc. as well as oxidizing agents. We also suggest potential strategies to transform these electrophiles into isozyme selective covalent PTP inhibitors.
Collapse
Affiliation(s)
- Kasi Viswanatharaju Ruddraraju
- Department of Medicinal Chemistry and Molecular Pharmacology, Department of Chemistry, Center for Cancer Research, and Institute for Drug Discovery, Purdue University, 720 Clinic Drive, West Lafayette, IN 47907, USA.
| | | |
Collapse
|
17
|
Iacopetta D, Mariconda A, Saturnino C, Caruso A, Palma G, Ceramella J, Muià N, Perri M, Sinicropi MS, Caroleo MC, Longo P. Novel Gold and Silver Carbene Complexes Exert Antitumor Effects Triggering the Reactive Oxygen Species Dependent Intrinsic Apoptotic Pathway. ChemMedChem 2017; 12:2054-2065. [PMID: 29120085 DOI: 10.1002/cmdc.201700634] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Indexed: 12/20/2022]
Abstract
Cisplatin and other platinum-based drugs are well-known valid anticancer drugs. However, during chemotherapy, the presence of numerous side effects and the onset of frequent phenomena of resistance has pushed many research groups to devise new metal-based compounds holding improved anticancer properties and fewer undesired effects. Amongst the variety of synthesized compounds, significant antiproliferative effects have been obtained by employing organometallic compounds, particularly those based on silver and gold. With this in mind, we synthesized four compounds, two silver complexes and two gold complexes, with good inhibitory effects on the in vitro proliferation of breast and ovarian cancer-cell models. The antitumor activity of the most active compound, that is, AuL4, was found to be ninefold higher than that of cisplatin, and this compound induced dramatic morphological changes in HeLa cells. AuL4 induced PARP-1 cleavage, caspases 3/7 and 9 activation, mitochondria disruption, cytochrome c release in cancer-cell cytoplasm, and the intracellular production of reactive oxygen species. Thus, AuL4 treatment caused cancer-cell death by the intrinsic apoptotic pathway, whereas no cytotoxic effects were recorded upon treating non-tumor cell lines. The reported outcomes may be an important contribution to the expanding knowledge of medicinal bio-organometallic chemistry and enlarge the available anticancer toolbox, offering improved features, such as higher activity and/or selectivity, and opening the way to new discoveries and applications.
Collapse
Affiliation(s)
- Domenico Iacopetta
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via Pietro Bucci, 87036, Arcavacata di Rende, Italy
| | - Annaluisa Mariconda
- Department of Biology and Chemistry, University of Salerno, Via Giovanni Paolo II, 132, Fisciano, 84084, Italy
| | - Carmela Saturnino
- Department of Science, University of Basilicata, Viale dell'Ateneo Lucano 10, Potenza, 85100, Italy
| | - Anna Caruso
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via Pietro Bucci, 87036, Arcavacata di Rende, Italy
| | - Giuseppe Palma
- SSD Sperimentazione Animale, Istituto Nazionale Tumori, IRCCS, "Fondazione G. Pascale", Via Mariano Semmola, Napoli, 80131, Italy
| | - Jessica Ceramella
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via Pietro Bucci, 87036, Arcavacata di Rende, Italy
| | - Noemi Muià
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via Pietro Bucci, 87036, Arcavacata di Rende, Italy
| | - Mariarita Perri
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via Pietro Bucci, 87036, Arcavacata di Rende, Italy
| | - Maria Stefania Sinicropi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via Pietro Bucci, 87036, Arcavacata di Rende, Italy
| | - Maria Cristina Caroleo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via Pietro Bucci, 87036, Arcavacata di Rende, Italy
| | - Pasquale Longo
- Department of Biology and Chemistry, University of Salerno, Via Giovanni Paolo II, 132, Fisciano, 84084, Italy
| |
Collapse
|
18
|
González-Liste PJ, García-Garrido SE, Cadierno V. Gold(i)-catalyzed addition of carboxylic acids to internal alkynes in aqueous medium. Org Biomol Chem 2017; 15:1670-1679. [DOI: 10.1039/c6ob02800d] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A broad scope catalytic system for the intermolecular addition of carboxylic acids to internal alkynes, in water under mild conditions, has been developed.
Collapse
Affiliation(s)
- Pedro J. González-Liste
- Laboratorio de Compuestos Organometálicos y Catálisis (Unidad Asociada al CSIC)
- Centro de Innovación en Química Avanzada (ORFEO-CINQA)
- Departamento de Química Orgánica e Inorgánica
- IUQOEM
- Universidad de Oviedo
| | - Sergio E. García-Garrido
- Laboratorio de Compuestos Organometálicos y Catálisis (Unidad Asociada al CSIC)
- Centro de Innovación en Química Avanzada (ORFEO-CINQA)
- Departamento de Química Orgánica e Inorgánica
- IUQOEM
- Universidad de Oviedo
| | - Victorio Cadierno
- Laboratorio de Compuestos Organometálicos y Catálisis (Unidad Asociada al CSIC)
- Centro de Innovación en Química Avanzada (ORFEO-CINQA)
- Departamento de Química Orgánica e Inorgánica
- IUQOEM
- Universidad de Oviedo
| |
Collapse
|
19
|
Tomaschautzky J, Neumann B, Stammler HG, Mix A, Mitzel NW. Tridentate Lewis-acids based on triphenylsilane. Dalton Trans 2017; 46:1645-1659. [DOI: 10.1039/c6dt04293g] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Triphenylsilanes are versatile propeller-shaped building blocks and have been used for the syntheses of several novel poly-Lewis-acids. The first solid-state structure of a bisma-silatriptycene as well as investigations in host–guest chemistry of a triple alane with a threefold amine are reported.
Collapse
Affiliation(s)
- Janek Tomaschautzky
- Inorganic and Structural Chemistry
- Centre for Molecular Materials CM2
- Fakultät für Chemie
- Universität Bielefeld
- 33615 Bielefeld
| | - Beate Neumann
- Inorganic and Structural Chemistry
- Centre for Molecular Materials CM2
- Fakultät für Chemie
- Universität Bielefeld
- 33615 Bielefeld
| | - Hans-Georg Stammler
- Inorganic and Structural Chemistry
- Centre for Molecular Materials CM2
- Fakultät für Chemie
- Universität Bielefeld
- 33615 Bielefeld
| | - Andreas Mix
- Inorganic and Structural Chemistry
- Centre for Molecular Materials CM2
- Fakultät für Chemie
- Universität Bielefeld
- 33615 Bielefeld
| | - Norbert W. Mitzel
- Inorganic and Structural Chemistry
- Centre for Molecular Materials CM2
- Fakultät für Chemie
- Universität Bielefeld
- 33615 Bielefeld
| |
Collapse
|
20
|
Hopper M, Yun JF, Zhou B, Le C, Kehoe K, Le R, Hill R, Jongeward G, Debnath A, Zhang L, Miyamoto Y, Eckmann L, Land KM, Wrischnik LA. Auranofin inactivates Trichomonas vaginalis thioredoxin reductase and is effective against trichomonads in vitro and in vivo. Int J Antimicrob Agents 2016; 48:690-694. [PMID: 27839893 DOI: 10.1016/j.ijantimicag.2016.09.020] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Revised: 09/06/2016] [Accepted: 09/15/2016] [Indexed: 11/26/2022]
Abstract
Trichomoniasis, caused by the protozoan parasite Trichomonas vaginalis, is the most common, non-viral, sexually transmitted infection in the world, but only two closely related nitro drugs are approved for its treatment. New antimicrobials against trichomoniasis remain an urgent need. Several organic gold compounds were tested for activity against T. vaginalis thioredoxin reductase (TrxR) in cell-free systems as well as for activity against different trichomonads in vitro and in a murine infection model. The organic gold(I) compounds auranofin and chloro(diethylphenylphosphine)gold(I) inhibited TrxR in a concentration-dependent manner in assays with recombinant purified reductase and in cytoplasmic extracts of T. vaginalis transfected with a haemagglutinin epitope-tagged form of the reductase. Auranofin potently suppressed the growth of three independent clinical T. vaginalis isolates as well as several strains of another trichomonad (Tritrichomonas foetus) in a 24 h-assay, with 50% inhibitory concentrations of 0.7-2.5 µM and minimum lethal concentrations of 2-6 µM. The drug also compromised the ability of the parasite to overcome oxidant stress, supporting the notion that auranofin acts, in part, by inactivating TrxR-dependent antioxidant defences. Chloro(diethylphenylphosphine)gold(I) was 10-fold less effective against T. vaginalis in vitro than auranofin. Oral administration of auranofin for 4 days cleared the parasites in a murine model of vaginal T. foetus infection without displaying any apparent adverse effects. The approved human drug auranofin may be a promising agent as an alternative treatment of trichomoniasis in cases when standard nitro drug therapies have failed.
Collapse
Affiliation(s)
- Melissa Hopper
- Department of Biological Sciences, University of the Pacific, Stockton, CA, USA
| | - Jeong-Fil Yun
- Department of Biological Sciences, University of the Pacific, Stockton, CA, USA
| | - Bianhua Zhou
- Department of Medicine, School of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Christine Le
- Department of Medicine, School of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Katelin Kehoe
- Department of Biological Sciences, University of the Pacific, Stockton, CA, USA
| | - Ryan Le
- Department of Biological Sciences, University of the Pacific, Stockton, CA, USA
| | - Ryan Hill
- Department of Biological Sciences, University of the Pacific, Stockton, CA, USA
| | - Gregg Jongeward
- Department of Biological Sciences, University of the Pacific, Stockton, CA, USA
| | - Anjan Debnath
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Liangfang Zhang
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA, USA
| | - Yukiko Miyamoto
- Department of Medicine, School of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Lars Eckmann
- Department of Medicine, School of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA.
| | - Kirkwood M Land
- Department of Biological Sciences, University of the Pacific, Stockton, CA, USA
| | - Lisa A Wrischnik
- Department of Biological Sciences, University of the Pacific, Stockton, CA, USA
| |
Collapse
|
21
|
Bhabak KP, Bhowmick D. Synthetic strategies of gold(I)-selenolates from ortho-substituted diaryl diselenides via selenol and selenenyl sulfide intermediates. Inorganica Chim Acta 2016. [DOI: 10.1016/j.ica.2016.06.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
22
|
Maftei E, Maftei CV, Jones PG, Freytag M, Franz MH, Kelter G, Fiebig HH, Tamm M, Neda I. Trifluoromethylpyridine-SubstitutedN-Heterocyclic Carbenes Related to Natural Products: Synthesis, Structure, and Potential Antitumor Activity of some Corresponding Gold(I), Rhodium(I), and Iridium(I) Complexes. Helv Chim Acta 2016. [DOI: 10.1002/hlca.201500529] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Elena Maftei
- Institut für Anorganische und Analytische Chemie; Technische Universität Carola Wilhelmina; Hagenring 30 DE-38106 Braunschweig
- Institutul National de Cercetare Dezvoltare pentru Electrochimie si Materie Condensata; Str. Dr. A. Paunescu Podeanu Nr. 144 RO-300569 Timisoara
| | - Catalin V. Maftei
- Institut für Anorganische und Analytische Chemie; Technische Universität Carola Wilhelmina; Hagenring 30 DE-38106 Braunschweig
- Institutul National de Cercetare Dezvoltare pentru Electrochimie si Materie Condensata; Str. Dr. A. Paunescu Podeanu Nr. 144 RO-300569 Timisoara
| | - Peter G. Jones
- Institut für Anorganische und Analytische Chemie; Technische Universität Carola Wilhelmina; Hagenring 30 DE-38106 Braunschweig
| | - Matthias Freytag
- Institut für Anorganische und Analytische Chemie; Technische Universität Carola Wilhelmina; Hagenring 30 DE-38106 Braunschweig
| | - M. Heiko Franz
- Institutul National de Cercetare Dezvoltare pentru Electrochimie si Materie Condensata; Str. Dr. A. Paunescu Podeanu Nr. 144 RO-300569 Timisoara
- InnoChemTech GmbH; Hagenring 30 DE-38106 Braunschweig
| | | | | | - Matthias Tamm
- Institut für Anorganische und Analytische Chemie; Technische Universität Carola Wilhelmina; Hagenring 30 DE-38106 Braunschweig
| | - Ion Neda
- Institut für Anorganische und Analytische Chemie; Technische Universität Carola Wilhelmina; Hagenring 30 DE-38106 Braunschweig
- Institutul National de Cercetare Dezvoltare pentru Electrochimie si Materie Condensata; Str. Dr. A. Paunescu Podeanu Nr. 144 RO-300569 Timisoara
| |
Collapse
|
23
|
|
24
|
Braunschweig H, Ewing WC, Kramer T, Mattock JD, Vargas A, Werner C. Organometallic Probe for the Electronics of Base-Stabilized Group 11 Metal Cations. Chemistry 2015; 21:12347-56. [DOI: 10.1002/chem.201500788] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Indexed: 11/10/2022]
|
25
|
Maftei CV, Fodor E, Jones PG, Freytag M, Franz MH, Kelter G, Fiebig HH, Tamm M, Neda I. N-heterocyclic carbenes (NHC) with 1,2,4-oxadiazole-substituents related to natural products: synthesis, structure and potential antitumor activity of some corresponding gold(I) and silver(I) complexes. Eur J Med Chem 2015; 101:431-41. [PMID: 26185007 DOI: 10.1016/j.ejmech.2015.06.053] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Revised: 05/11/2015] [Accepted: 06/28/2015] [Indexed: 01/06/2023]
Abstract
This work presents the synthesis, characterization and application of eleven new gold (I) complexes 13-23 with 1,2,4-oxadiazole-containing N-heterocyclic carbene (NHC) ligands and of the NHC silver(I) complex 24. The 1,2,4-oxadiazole unit, which can be found in a variety of biologically active natural products such as phidianidines or quisqualic acid, was incorporated, along with a variety of other biologically active moieties (anthracene, indole, 2-pyridine, 2,3,4,5-tetra-O-acetyl-D-glucopyranose, quincorine and quincoridine), in order to change the lipophilicity of the complexes, so that the transport of the active units (M-NHC) though the cell wall barrier is facilitated. The biological activity of the complexes was investigated. In vitro assessment of anti-tumor activity in a panel of 12 human tumor cell lines by a monolayer assay revealed impressive potency (mean IC50 < 0.1 μM) and tumor selectivity for 6 compounds, with individual IC50 values in the low nanomolar range. The solid state structures of compounds 13, 14, 15, 17, 18, 19 and 24 were determined by X-ray diffraction analyses.
Collapse
Affiliation(s)
- Catalin V Maftei
- Institut für Anorganische und Analytische Chemie, Technische Universität Braunschweig, Hagenring 30, D-38106 Braunschweig, Germany; Institutul National de Cercetare Dezvoltare pentru Electrochimie si Materie Condensata, Str. Dr. A. Paunescu Podeanu, Nr. 144, Ro-300569 Timisoara, Romania
| | - Elena Fodor
- Institut für Anorganische und Analytische Chemie, Technische Universität Braunschweig, Hagenring 30, D-38106 Braunschweig, Germany; Institutul National de Cercetare Dezvoltare pentru Electrochimie si Materie Condensata, Str. Dr. A. Paunescu Podeanu, Nr. 144, Ro-300569 Timisoara, Romania
| | - Peter G Jones
- Institut für Anorganische und Analytische Chemie, Technische Universität Braunschweig, Hagenring 30, D-38106 Braunschweig, Germany
| | - Matthias Freytag
- Institut für Anorganische und Analytische Chemie, Technische Universität Braunschweig, Hagenring 30, D-38106 Braunschweig, Germany
| | - M Heiko Franz
- InnoChemTech GmbH, Hagenring 30, D-38106 Braunschweig, Germany; Institutul National de Cercetare Dezvoltare pentru Electrochimie si Materie Condensata, Str. Dr. A. Paunescu Podeanu, Nr. 144, Ro-300569 Timisoara, Romania
| | - Gerhard Kelter
- Oncotest GmbH, Am Flughafen 12-14, D-79108 Freiburg, Germany
| | | | - Matthias Tamm
- Institut für Anorganische und Analytische Chemie, Technische Universität Braunschweig, Hagenring 30, D-38106 Braunschweig, Germany.
| | - Ion Neda
- Institut für Anorganische und Analytische Chemie, Technische Universität Braunschweig, Hagenring 30, D-38106 Braunschweig, Germany; Institutul National de Cercetare Dezvoltare pentru Electrochimie si Materie Condensata, Str. Dr. A. Paunescu Podeanu, Nr. 144, Ro-300569 Timisoara, Romania.
| |
Collapse
|
26
|
Protein tyrosine phosphatase-PEST and β8 integrin regulate spatiotemporal patterns of RhoGDI1 activation in migrating cells. Mol Cell Biol 2015; 35:1401-13. [PMID: 25666508 DOI: 10.1128/mcb.00112-15] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Directional cell motility is essential for normal development and physiology, although how motile cells spatiotemporally activate signaling events remains largely unknown. Here, we have characterized an adhesion and signaling unit comprised of protein tyrosine phosphatase (PTP)-PEST and the extracellular matrix (ECM) adhesion receptor β8 integrin that plays essential roles in directional cell motility. β8 integrin and PTP-PEST form protein complexes at the leading edge of migrating cells and balance patterns of Rac1 and Cdc42 signaling by controlling the subcellular localization and phosphorylation status of Rho GDP dissociation inhibitor 1 (RhoGDI1). Translocation of Src-phosphorylated RhoGDI1 to the cell's leading edge promotes local activation of Rac1 and Cdc42, whereas dephosphorylation of RhoGDI1 by integrin-bound PTP-PEST promotes RhoGDI1 release from the membrane and sequestration of inactive Rac1/Cdc42 in the cytoplasm. Collectively, these data reveal a finely tuned regulatory mechanism for controlling signaling events at the leading edge of directionally migrating cells.
Collapse
|
27
|
Zou T, Lum CT, Lok CN, Zhang JJ, Che CM. Chemical biology of anticancer gold(iii) and gold(i) complexes. Chem Soc Rev 2015; 44:8786-801. [DOI: 10.1039/c5cs00132c] [Citation(s) in RCA: 420] [Impact Index Per Article: 46.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Anticancer gold complexes, including their mechanisms of action and the approaches adopted to improve the anticancer efficiency are described.
Collapse
Affiliation(s)
- Taotao Zou
- State Key Laboratory of Synthetic Chemistry
- Institute of Molecular Functional Materials
- Chemical Biology Centre and Department of Chemistry
- The University of Hong Kong
- Hong Kong
| | - Ching Tung Lum
- State Key Laboratory of Synthetic Chemistry
- Institute of Molecular Functional Materials
- Chemical Biology Centre and Department of Chemistry
- The University of Hong Kong
- Hong Kong
| | - Chun-Nam Lok
- State Key Laboratory of Synthetic Chemistry
- Institute of Molecular Functional Materials
- Chemical Biology Centre and Department of Chemistry
- The University of Hong Kong
- Hong Kong
| | - Jing-Jing Zhang
- State Key Laboratory of Synthetic Chemistry
- Institute of Molecular Functional Materials
- Chemical Biology Centre and Department of Chemistry
- The University of Hong Kong
- Hong Kong
| | - Chi-Ming Che
- State Key Laboratory of Synthetic Chemistry
- Institute of Molecular Functional Materials
- Chemical Biology Centre and Department of Chemistry
- The University of Hong Kong
- Hong Kong
| |
Collapse
|
28
|
Kriechbaum M, List M, Himmelsbach M, Redhammer GJ, Monkowius U. Peptide Coupling between Amino Acids and the Carboxylic Acid of a Functionalized Chlorido-gold(I)-phosphane. Inorg Chem 2014; 53:10602-10. [DOI: 10.1021/ic5017142] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
| | | | | | - Günther J. Redhammer
- Materialwissenschaften
und Physik, Abteilung für Mineralogie, Paris-Lodron Universität Salzburg, Hellabrunner Strasse 34, 5020 Salzburg, Austria
| | | |
Collapse
|
29
|
Thorson MK, Puerta DT, Cohen SM, Barrios AM. Inhibition of the lymphoid tyrosine phosphatase: the effect of zinc(II) ions and chelating ligand fragments on enzymatic activity. Bioorg Med Chem Lett 2014; 24:4019-22. [PMID: 24997687 PMCID: PMC4497560 DOI: 10.1016/j.bmcl.2014.06.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Revised: 06/04/2014] [Accepted: 06/06/2014] [Indexed: 11/22/2022]
Abstract
A 96-member chelator fragment library (CFL-1.1) was screened to identify inhibitors of the lymphoid tyrosine phosphatase in the absence and presence of zinc acetate. Fragments that inhibit LYP activity more potently in the presence of zinc, fragments that rescue LYP activity in the presence of inhibitory concentrations of zinc, and fragments that inhibit LYP activity independent of zinc concentration were identified. Of these, 1,2-dihydroxynaphthalene was the most potent inhibitor with an IC50 value of 2.52±0.06 μM after 2 h of incubation. LYP inhibition by 1,2-dihydroxynaphthalene was very similar to inhibition by 1,2-naphthoquinone (IC50=1.10±0.03 µM), indicating that the oxidized quinone species is likely the active inhibitor. The inhibition was time-dependent, consistent with covalent modification of the enzyme.
Collapse
Affiliation(s)
- Megan K Thorson
- Department of Medicinal Chemistry, University of Utah, 30 South 2000 East, Salt Lake City, UT 84112, USA
| | - David T Puerta
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA 92093, USA
| | - Seth M Cohen
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA 92093, USA
| | - Amy M Barrios
- Department of Medicinal Chemistry, University of Utah, 30 South 2000 East, Salt Lake City, UT 84112, USA.
| |
Collapse
|
30
|
Abstract
SIGNIFICANCE Protein tyrosine phosphatases (PTPs) play essential roles in controlling cell proliferation, differentiation, communication, and adhesion. The dysregulated activities of PTPs are involved in the pathogenesis of a number of human diseases such as cancer, diabetes, and autoimmune diseases. RECENT ADVANCES Many PTPs have emerged as potential new targets for novel drug discovery. PTP inhibitors have attracted much attention. Many PTP inhibitors have been developed. Some of them have been proven to be efficient in lowering blood glucose levels in vivo or inhibiting tumor xenograft growth. CRITICAL ISSUES Some metal ions and metal complexes potently inhibit PTPs. The metal atoms within metal complexes play an important role in PTP binding, while ligand structures influence the inhibitory potency and selectivity. Some metal complexes can penetrate the cell membrane and selectively bind to their targeting PTPs, enhancing the phosphorylation of the related substrates and influencing cellular metabolism. PTP inhibition is potentially involved in the pathophysiological and toxicological processes of metals and some PTPs may be cellular targets of certain metal-based therapeutic agents. FUTURE DIRECTIONS Investigating the structural basis of the interactions between metal complexes and PTPs would facilitate a comprehensive understanding of the structure-activity relationship and accelerate the development of promising metal-based drugs targeting specific PTPs.
Collapse
Affiliation(s)
- Liping Lu
- Key Laboratory of Chemical Biology and Molecular Engineering of the Education Ministry, Institute of Molecular Science, Shanxi University , Taiyuan, People's Republic of China
| | | |
Collapse
|
31
|
Sharlow ER, Leimgruber S, Murray S, Lira A, Sciotti RJ, Hickman M, Hudson T, Leed S, Caridha D, Barrios AM, Close D, Grögl M, Lazo JS. Auranofin is an apoptosis-simulating agent with in vitro and in vivo anti-leishmanial activity. ACS Chem Biol 2014; 9:663-72. [PMID: 24328400 DOI: 10.1021/cb400800q] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Cutaneous leishmaniasis remains ignored in therapeutic drug discovery programs worldwide. This is mainly because cutaneous leishmaniasis is frequently a disease of impoverished populations in countries where funds are limited for research and patient care. However, the health burden of individuals in endemic areas mandates readily available, effective, and safe treatments. Of the existing cutaneous leishmaniasis therapeutics, many are growth inhibitory to Leishmania parasites, potentially creating dormant parasite reservoirs that can be activated when host immunity is compromised, enabling the reemergence of cutaneous leishmaniasis lesions or worse spread of Leishmania parasites to other body sites. To accelerate the identification and development of novel cutaneous leishmaniasis therapeutics, we designed an integrated in vitro and in vivo screening platform that incorporated multiple Leishmania life cycles and species and probed a focused library of pharmaceutically active compounds. The objective of this phenotypic drug discovery platform was the identification and prioritization of bona fide cytotoxic chemotypes toward Leishmania parasites. We identified the Food and Drug Administration-approved drug auranofin, a known inhibitor of Leishmania promastigote growth, as a potent cytotoxic anti-leishmanial agent and inducer of apoptotic-like death in promastigotes. Significantly, the anti-leishmanial activity of auranofin transferred to cell-based amastigote assays as well as in vivo murine models. With appropriate future investigation, these data may provide the foundation for potential exploitation of gold(I)-based complexes as chemical tools or the basis of therapeutics for leishmaniasis. Thus, auranofin may represent a prototype drug that can be used to identify signaling pathways within the parasite and host cell critical for parasite growth and survival.
Collapse
Affiliation(s)
| | | | | | | | - Richard J. Sciotti
- Department
of Discovery, Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Mark Hickman
- Department
of Discovery, Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Thomas Hudson
- Department
of Discovery, Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Susan Leed
- Department
of Discovery, Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Diana Caridha
- Department
of Discovery, Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Amy M. Barrios
- Department
of Medicinal Chemistry, University of Utah, Salt Lake City, Utah 84112, United States
| | - David Close
- Department
of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Max Grögl
- Department
of Discovery, Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | | |
Collapse
|
32
|
Ahmed VF, Bottini N, Barrios AM. Covalent inhibition of the lymphoid tyrosine phosphatase. ChemMedChem 2014; 9:296-9. [PMID: 24403103 PMCID: PMC4096870 DOI: 10.1002/cmdc.201300404] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Indexed: 01/14/2023]
Abstract
Covalent inhibitors of lymphoid tyrosine phosphatase (LYP) were identified from a screen of the NIH Molecular Libraries Small Molecules Repository (MLSMR). Both of the two lead compounds identified have phosphotyrosine-mimetic benzoic acid moieties as well as electrophilic acrylonitrile groups. Inhibition kinetics of both compounds are consistent with covalent modification of the enzyme, with nanomolar KI and reciprocal millisecond kinact values, representing the best efficiency ratios (kinact /KI ) among currently reported covalent LYP inhibitors. Covalent inhibitors can provide longer efficacy and better selectivity than more conventional noncovalent inhibitors, and these lead compounds are an important step toward the development of protein tyrosine phosphatase (PTP)-targeted covalent therapeutic compounds.
Collapse
Affiliation(s)
- Vanessa F. Ahmed
- Department of Medicinal Chemistry, University of Utah, Salt Lake City, UT 84112, USA
| | - Nunzio Bottini
- Division of Cellular Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | - Amy M. Barrios
- Department of Medicinal Chemistry, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
33
|
Hristova YR, Kemper B, Besenius P. Water-soluble Au(I) complexes, their synthesis and applications. Tetrahedron 2013. [DOI: 10.1016/j.tet.2013.09.096] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
34
|
Abbehausen C, Peterson EJ, de Paiva REF, Corbi PP, Formiga ALB, Qu Y, Farrell NP. Gold(I)-Phosphine-N-Heterocycles: Biological Activity and Specific (Ligand) Interactions on the C-Terminal HIVNCp7 Zinc Finger. Inorg Chem 2013; 52:11280-7. [DOI: 10.1021/ic401535s] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Camilla Abbehausen
- Institute of Chemistry, University of Campinas—UNICAMP, P.O. Box
6154, CEP 13083-970, Campinas, São Paulo, Brazil
| | - Erica J. Peterson
- Goodwin Laboratory, Massey Cancer Center, Virginia Commonwealth University, 401 College Street, Richmond, Virginia 23298, United States
| | - Raphael E. F. de Paiva
- Institute of Chemistry, University of Campinas—UNICAMP, P.O. Box
6154, CEP 13083-970, Campinas, São Paulo, Brazil
| | - Pedro P. Corbi
- Institute of Chemistry, University of Campinas—UNICAMP, P.O. Box
6154, CEP 13083-970, Campinas, São Paulo, Brazil
| | - André L. B. Formiga
- Institute of Chemistry, University of Campinas—UNICAMP, P.O. Box
6154, CEP 13083-970, Campinas, São Paulo, Brazil
| | - Yun Qu
- Goodwin Laboratory, Massey Cancer Center, Virginia Commonwealth University, 401 College Street, Richmond, Virginia 23298, United States
- Department of Chemistry, Virginia Commonwealth University, 1001 W. Main Street, Richmond, Virginia 23284-2006, United States
| | - Nicholas P. Farrell
- Goodwin Laboratory, Massey Cancer Center, Virginia Commonwealth University, 401 College Street, Richmond, Virginia 23298, United States
- Department of Chemistry, Virginia Commonwealth University, 1001 W. Main Street, Richmond, Virginia 23284-2006, United States
| |
Collapse
|
35
|
Kulkarni RA, Vellore NA, Bliss MR, Stanford SM, Falk MD, Bottini N, Baron R, Barrios AM. Substrate selection influences molecular recognition in a screen for lymphoid tyrosine phosphatase inhibitors. Chembiochem 2013; 14:1640-7. [PMID: 23956195 PMCID: PMC3874405 DOI: 10.1002/cbic.201300273] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Indexed: 11/09/2022]
Abstract
Assay design is an important variable that influences the outcome of an inhibitor screen. Here, we have investigated the hypothesis that protein tyrosine phosphatase inhibitors with improved biological activity could be identified from a screen by using a biologically relevant peptide substrate, rather than traditional phosphotyrosine mimetic substrates. A 2000-member library of drugs and drug-like compounds was screened for inhibitors of lymphoid tyrosine phosphatase (LYP) by using both a peptide substrate (Ac-ARLIEDNE-pCAP-TAREG-NH₂, peptide 1) and a small-molecule phosphotyrosine mimetic substrate (difluoromethyl umbelliferyl phosphate, DiFMUP). The results demonstrate that compounds that inhibited enzyme activity on the peptide substrate had greater biological activity than compounds that only inhibited enzyme activity on DiFMUP. Finally, epigallocatechin-3,5-digallate was identified as the most potent inhibitor of lymphoid tyrosine phosphatase activity to date, with an IC₅₀ of 50 nM and significant activity in T-cells. Molecular docking simulations provided a first model for binding of this potent inhibitor to LYP; this will constitute the platform for ongoing lead optimization efforts.
Collapse
Affiliation(s)
| | - Nadeem A. Vellore
- Department of Medicinal Chemistry, University of Utah, Salt Lake City, UT 84112
- Henry Eyring Center for Theoretical Chemistry, University of Utah, Salt Lake City, UT 84112
| | - Matthew R. Bliss
- Division of Cellular Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037
| | - Stephanie M. Stanford
- Division of Cellular Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037
| | - Matthew D. Falk
- Division of Cellular Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037
| | - Nunzio Bottini
- Division of Cellular Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037
| | - Riccardo Baron
- Department of Medicinal Chemistry, University of Utah, Salt Lake City, UT 84112
- Henry Eyring Center for Theoretical Chemistry, University of Utah, Salt Lake City, UT 84112
| | - Amy M. Barrios
- Department of Medicinal Chemistry, University of Utah, Salt Lake City, UT 84112
| |
Collapse
|
36
|
Kulkarni RA, Stanford SM, Vellore NA, Krishnamurthy D, Bliss MR, Baron R, Bottini N, Barrios AM. Thiuram disulfides as pseudo-irreversible inhibitors of lymphoid tyrosine phosphatase. ChemMedChem 2013; 8:1561-8. [PMID: 23873737 PMCID: PMC3863632 DOI: 10.1002/cmdc.201300215] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Revised: 06/21/2013] [Indexed: 12/24/2022]
Abstract
We screened a small library of thiuram disulfides for inhibition of lymphoid tyrosine phosphatase (LYP) activity. The parent thiuram disulfide, disulfiram, inhibited LYP activity in vitro and in Jurkat T cells, whereas diethyldithiocarbamate failed to inhibit LYP at the concentrations tested. Compound 13, an N-(2-thioxothiazolidin-4-one) analogue, was found to be the most potent LYP inhibitor in this series, with an IC50 value of 3 μM. Compound 13 inhibits LYP pseudo-irreversibly, as evidenced by the time-dependence of inhibition, with a K(i) value of 1.1 μM and a k(inact) value of 0.004 s⁻¹. The inhibition of LYP by compound 13 could not be reversed significantly by incubation with glutathione or by prolonged dialysis, but could be partially reversed by incubation with dithiothreitol. Compound 13 also inhibited LYP activity in Jurkat T cells.
Collapse
Affiliation(s)
- Rhushikesh A Kulkarni
- Department of Medicinal Chemistry, University of Utah, Salt Lake City, UT 84112, USA
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Integrating virtual and biochemical screening for protein tyrosine phosphatase inhibitor discovery. Methods 2013; 65:219-28. [PMID: 23969317 DOI: 10.1016/j.ymeth.2013.08.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Revised: 08/09/2013] [Accepted: 08/13/2013] [Indexed: 12/14/2022] Open
Abstract
Protein tyrosine phosphatases (PTPs) represent an important class of enzymes that mediate signal transduction and control diverse aspects of cell behavior. The importance of their activity is exemplified by their significant contribution to disease etiology with over half of all human PTP genes implicated in at least one disease. Small molecule inhibitors targeting individual PTPs are important biological tools, and are needed to fully characterize the function of these enzymes. Moreover, potent and selective PTP inhibitors hold the promise to transform the treatment of many diseases. While numerous methods exist to develop PTP-directed small molecules, we have found that complimentary use of both virtual (in silico) and biochemical (in vitro) screening approaches expedite compound identification and drug development. Here, we summarize methods pertinent to our work and others. Focusing on specific challenges and successes we have experienced, we discuss the considerable caution that must be taken to avoid enrichment of inhibitors that function by non-selective oxidation. We also discuss the utility of using "open" PTP structures to identify active-site directed compounds, a rather unconventional choice for virtual screening. When integrated closely, virtual and biochemical screening can be used in a productive workflow to identify small molecules targeting PTPs.
Collapse
|
38
|
Synthesis and evaluation of copper complexes of Schiff-base condensates from 5-substituted-2-hydroxybenzaldehyde and 2-substituted-benzenamine as selective inhibitors of protein tyrosine phosphatases. Inorganica Chim Acta 2013. [DOI: 10.1016/j.ica.2013.05.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
39
|
Stanford SM, Krishnamurthy D, Kulkarni RA, Karver CE, Bruenger E, Walker LM, Ma CT, Chung TDY, Sergienko E, Bottini N, Barrios AM. pCAP-based peptide substrates: the new tool in the box of tyrosine phosphatase assays. Methods 2013; 65:165-74. [PMID: 23886911 DOI: 10.1016/j.ymeth.2013.07.022] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Revised: 07/10/2013] [Accepted: 07/13/2013] [Indexed: 10/26/2022] Open
Abstract
Robust, facile high throughput assays based on non-peptidic probes are available to detect the enzyme activity of protein tyrosine phosphatases. However, these assays cannot replace the use of peptide-based probes in many applications; for example when a closer mimic of the physiological target is desired or in substrate profiling expeditions. Phosphotyrosine peptides are often used in these assays, but their use is complicated by either poor sensitivity or the need for indirect detection methods, among other pitfalls. Novel peptide-based probes for protein tyrosine phosphatases are needed to replace phosphotyrosine peptides and accelerate the field of tyrosine phosphatase substrate profiling. Here we review a type of peptidic probe for tyrosine phosphatases, which is based on the incorporation of the phosphotyrosine-mimic phosphocoumaryl amino propionic acid (pCAP) into peptides. The resulting fluorogenic pCAP peptides are dephosphorylated by tyrosine phosphatases with similar efficiency as the homologous phosphotyrosine peptides. pCAP peptides outperform phosphotyrosine peptides, providing an assay that is as robust, sensitive and facile as the non-peptidic fluorogenic probes on the market. Finally the use of pCAP can expand the range of phosphatase assays, facilitating the investigation of multiphosphorylated peptides and providing an in-gel assay for phosphatase activity.
Collapse
Affiliation(s)
- Stephanie M Stanford
- Division of Cellular Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | - Divya Krishnamurthy
- Department of Chemistry, University of Southern California, Los Angeles, CA 90089, USA; Department of Medicinal Chemistry, University of Utah, Salt Lake City, UT 84112, USA
| | - Rhushikesh A Kulkarni
- Department of Medicinal Chemistry, University of Utah, Salt Lake City, UT 84112, USA
| | - Caitlin E Karver
- Department of Chemistry, University of Southern California, Los Angeles, CA 90089, USA; Department of Medicinal Chemistry, University of Utah, Salt Lake City, UT 84112, USA
| | - Eveline Bruenger
- Department of Medicinal Chemistry, University of Utah, Salt Lake City, UT 84112, USA
| | - Logan M Walker
- Division of Cellular Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | - Chen-Ting Ma
- Conrad Prebys Center for Chemical Genomics, Sanford
- Burnham Medical Research Institute, La Jolla, CA 92037, USA
| | - Thomas D Y Chung
- Conrad Prebys Center for Chemical Genomics, Sanford
- Burnham Medical Research Institute, La Jolla, CA 92037, USA
| | - Eduard Sergienko
- Conrad Prebys Center for Chemical Genomics, Sanford
- Burnham Medical Research Institute, La Jolla, CA 92037, USA
| | - Nunzio Bottini
- Division of Cellular Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA.
| | - Amy M Barrios
- Department of Medicinal Chemistry, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
40
|
He Y, Liu S, Menon A, Stanford S, Oppong E, Gunawan AM, Wu L, Wu DJ, Barrios AM, Bottini N, Cato ACB, Zhang ZY. A potent and selective small-molecule inhibitor for the lymphoid-specific tyrosine phosphatase (LYP), a target associated with autoimmune diseases. J Med Chem 2013; 56:4990-5008. [PMID: 23713581 PMCID: PMC3711248 DOI: 10.1021/jm400248c] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Lymphoid-specific tyrosine phosphatase (LYP), a member of the protein tyrosine phosphatase (PTP) family of signaling enzymes, is associated with a broad spectrum of autoimmune diseases. Herein we describe our structure-based lead optimization efforts within a 6-hydroxy-benzofuran-5-carboxylic acid series culminating in the identification of compound 8b, a potent and selective inhibitor of LYP with a K(i) value of 110 nM and more than 9-fold selectivity over a large panel of PTPs. The structure of LYP in complex with 8b was obtained by X-ray crystallography, providing detailed information about the molecular recognition of small-molecule ligands binding LYP. Importantly, compound 8b possesses highly efficacious cellular activity in both T- and mast cells and is capable of blocking anaphylaxis in mice. Discovery of 8b establishes a starting point for the development of clinically useful LYP inhibitors for treating a wide range of autoimmune disorders.
Collapse
Affiliation(s)
- Yantao He
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, 635 Barnhill Drive, Indianapolis, Indiana 46202, USA
| | - Sijiu Liu
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, 635 Barnhill Drive, Indianapolis, Indiana 46202, USA
| | - Ambili Menon
- Karlsruhe Institute of Technology, Institute of Toxicology and Genetics, Hermann-von- Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
| | - Stephanie Stanford
- Division of Cellular Biology, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Emmanuel Oppong
- Karlsruhe Institute of Technology, Institute of Toxicology and Genetics, Hermann-von- Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
| | - Andrea M. Gunawan
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, 635 Barnhill Drive, Indianapolis, Indiana 46202, USA
| | - Li Wu
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, 635 Barnhill Drive, Indianapolis, Indiana 46202, USA
| | - Dennis J. Wu
- Division of Cellular Biology, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Amy M. Barrios
- Department of Medicinal Chemistry, University of Utah, Salt Lake City, UT 84112, USA
| | - Nunzio Bottini
- Division of Cellular Biology, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Andrew C. B. Cato
- Karlsruhe Institute of Technology, Institute of Toxicology and Genetics, Hermann-von- Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
| | - Zhong-Yin Zhang
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, 635 Barnhill Drive, Indianapolis, Indiana 46202, USA
| |
Collapse
|
41
|
Liu W, Gust R. Metal N-heterocyclic carbene complexes as potential antitumor metallodrugs. Chem Soc Rev 2013; 42:755-73. [PMID: 23147001 DOI: 10.1039/c2cs35314h] [Citation(s) in RCA: 586] [Impact Index Per Article: 53.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Wukun Liu
- Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Str 2+4, 14195 Berlin, Germany
| | | |
Collapse
|
42
|
Ong JX, Yap CW, Ang WH. Rational Design of Selective Organoruthenium Inhibitors of Protein Tyrosine Phosphatase 1B. Inorg Chem 2012; 51:12483-92. [DOI: 10.1021/ic301884j] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Jun Xiang Ong
- Department of Chemistry, National University of Singapore, 3 Science Drive 3,
Singapore 117543
| | - Chun Wei Yap
- Department
of Pharmacy, National University of Singapore, 18 Science Drive
4, Singapore 117543
| | - Wee Han Ang
- Department of Chemistry, National University of Singapore, 3 Science Drive 3,
Singapore 117543
| |
Collapse
|
43
|
He R, Zeng LF, He Y, Zhang S, Zhang ZY. Small molecule tools for functional interrogation of protein tyrosine phosphatases. FEBS J 2012; 280:731-50. [PMID: 22816879 DOI: 10.1111/j.1742-4658.2012.08718.x] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The importance of protein tyrosine phosphatases (PTPs) in the regulation of cellular signalling is well established. Malfunction of PTP activity is also known to be associated with cancer, metabolic syndromes and autoimmune disorders, as well as neurodegenerative and infectious diseases. However, a detailed understanding of the roles played by the PTPs in normal physiology and in pathogenic conditions has been hampered by the absence of PTP-specific small molecule agents. In addition, the therapeutic benefits of modulating this target class are underexplored as a result of a lack of suitable chemical probes. Potent and specific PTP inhibitors could significantly facilitate functional analysis of the PTPs in complex cellular signal transduction pathways and may constitute valuable therapeutics in the treatment of several human diseases. We highlight the current challenges to and opportunities for developing PTP-specific small molecule agents. We also review available selective small molecule inhibitors developed for a number of PTPs, including PTP1B, TC-PTP, SHP2, lymphoid-specific tyrosine phosphatase, haematopoietic protein tyrosine phosphatase, CD45, PTPβ, PTPγ, PTPRO, Vaccinia H1-related phosphatase, mitogen-activated protein kinase phosphatase-1, mitogen-activated protein kinase phosphatase-3, Cdc25, YopH, mPTPA and mPTPB.
Collapse
Affiliation(s)
- Rongjun He
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | | | | | | |
Collapse
|
44
|
High-throughput screen using a single-cell tyrosine phosphatase assay reveals biologically active inhibitors of tyrosine phosphatase CD45. Proc Natl Acad Sci U S A 2012; 109:13972-7. [PMID: 22891353 DOI: 10.1073/pnas.1205028109] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Many cellular signaling events are regulated by tyrosine phosphorylation and mediated by the opposing actions of protein tyrosine kinases and phosphatases. Protein tyrosine phosphatases are emerging as drug targets, but poor cell permeability of inhibitors has limited the development of drugs targeting these enzymes [Tautz L, et al. (2006) Expert Opin Ther Targets 10:157-177]. Here we developed a method to monitor tyrosine phosphatase activity at the single-cell level and applied it to the identification of cell-permeable inhibitors. The method takes advantage of the fluorogenic properties of phosphorylated coumaryl amino propionic acid (pCAP), an analog of phosphotyrosine, which can be incorporated into peptides. Once delivered into cells, pCAP peptides were dephosphorylated by protein tyrosine phosphatases, and the resulting cell fluorescence could be monitored by flow cytometry and high-content imaging. The robustness and sensitivity of the assay was validated using peptides preferentially dephosphorylated by CD45 and T-cell tyrosine phosphatase and available inhibitors of these two enzymes. The assay was applied to high-throughput screening for inhibitors of CD45, an important target for autoimmunity and infectious diseases [Hermiston ML, et al. (2003) Annu Rev Immunol 21:107-137]. We identified four CD45 inhibitors that showed activity in T cells and macrophages. These results indicate that our assay can be applied to primary screening for inhibitors of CD45 and of other protein tyrosine phosphatases to increase the yield of biologically active inhibitors.
Collapse
|
45
|
Meyer A, Bagowski CP, Kokoschka M, Stefanopoulou M, Alborzinia H, Can S, Vlecken DH, Sheldrick WS, Wölfl S, Ott I. Über die biologischen Eigenschaften von Alkinyl(phosphan)gold(I)-Komplexen. Angew Chem Int Ed Engl 2012. [DOI: 10.1002/ange.201202939] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
46
|
Meyer A, Bagowski CP, Kokoschka M, Stefanopoulou M, Alborzinia H, Can S, Vlecken DH, Sheldrick WS, Wölfl S, Ott I. On the Biological Properties of Alkynyl Phosphine Gold(I) Complexes. Angew Chem Int Ed Engl 2012; 51:8895-9. [DOI: 10.1002/anie.201202939] [Citation(s) in RCA: 144] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Revised: 05/17/2012] [Indexed: 01/20/2023]
|
47
|
Liu W, Bensdorf K, Proetto M, Hagenbach A, Abram U, Gust R. Synthesis, Characterization, and in Vitro Studies of Bis[1,3-diethyl-4,5-diarylimidazol-2-ylidene]gold(I/III) Complexes. J Med Chem 2012; 55:3713-24. [DOI: 10.1021/jm3000196] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Wukun Liu
- Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Strasse 2 + 4, 14195 Berlin, Germany
| | - Kerstin Bensdorf
- Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Strasse 2 + 4, 14195 Berlin, Germany
| | - Maria Proetto
- Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Strasse 2 + 4, 14195 Berlin, Germany
| | - Adelheid Hagenbach
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Fabeckstrasse 34-36, 14195 Berlin, Germany
| | - Ulrich Abram
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Fabeckstrasse 34-36, 14195 Berlin, Germany
| | - Ronald Gust
- Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Strasse 2 + 4, 14195 Berlin, Germany
- Institute of Pharmacy, University Innsbruck, Innrain 80/82, A-6020 Innsbruck, Austria
| |
Collapse
|
48
|
Jiang B, Shi D, Cui Y, Guo S. Design, Synthesis, and Biological Evaluation of Bromophenol Derivatives as Protein Tyrosine Phosphatase 1B Inhibitors. Arch Pharm (Weinheim) 2012; 345:444-53. [DOI: 10.1002/ardp.201100373] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2011] [Revised: 12/01/2011] [Accepted: 12/06/2011] [Indexed: 11/11/2022]
|
49
|
|
50
|
Han H, Lu L, Wang Q, Zhu M, Yuan C, Xing S, Fu X. Synthesis and evaluation of oxovanadium(iv) complexes of Schiff-base condensates from 5-substituted-2-hydroxybenzaldehyde and 2-substituted-benzenamine as selective inhibitors of protein tyrosine phosphatase 1B. Dalton Trans 2012; 41:11116-24. [DOI: 10.1039/c2dt30198a] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|