1
|
Yawson GK, Will MF, Huffman SE, Strandquist ET, Bothwell PJ, Oliver EB, Apuzzo CF, Platt DC, Weitzel CS, Jones MA, Ferrence GM, Hamaker CG, Webb MI. A Dual-Pronged Approach: A Ruthenium(III) Complex That Modulates Amyloid-β Aggregation and Disrupts Its Formed Aggregates. Inorg Chem 2022; 61:2733-2744. [PMID: 35102739 DOI: 10.1021/acs.inorgchem.1c01651] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Alzheimer's disease (AD) is a devastating neurological disorder for which soluble oligomers of the peptide amyloid-β (Aβ) are now recognized as the neurotoxic species. Metal-based therapeutics are uniquely suited to target Aβ, with ruthenium-based (Ru) complexes emerging as propitious candidates. Recently, azole-based Ru(III) complexes were observed to modulate the aggregation of Aβ in solution, where the inclusion of a primary amine proximal to the ligand coordination site improved the activity of the complexes. To advance these structure-activity relationships, a series of oxazole-based Ru complexes were prepared and evaluated for their ability to modulate Aβ aggregation. From these studies, a lead candidate, Oc, emerged that had superior activity relative to its azole predecessors in modulating the aggregation of soluble Aβ and diminishing its cytotoxicity. Further evaluation of Oc demonstrated its ability to disrupt formed Aβ aggregates, resulting in smaller amorphous species. Because altering both sides of the aggregation equilibrium for Aβ has not been previously suggested for metal-based complexes for AD, this work represents an exciting new avenue for improved therapeutic success.
Collapse
Affiliation(s)
- Gideon K Yawson
- Department of Chemistry, Illinois State University, Normal, Illinois 61790-4160, United States
| | - Mark F Will
- Department of Chemistry, Illinois State University, Normal, Illinois 61790-4160, United States
| | - Samantha E Huffman
- Department of Chemistry, Illinois State University, Normal, Illinois 61790-4160, United States
| | - Evan T Strandquist
- Department of Chemistry, Illinois State University, Normal, Illinois 61790-4160, United States
| | - Paige J Bothwell
- Core Microscope Facility, Department of Biological Sciences, Northern Illinois University, DeKalb, Illinois 60115, United States
| | - Ethan B Oliver
- Department of Chemistry, Illinois State University, Normal, Illinois 61790-4160, United States
| | - C Fiore Apuzzo
- Department of Chemistry, Illinois State University, Normal, Illinois 61790-4160, United States
| | - David C Platt
- Department of Chemistry, Illinois State University, Normal, Illinois 61790-4160, United States
| | - Christopher S Weitzel
- Department of Chemistry, Illinois State University, Normal, Illinois 61790-4160, United States
| | - Marjorie A Jones
- Department of Chemistry, Illinois State University, Normal, Illinois 61790-4160, United States
| | - Gregory M Ferrence
- Department of Chemistry, Illinois State University, Normal, Illinois 61790-4160, United States
| | - Christopher G Hamaker
- Department of Chemistry, Illinois State University, Normal, Illinois 61790-4160, United States
| | - Michael I Webb
- Department of Chemistry, Illinois State University, Normal, Illinois 61790-4160, United States
| |
Collapse
|
2
|
|
3
|
Davis J, Cetto A, Campbell M, Scoggins S, Stultz L, Hanson P. DMSO reduces the cytotoxicity of anticancer ruthenium complex KP1019 in yeast. MICROPUBLICATION BIOLOGY 2021; 2021:10.17912/micropub.biology.000436. [PMID: 34377963 PMCID: PMC8339913 DOI: 10.17912/micropub.biology.000436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/29/2021] [Accepted: 07/29/2021] [Indexed: 11/25/2022]
Abstract
Low solubility in aqueous solutions is a significant limitation of the otherwise promising anticancer ruthenium complex KP1019. In laboratory studies, this challenge is often overcome by using DMSO to help drive the drug into solution. Since DMSO was previously shown to alter the bioactivity of platinum-based chemotherapeutics, here we examine DMSO's effects on KP1019. Using Saccharomyces cerevisiae as a model organism, we apply multiple measures of growth inhibition to demonstrate that DMSO reduces the drug's toxicity. This reduction in bioactivity correlates with spectrophotometric changes consistent with DMSO-dependent increases in the stability of the KP1019 pro-drug. The impact of DMSO on the biology and chemistry of KP1019 suggests this solvent should not be used in studies of this and similar anticancer ruthenium complexes.
Collapse
Affiliation(s)
- Jonathan Davis
- Department of Biology, Furman University, Greenville, SC 29613, USA
| | - Anne Cetto
- Department of Chemistry, Birmingham-Southern College, Birmingham, AL 35254, USA
| | - Mary Campbell
- Department of Chemistry, Birmingham-Southern College, Birmingham, AL 35254, USA
| | - Seth Scoggins
- Department of Chemistry, Birmingham-Southern College, Birmingham, AL 35254, USA
| | - Laura Stultz
- Department of Chemistry, Birmingham-Southern College, Birmingham, AL 35254, USA
| | - Pamela Hanson
- Department of Biology, Furman University, Greenville, SC 29613, USA,
Correspondence to: Pamela Hanson ()
| |
Collapse
|
4
|
Qin LQ, Liang CJ, Zhou Z, Qin QP, Wei ZZ, Tan MX, Liang H. Mitochondria-localizing curcumin-cryptolepine Zn(II) complexes and their antitumor activity. Bioorg Med Chem 2021; 30:115948. [PMID: 33360578 DOI: 10.1016/j.bmc.2020.115948] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 12/03/2020] [Accepted: 12/06/2020] [Indexed: 01/08/2023]
Abstract
Many metal complexes are potent candidates as mitochondrial-targeting agents. In this study, four novel Zn(II) complexes, [Zn(BPQA)Cl2] (Zn1), [Zn(BPQA)(Curc)]Cl (Zn2), [Zn(PQA)Cl2] (Zn3), and [Zn(PQA)(Curc)]Cl (Zn4), containing N,N-bis(pyridin-2-ylmethyl)benzofuro[3,2-b]quinolin-11-amine (BPQA), N-(pyridin-2-ylmethyl)benzofuro[3,2-b]quinolin-11-amine (PQA), and curcumin (H-Curc) were synthesized. An MTT assay showed that Zn1-Zn4 had strong anticancer activities against SK-OV-3/DDP and T-24 tumor cells with IC50 values of 0.03-6.19 μM. Importantly, Zn1 and Zn2 displayed low toxicities against normal HL-7702 cells. Mechanism experiments demonstrated that probe Zn2 showed appreciable fluorescence in the red region of the spectrum, and substantial accumulation of Zn2 occurred in the mitochondria after treatment, indicating increases in Ca2+ and reactive oxygen species levels, loss of the mitochondrial membrane potential, and consequent induction of mitochondrial dysfunction at low concentrations. In addition, the probe Zn2 effectively (50.7%) inhibited the growth of T-24 bladder tumor cells in vivo. The probe Zn2 shows potential for use in cancer therapy while retaining the H-Curc as an imaging probe.
Collapse
Affiliation(s)
- Li-Qin Qin
- Guangxi Key Lab of Agricultural Resources Chemistry and Biotechnology, College of Chemistry and Food Science, Yulin Normal University, 1303 Jiaoyudong Road, Yulin 537000, PR China
| | - Chun-Jie Liang
- Guangxi Key Lab of Agricultural Resources Chemistry and Biotechnology, College of Chemistry and Food Science, Yulin Normal University, 1303 Jiaoyudong Road, Yulin 537000, PR China
| | - Zhen Zhou
- Guangxi Key Lab of Agricultural Resources Chemistry and Biotechnology, College of Chemistry and Food Science, Yulin Normal University, 1303 Jiaoyudong Road, Yulin 537000, PR China
| | - Qi-Pin Qin
- Guangxi Key Lab of Agricultural Resources Chemistry and Biotechnology, College of Chemistry and Food Science, Yulin Normal University, 1303 Jiaoyudong Road, Yulin 537000, PR China; State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmacy, Guangxi Normal University, 15 Yucai Road, Guilin 541004, PR China.
| | - Zu-Zhuang Wei
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, PR China.
| | - Ming-Xiong Tan
- Guangxi Key Lab of Agricultural Resources Chemistry and Biotechnology, College of Chemistry and Food Science, Yulin Normal University, 1303 Jiaoyudong Road, Yulin 537000, PR China
| | - Hong Liang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmacy, Guangxi Normal University, 15 Yucai Road, Guilin 541004, PR China.
| |
Collapse
|
5
|
Steel TR, Hartinger CG. Metalloproteomics for molecular target identification of protein-binding anticancer metallodrugs. Metallomics 2020; 12:1627-1636. [PMID: 33063808 DOI: 10.1039/d0mt00196a] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Proteomics has played an important role in elucidating the fundamental processes occuring in living cells. Translating these methods to metallodrug research ('metalloproteomics') has provided a means for molecular target identification of metal-based anticancer agents which should signifcantly advance the research field. In combination with biological assays, these techniques have enabled the mechanisms of action of metallodrugs to be linked to their interactions with molecular targets and aid understanding of their biological properties. Such investigations have profoundly increased our knowledge of the complex and dynamic nature of metallodrug-biomolecule interactions and have provided, at least for some compound types, a more detailed picture on their specific protein-binding patterns. This perspective highlights the progression of metallodrug proteomics research for the identification of non-DNA targets from standard analytical techniques to powerful metallodrug pull-down methods.
Collapse
Affiliation(s)
- Tasha R Steel
- School of Chemical Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.
| | | |
Collapse
|
6
|
Inhibition of DNA Repair Pathways and Induction of ROS Are Potential Mechanisms of Action of the Small Molecule Inhibitor BOLD-100 in Breast Cancer. Cancers (Basel) 2020; 12:cancers12092647. [PMID: 32947941 PMCID: PMC7563761 DOI: 10.3390/cancers12092647] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/08/2020] [Accepted: 09/10/2020] [Indexed: 12/16/2022] Open
Abstract
BOLD-100, a ruthenium-based complex, sodium trans-[tetrachloridobis (1H-indazole) ruthenate (III)] (also known as IT-139, NKP1339 or KP1339), is a novel small molecule drug that demonstrated a manageable safety profile at the maximum tolerated dose and modest antitumor activity in a phase I clinical trial. BOLD-100 has been reported to inhibit the upregulation of the endoplasmic reticulum stress sensing protein GRP78. However, response to BOLD-100 varies in different cancer models and the precise mechanism of action in high-response versus low-response cancer cells remains unclear. In vitro studies have indicated that BOLD-100 induces cytostatic rather than cytotoxic effects as a monotherapy. To understand BOLD-100-mediated signaling mechanism in breast cancer cells, we used estrogen receptor positive (ER+) MCF7 breast cancer cells to obtain gene-metabolite integrated models. At 100 μM, BOLD-100 significantly reduced cell proliferation and expression of genes involved in the DNA repair pathway. BOLD-100 also induced reactive oxygen species (ROS) and phosphorylation of histone H2AX, gamma-H2AX (Ser139), suggesting disruption of proper DNA surveillance. In estrogen receptor negative (ER-) breast cancer cells, combination of BOLD-100 with a PARP inhibitor, olaparib, induced significant inhibition of cell growth and xenografts and increased gamma-H2AX. Thus, BOLD-100 is a novel DNA repair pathway targeting agent and can be used with other chemotherapies in ER- breast cancer.
Collapse
|