1
|
Mom R, Mocquet V, Auguin D, Réty S. Aquaporin Modulation by Cations, a Review. Curr Issues Mol Biol 2024; 46:7955-7975. [PMID: 39194687 DOI: 10.3390/cimb46080470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 07/18/2024] [Accepted: 07/22/2024] [Indexed: 08/29/2024] Open
Abstract
Aquaporins (AQPs) are transmembrane channels initially discovered for their role in water flux facilitation through biological membranes. Over the years, a much more complex and subtle picture of these channels appeared, highlighting many other solutes accommodated by AQPs and a dense regulatory network finely tuning cell membranes' water permeability. At the intersection between several transduction pathways (e.g., cell volume regulation, calcium signaling, potassium cycling, etc.), this wide and ancient protein family is considered an important therapeutic target for cancer treatment and many other pathophysiologies. However, a precise and isoform-specific modulation of these channels function is still challenging. Among the modulators of AQPs functions, cations have been shown to play a significant contribution, starting with mercury being historically associated with the inhibition of AQPs since their discovery. While the comprehension of AQPs modulation by cations has improved, a unifying molecular mechanism integrating all current knowledge is still lacking. In an effort to extract general trends, we reviewed all known modulations of AQPs by cations to capture a first glimpse of this regulatory network. We paid particular attention to the associated molecular mechanisms and pinpointed the residues involved in cation binding and in conformational changes tied up to the modulation of the channel function.
Collapse
Affiliation(s)
- Robin Mom
- Laboratoire de Biologie et Modelisation de la Cellule, Ecole Normale Superieure de Lyon, CNRS, UMR 5239, Inserm, U1293, Universite Claude Bernard Lyon 1, 46 allee d'Italie, F-69364 Lyon, France
| | - Vincent Mocquet
- Laboratoire de Biologie et Modelisation de la Cellule, Ecole Normale Superieure de Lyon, CNRS, UMR 5239, Inserm, U1293, Universite Claude Bernard Lyon 1, 46 allee d'Italie, F-69364 Lyon, France
| | - Daniel Auguin
- Laboratoire de Physiologie, Ecologie et Environnement (P2E), UPRES EA 1207/USC INRAE-1328, UFR Sciences et Techniques, Université d'Orléans, F-45067 Orléans, France
| | - Stéphane Réty
- Laboratoire de Biologie et Modelisation de la Cellule, Ecole Normale Superieure de Lyon, CNRS, UMR 5239, Inserm, U1293, Universite Claude Bernard Lyon 1, 46 allee d'Italie, F-69364 Lyon, France
| |
Collapse
|
2
|
da Silva IV, Pimpão C, Paccetti-Alves I, Thomas SR, Barateiro A, Casini A, Soveral G. Blockage of aquaporin-3 peroxiporin activity by organogold compounds affects melanoma cell adhesion, proliferation and migration. J Physiol 2024; 602:3111-3129. [PMID: 38323926 DOI: 10.1113/jp284155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 01/23/2024] [Indexed: 02/08/2024] Open
Abstract
Aquaporin-3 (AQP3) is a membrane channel with dual aquaglyceroporin/peroxiporin activity, facilitating the diffusion of water, glycerol and H2O2 across cell membranes. AQP3 shows aberrant expression in melanoma and its role in cell adhesion, migration and proliferation is well described. Gold compounds were shown to modulate AQP3 activity with reduced associated toxicity, making them promising molecules for cancer therapy. In this study, we validated the phenotype resulting from AQP3-silencing of two melanoma cell lines, MNT-1 and A375, which resulted in decreased H2O2 permeability. Subsequently, the AQP3 inhibitory effect of a new series of organogold compounds derived from Auphen, a potent AQP3 inhibitor, was first evaluated in red blood cells (RBCs) that highly express AQP3, and then in HEK-293T cells with AQP3 overexpression to ascertain the compounds' specificity. The first screening in RBCs unveiled two organogold compounds as promising blockers of AQP3 permeability. Moderate reduction of glycerol permeability but drastic inhibition of H2O2 permeability was detected for some of the gold derivatives in both AQP3-overexpressing cells and human melanoma cell lines. Additionally, all compounds were effective in impairing cell adhesion, proliferation and migration, although in a cell type-dependent manner. In conclusion, our data show that AQP3 peroxiporin activity is crucial for melanoma progression and highlight organogold compounds as promising AQP3 inhibitors with implications in melanoma cell adhesion, proliferation and migration, unveiling their potential as anticancer drugs against AQP3-overexpressing tumours. KEY POINTS: AQP3 affects cellular redox balance. Gold compounds inhibit AQP3 permeability in melanoma cells. AQP3 is involved in cell adhesion, proliferation and migration of melanoma. Blockage of AQP3 peroxiporin activity impairs melanoma cell migration. Gold compounds are potential anticancer drug leads for AQP3-overexpressing cancers.
Collapse
Affiliation(s)
- Inês V da Silva
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal
- Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal
| | - Catarina Pimpão
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal
- Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal
| | - Inês Paccetti-Alves
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal
- Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal
| | - Sophie R Thomas
- Department of Chemistry, School of Natural Sciences, Technical University of Munich, Garching bei München, Germany
| | - Andreia Barateiro
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal
- Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal
| | - Angela Casini
- Department of Chemistry, School of Natural Sciences, Technical University of Munich, Garching bei München, Germany
| | - Graça Soveral
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal
- Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
3
|
Mu K, Kitts DD. Intestinal polyphenol antioxidant activity involves redox signaling mechanisms facilitated by aquaporin activity. Redox Biol 2023; 68:102948. [PMID: 37922763 PMCID: PMC10643476 DOI: 10.1016/j.redox.2023.102948] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 10/26/2023] [Indexed: 11/07/2023] Open
Abstract
Ascertaining whether dietary polyphenols evoke an antioxidant or prooxidant activity, which translates to a functional role required to maintain intestinal cell homeostasis continues to be an active and controversial area of research for food chemists and biochemists alike. We have proposed that the paradoxical function of polyphenols to autoxidize to generate H2O2 is a required first step in the capacity of some plant phenolics to function as intracellular antioxidants. This is based on the fact that cell redox homeostasis is achieved by a balance between H2O2 formation and subsequent outcomes of antioxidant systems function. Maintaining optimal extracellular and intracellular H2O2 concentrations is required for cell survival, since low levels are important to upregulate endogenous antioxidant capacity; whereas, concentrations that go beyond homeostatic control typically result in an inflammatory response, growth arrest, or eventual cell death. Aquaporins (AQPs) are a family of water channel membrane proteins that facilitate cellular transportation of water and other small molecule-derived solutes, such as H2O2, in all organisms. In the intestine, AQPs act as gatekeepers to regulate intracellular uptake of H2O2, generated from extracellular polyphenol autoxidation, thus enabling an intracellular cell signaling responses to mitigate onset of oxidative stress and intestinal inflammation. In this review, we highlight the potential role of AQPs to control important underlying mechanisms that define downstream regulation of intestinal redox homeostasis, specifically. It has been established that polyphenols that undergo oxidation to the quinone form, resulting in subsequent adduction to a thiol group on Keap1-Nrf2 complex, trigger Nrf2 activation and a cascade of indirect intracellular antioxidant effects. Here, we propose a similar mechanism that involves H2O2 generated from specific dietary polyphenols with a predisposition to undergo autoxidation. The ultimate bioactivity is regulated and expressed by AQP membrane function and thus, by extension, represents expression of an intracellular antioxidant chemoprotection mechanism.
Collapse
Affiliation(s)
- Kaiwen Mu
- Food Science, Food Nutrition and Health Program. Faculty of Land and Food System, The University of British Columbia, 2205 East Mall, Vancouver, B.C, V6T 1Z4, Canada
| | - David D Kitts
- Food Science, Food Nutrition and Health Program. Faculty of Land and Food System, The University of British Columbia, 2205 East Mall, Vancouver, B.C, V6T 1Z4, Canada.
| |
Collapse
|
4
|
Banerjee S, Smith IM, Hengen AC, Stroka KM. Methods for studying mammalian aquaporin biology. Biol Methods Protoc 2023; 8:bpad031. [PMID: 38046463 PMCID: PMC10689382 DOI: 10.1093/biomethods/bpad031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/29/2023] [Accepted: 11/09/2023] [Indexed: 12/05/2023] Open
Abstract
Aquaporins (AQPs), transmembrane water-conducting channels, have earned a great deal of scrutiny for their critical physiological roles in healthy and disease cell states, especially in the biomedical field. Numerous methods have been implemented to elucidate the involvement of AQP-mediated water transport and downstream signaling activation in eliciting whole cell, tissue, and organ functional responses. To modulate these responses, other methods have been employed to investigate AQP druggability. This review discusses standard in vitro, in vivo, and in silico methods for studying AQPs, especially for biomedical and mammalian cell biology applications. We also propose some new techniques and approaches for future AQP research to address current gaps in methodology.
Collapse
Affiliation(s)
- Shohini Banerjee
- Fischell Department of Bioengineering, University of Maryland, MD 20742, United States
| | - Ian M Smith
- Fischell Department of Bioengineering, University of Maryland, MD 20742, United States
| | - Autumn C Hengen
- Fischell Department of Bioengineering, University of Maryland, MD 20742, United States
| | - Kimberly M Stroka
- Fischell Department of Bioengineering, University of Maryland, MD 20742, United States
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore MD 21201, United States
- Biophysics Program, University of Maryland, MD 20742, United States
- Center for Stem Cell Biology and Regenerative Medicine, University of Maryland, Baltimore MD 21201, United States
| |
Collapse
|
5
|
Moreno-Alcántar G, Picchetti P, Casini A. Gold Complexes in Anticancer Therapy: From New Design Principles to Particle-Based Delivery Systems. Angew Chem Int Ed Engl 2023; 62:e202218000. [PMID: 36847211 DOI: 10.1002/anie.202218000] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/26/2023] [Accepted: 02/27/2023] [Indexed: 02/28/2023]
Abstract
The discovery of the medicinal properties of gold complexes has fuelled the design and synthesis of new anticancer metallodrugs, which have received special attention due to their unique modes of action. Current research in the development of gold compounds with therapeutic properties is predominantly focused on the molecular design of drug leads with superior pharmacological activities, e.g., by introducing targeting features. Moreover, intensive research aims at improving the physicochemical properties of gold compounds, such as chemical stability and solubility in the physiological environment. In this regard, the encapsulation of gold compounds in nanocarriers or their chemical grafting onto targeted delivery vectors could lead to new nanomedicines that eventually reach clinical applications. Herein, we provide an overview of the state-of-the-art progress of gold anticancer compounds, andmore importantly we thoroughly revise the development of nanoparticle-based delivery systems for gold chemotherapeutics.
Collapse
Affiliation(s)
- Guillermo Moreno-Alcántar
- Chair of Medicinal and Bioinorganic Chemistry, School of Natural Sciences, Department of Chemistry, Technical University of Munich (TUM), Lichtenbergstr. 4, 85748, Garching b. München, Germany
| | - Pierre Picchetti
- Karlsruhe Institute of Technology (KIT), Institute of Nanotechnology, Hermann-von-Helmholtz-Platz 1, 76344, Eggenstein-Leopoldshafen, Germany
| | - Angela Casini
- Chair of Medicinal and Bioinorganic Chemistry, School of Natural Sciences, Department of Chemistry, Technical University of Munich (TUM), Lichtenbergstr. 4, 85748, Garching b. München, Germany
| |
Collapse
|
6
|
Abstract
Aquaporins (AQP) working as membrane channels facilitated water transport, play vital roles in various physiological progress including cell migration, energy metabolism, inflammation, etc. They are quite important drug targets, but elusive for discovery due to their undruggable properties. In this chapter, we summarized most fluently used methods for screening AQP inhibitors, including cell swelling assay, cell shrinking assay, and stopped-flow assay. And three classes of AQP inhibitors have been discussed, including metal-related inhibitors, quaternary ammonium salts, and small molecule inhibitors which further divided into four parts, sulfanilamide analogies, TGN-020, antiepileptic drugs, and others. It has been suggested that although they showed inhibition effects on AQP1, AQP3, AQP4, AQP7, or AQP9 in some researches, none of them could be asserted as AQP inhibitors to some extent. Discovering AQP inhibitors is a big challenge, but if successful, it will be a great contribution for human health.
Collapse
Affiliation(s)
- Shuyuan Wang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | | | - Baoxue Yang
- School of Basic Medical Sciences, Peking University, Beijing, China.
| |
Collapse
|
7
|
The role of tridentate ligands on the redox stability of anticancer gold(III) complexes. J Inorg Biochem 2022; 236:111970. [PMID: 36049259 DOI: 10.1016/j.jinorgbio.2022.111970] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 08/12/2022] [Accepted: 08/13/2022] [Indexed: 12/15/2022]
Abstract
Gold(III) complexes are promising compounds for cancer chemotherapy, whose action depends on their redox stability. In this context, the choice of ligands is crucial to adjust their reactivity and biological response. The present study addressed the effect of the gold coordination sphere on the reduction potential (Eo) for ten gold(III) complexes containing five or six-membered rings tridentate ligands - [AuIII(trident)Cl]3+n (trident = N^N^N, C^N^N, C^C^N, C^N^C, and N^C^N). The calculated Eo covered a broad range of 2500 mV with the most stable complexes containing two AuC bonds (Eo = -1.85 V for [AuIII(C^C^N)Cl] - f). For complexes with one AuC bond, the N^C^N ligands stabilize the gold(III) complex more efficiently than N^N^C; however, the inclusion of the non-innocent ligand bipy (2,2'-bipyridine) in N^N portion provides an extra stabilization effect. Among the derivatives with one AuC bond, [AuIII(N^N^C)Cl]+ (N^N = bipy) (a) showed Eo = -1.20 V. For the complexes with N^N^N ligands, Eo was positive and almost constant (+0.60 V). Furthermore, the kinetics for ligand exchange reactions (Cl-/H2O, H2O/Cys and Cl-/Cys) were monitored for the most stable compounds and the energy profiles compared to the reduction pathways.
Collapse
|
8
|
Charlestin V, Fulkerson D, Arias Matus CE, Walker ZT, Carthy K, Littlepage LE. Aquaporins: New players in breast cancer progression and treatment response. Front Oncol 2022; 12:988119. [PMID: 36212456 PMCID: PMC9532844 DOI: 10.3389/fonc.2022.988119] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 08/09/2022] [Indexed: 11/30/2022] Open
Abstract
Aquaporins (AQPs) are a family of small transmembrane proteins that selectively transport water and other small molecules and ions following an osmotic gradient across cell plasma membranes. This enables them to regulate numerous functions including water homeostasis, fat metabolism, proliferation, migration, and adhesion. Previous structural and functional studies highlight a strong biological relationship between AQP protein expression, localization, and key biological functions in normal and cancer tissues, where aberrant AQP expression correlates with tumorigenesis and metastasis. In this review, we discuss the roles of AQP1, AQP3, AQP4, AQP5, and AQP7 in breast cancer progression and metastasis, including the role of AQPs in the tumor microenvironment, to highlight potential contributions of stromal-derived to epithelial-derived AQPs to breast cancer. Emerging evidence identifies AQPs as predictors of response to cancer therapy and as targets for increasing their sensitivity to treatment. However, these studies have not evaluated the requirements for protein structure on AQP function within the context of breast cancer. We also examine how AQPs contribute to a patient's response to cancer treatment, existing AQP inhibitors and how AQPs could serve as novel predictive biomarkers of therapy response in breast cancer. Future studies also should evaluate AQP redundancy and compensation as mechanisms used to overcome aberrant AQP function. This review highlights the need for additional research into how AQPs contribute molecularly to therapeutic resistance and by altering the tumor microenvironment.
Collapse
Affiliation(s)
- Verodia Charlestin
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, United States
- Harper Cancer Research Institute, University of Notre Dame, South Bend, IN, United States
| | - Daniel Fulkerson
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, United States
- Harper Cancer Research Institute, University of Notre Dame, South Bend, IN, United States
| | - Carlos E. Arias Matus
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, United States
- Harper Cancer Research Institute, University of Notre Dame, South Bend, IN, United States
- Department of Biotechnology, Universidad Popular Autónoma del Estado de Puebla, Pue, Mexico
| | - Zachary T. Walker
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, United States
- Harper Cancer Research Institute, University of Notre Dame, South Bend, IN, United States
| | - Kevin Carthy
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, United States
- Harper Cancer Research Institute, University of Notre Dame, South Bend, IN, United States
| | - Laurie E. Littlepage
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, United States
- Harper Cancer Research Institute, University of Notre Dame, South Bend, IN, United States
| |
Collapse
|
9
|
Pimpão C, Wragg D, da Silva IV, Casini A, Soveral G. Aquaglyceroporin Modulators as Emergent Pharmacological Molecules for Human Diseases. Front Mol Biosci 2022; 9:845237. [PMID: 35187089 PMCID: PMC8850838 DOI: 10.3389/fmolb.2022.845237] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 01/13/2022] [Indexed: 12/26/2022] Open
Abstract
Aquaglyceroporins, a sub-class of aquaporins that facilitate the diffusion of water, glycerol and other small uncharged solutes across cell membranes, have been recognized for their important role in human physiology and their involvement in multiple disorders, mostly related to disturbed energy homeostasis. Aquaglyceroporins dysfunction in a variety of pathological conditions highlighted their targeting as novel therapeutic strategies, boosting the search for potent and selective modulators with pharmacological properties. The identification of selective inhibitors with potential clinical applications has been challenging, relying on accurate assays to measure membrane glycerol permeability and validate effective functional blockers. Additionally, biologicals such as hormones and natural compounds have been revealed as alternative strategies to modulate aquaglyceroporins via their gene and protein expression. This review summarizes the current knowledge of aquaglyceroporins’ involvement in several pathologies and the experimental approaches used to evaluate glycerol permeability and aquaglyceroporin modulation. In addition, we provide an update on aquaglyceroporins modulators reported to impact disease, unveiling aquaglyceroporin pharmacological targeting as a promising approach for innovative therapeutics.
Collapse
Affiliation(s)
- Catarina Pimpão
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal
- Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal
| | - Darren Wragg
- Department of Chemistry, Technical University of Munich, Munich, Germany
| | - Inês V. da Silva
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal
- Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal
| | - Angela Casini
- Department of Chemistry, Technical University of Munich, Munich, Germany
- *Correspondence: Angela Casini, ; Graça Soveral,
| | - Graça Soveral
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal
- Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal
- *Correspondence: Angela Casini, ; Graça Soveral,
| |
Collapse
|
10
|
Henderson SW, Nourmohammadi S, Ramesh SA, Yool AJ. Aquaporin ion conductance properties defined by membrane environment, protein structure, and cell physiology. Biophys Rev 2022; 14:181-198. [PMID: 35340612 PMCID: PMC8921385 DOI: 10.1007/s12551-021-00925-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 12/09/2021] [Indexed: 01/13/2023] Open
Abstract
Aquaporins (AQPs) are multifunctional transmembrane channel proteins permeable to water and an expanding array of solutes. AQP-mediated ion channel activity was first observed when purified AQP0 from bovine lens was incorporated into lipid bilayers. Electrophysiological properties of ion-conducting AQPs since discovered in plants, invertebrates, and mammals have been assessed using native, reconstituted, and heterologously expressed channels. Accumulating evidence is defining amino acid residues that govern differential solute permeability through intrasubunit and central pores of AQP tetramers. Rings of charged and hydrophobic residues around pores influence AQP selectivity, and are candidates for further work to define motifs that distinguish ion conduction capability, versus strict water and glycerol permeability. Similarities between AQP ion channels thus far include large single channel conductances and long open times, but differences in ionic selectivity, permeability to divalent cations, and mechanisms of gating (e.g., by voltage, pH, and cyclic nucleotides) are unique to subtypes. Effects of lipid environments in modulating parameters such as single channel amplitude could explain in part the variations in AQP ion channel properties observed across preparations. Physiological roles of the ion-conducting AQP classes span diverse processes including regulation of cell motility, organellar pH, neural development, signaling, and nutrient acquisition. Advances in computational methods can generate testable predictions of AQP structure-function relationships, which combined with innovative high-throughput assays could revolutionize the field in defining essential properties of ion-conducting AQPs, discovering new AQP ion channels, and understanding the effects of AQP interactions with proteins, signaling cascades, and membrane lipids.
Collapse
Affiliation(s)
- Sam W. Henderson
- School of Biomedicine, University of Adelaide, Adelaide, SA 5005 Australia
| | | | - Sunita A. Ramesh
- College of Science and Engineering, Flinders University, Bedford Park, SA 5042 Australia
| | - Andrea J. Yool
- School of Biomedicine, University of Adelaide, Adelaide, SA 5005 Australia
| |
Collapse
|
11
|
Pinto A, Roma-Rodrigues C, Ward JS, Puttreddy R, Rissanen K, Baptista PV, Fernandes AR, Lima JC, Rodríguez L. Aggregation versus Biological Activity in Gold(I) Complexes. An Unexplored Concept. Inorg Chem 2021; 60:18753-18763. [PMID: 34719915 DOI: 10.1021/acs.inorgchem.1c02359] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The aggregation process of a series of mono- and dinuclear gold(I) complexes containing a 4-ethynylaniline ligand and a phosphane at the second coordination position (PR3-Au-C≡CC6H4-NH2, complexes 1-5, and (diphos)(Au-C≡CC6H4-NH2)2, complexes 6-8), whose biological activity was previously studied by us, has been carefully analyzed through absorption, emission, and NMR spectroscopy, together with dynamic light scattering and small-angle X-ray scattering. These experiments allow us to retrieve information about how the compounds enter the cells. It was observed that all compounds present aggregation in fresh solutions, before biological treatment, and thus they must be entering the cells as aggregates. Inductively coupled plasma atomic emission spectrometry measurements showed that mononuclear complexes are mainly found in the cytosolic fraction; the dinuclear complexes are mainly found in a subsequent fraction composed of nuclei and cytoskeleton. Additionally, dinuclear complex 8 affects the actin aggregation to a larger extent, suggesting a cooperative effect of dinuclear compounds.
Collapse
Affiliation(s)
- Andrea Pinto
- Departament de Química Inorgànica i Orgànica, Secció de Química Inorgànica, Universitat de Barcelona, Martí i Franquès 1-11, E-08028 Barcelona, Spain
- Institut de Nanociència i Nanotecnologia (IN2UB). Universitat de Barcelona, 08028 Barcelona, Spain
| | - Catarina Roma-Rodrigues
- UCIBIO─Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal
| | - Jas S Ward
- Department of Chemistry, University of Jyvaskyla, P.O. Box 35, 40014 Jväskylä, Finland
| | - Rakesh Puttreddy
- Faculty of Engineering and Natural Sciences, Tampere University, P.O. Box 541, FI-33101 Tampere, Finland
| | - Kari Rissanen
- Department of Chemistry, University of Jyvaskyla, P.O. Box 35, 40014 Jväskylä, Finland
| | - Pedro V Baptista
- UCIBIO─Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal
| | - Alexandra R Fernandes
- UCIBIO─Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal
| | - João Carlos Lima
- LAQV-REQUIMTE, Departamento de Química, CQFB, Universidade Nova de Lisboa, 2825-152 Monte de Caparica, Portugal
| | - Laura Rodríguez
- Departament de Química Inorgànica i Orgànica, Secció de Química Inorgànica, Universitat de Barcelona, Martí i Franquès 1-11, E-08028 Barcelona, Spain
- Institut de Nanociència i Nanotecnologia (IN2UB). Universitat de Barcelona, 08028 Barcelona, Spain
| |
Collapse
|
12
|
Tolbatov I, Marrone A, Coletti C, Re N. Computational Studies of Au(I) and Au(III) Anticancer MetalLodrugs: A Survey. Molecules 2021; 26:7600. [PMID: 34946684 PMCID: PMC8707411 DOI: 10.3390/molecules26247600] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/29/2021] [Accepted: 12/12/2021] [Indexed: 11/16/2022] Open
Abstract
Owing to the growing hardware capabilities and the enhancing efficacy of computational methodologies, computational chemistry approaches have constantly become more important in the development of novel anticancer metallodrugs. Besides traditional Pt-based drugs, inorganic and organometallic complexes of other transition metals are showing increasing potential in the treatment of cancer. Among them, Au(I)- and Au(III)-based compounds are promising candidates due to the strong affinity of Au(I) cations to cysteine and selenocysteine side chains of the protein residues and to Au(III) complexes being more labile and prone to the reduction to either Au(I) or Au(0) in the physiological milieu. A correct prediction of metal complexes' properties and of their bonding interactions with potential ligands requires QM computations, usually at the ab initio or DFT level. However, MM, MD, and docking approaches can also give useful information on their binding site on large biomolecular targets, such as proteins or DNA, provided a careful parametrization of the metal force field is employed. In this review, we provide an overview of the recent computational studies of Au(I) and Au(III) antitumor compounds and of their interactions with biomolecular targets, such as sulfur- and selenium-containing enzymes, like glutathione reductases, glutathione peroxidase, glutathione-S-transferase, cysteine protease, thioredoxin reductase and poly (ADP-ribose) polymerase 1.
Collapse
Affiliation(s)
- Iogann Tolbatov
- Institut de Chimie Moleculaire de l’Université de Bourgogne (ICMUB), Université de Bourgogne Franche-Comté (UBFC), Avenue Alain Savary 9, 21078 Dijon, France;
| | - Alessandro Marrone
- Dipartimento di Farmacia, Università degli Studi “G. D’Annunzio” Chieti-Pescara, Via dei Vestini, 66100 Chieti, Italy; (A.M.); (C.C.)
| | - Cecilia Coletti
- Dipartimento di Farmacia, Università degli Studi “G. D’Annunzio” Chieti-Pescara, Via dei Vestini, 66100 Chieti, Italy; (A.M.); (C.C.)
| | - Nazzareno Re
- Dipartimento di Farmacia, Università degli Studi “G. D’Annunzio” Chieti-Pescara, Via dei Vestini, 66100 Chieti, Italy; (A.M.); (C.C.)
| |
Collapse
|
13
|
Zhang H, Cai W, Shao X. Regulation of aquaporin-3 water permeability by hyaluronan. Phys Chem Chem Phys 2021; 23:25706-25711. [PMID: 34755729 DOI: 10.1039/d1cp02867g] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Hyaluronan (HA) is a major component in the extracellular matrix and is responsible for maintaining the water content of the skin. However, the function and moisturizing mechanism at the atomic level of HA remain only partially understood. Investigating the interactions of HA and other skin components can help us understand how the former moisturizes the skin. Considering that aquaporin-3 (AQP3) is a protein responsible for transmembrane water transport in the human skin, we have, therefore, investigated the interactions of AQP3 and HA with different molecular weights using molecular dynamics simulations in the present work. Our results indicate that HA can adsorb onto AQP3 and decrease water mobility around the latter. In addition, the permeation rate of water through AQP3 can also be decreased by HA, and this phenomenon is particularly obvious for small molecular HA. Moreover, we found that large molecular HA can link two adjacent membranes in the extracellular matrix, increasing the adhesion between the membranes in the periplasm. The results of the present study indicate that HA is a natural regulator of AQP3, revealing the synergetic function of HA and AQP3 in the extracellular matrix of the skin.
Collapse
Affiliation(s)
- Hong Zhang
- Research Center for Analytical Sciences, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin Key Laboratory of Biosensing and Molecular Recognition, State Key Laboratory of Medicinal Chemical Biology, Tianjin 300071, China.
| | - Wensheng Cai
- Research Center for Analytical Sciences, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin Key Laboratory of Biosensing and Molecular Recognition, State Key Laboratory of Medicinal Chemical Biology, Tianjin 300071, China.
| | - Xueguang Shao
- Research Center for Analytical Sciences, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin Key Laboratory of Biosensing and Molecular Recognition, State Key Laboratory of Medicinal Chemical Biology, Tianjin 300071, China.
| |
Collapse
|
14
|
Chakraborty P, Oosterhuis D, Bonsignore R, Casini A, Olinga P, Scheffers D. An Organogold Compound as Potential Antimicrobial Agent against Drug-Resistant Bacteria: Initial Mechanistic Insights. ChemMedChem 2021; 16:3060-3070. [PMID: 34181818 PMCID: PMC8518660 DOI: 10.1002/cmdc.202100342] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Indexed: 01/07/2023]
Abstract
The rise of antimicrobial resistance has necessitated novel strategies to efficiently combat pathogenic bacteria. Metal-based compounds have been proven as a possible alternative to classical organic drugs. Here, we have assessed the antibacterial activity of seven gold complexes of different families. One compound, a cyclometalated Au(III) C^N complex, showed activity against Gram-positive bacteria, including multi-drug resistant clinical strains. The mechanism of action of this compound was studied in Bacillus subtilis. Overall, the studies point towards a complex mode of antibacterial action, which does not include induction of oxidative stress or cell membrane damage. A number of genes related to metal transport and homeostasis were upregulated upon short treatment of the cells with gold compound. Toxicity tests conducted on precision-cut mouse tissue slices ex vivo revealed that the organogold compound is poorly toxic to mouse liver and kidney tissues, and may thus, be treated as an antibacterial drug candidate.
Collapse
Affiliation(s)
- Parichita Chakraborty
- Department of Molecular MicrobiologyGroningen Institute for Biomolecular Sciences and BiotechnologyUniversity of Groningen9747 AGGroningenThe Netherlands
| | - Dorenda Oosterhuis
- Department of Pharmaceutical Technology and BiopharmacyGroningen Research Institute of PharmacyUniversity of Groningen9713AVGroningenThe Netherlands
| | - Riccardo Bonsignore
- Chair of Medicinal and Bioinorganic ChemistryDepartment of ChemistryTechnical University of MunichLichtenbergstr. 485748Garching b. MünchenGermany
| | - Angela Casini
- Chair of Medicinal and Bioinorganic ChemistryDepartment of ChemistryTechnical University of MunichLichtenbergstr. 485748Garching b. MünchenGermany
| | - Peter Olinga
- Department of Pharmaceutical Technology and BiopharmacyGroningen Research Institute of PharmacyUniversity of Groningen9713AVGroningenThe Netherlands
| | - Dirk‐Jan Scheffers
- Department of Molecular MicrobiologyGroningen Institute for Biomolecular Sciences and BiotechnologyUniversity of Groningen9747 AGGroningenThe Netherlands
| |
Collapse
|
15
|
Pimpão C, Wragg D, Bonsignore R, Aikman B, Pedersen PA, Leoni S, Soveral G, Casini A. Mechanisms of irreversible aquaporin-10 inhibition by organogold compounds studied by combined biophysical methods and atomistic simulations. Metallomics 2021; 13:6360981. [PMID: 34468767 DOI: 10.1093/mtomcs/mfab053] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 08/18/2021] [Indexed: 11/14/2022]
Abstract
The inhibition of glycerol permeation via human aquaporin-10 (hAQP10) by organometallic gold complexes has been studied by stopped-flow fluorescence spectroscopy, and its mechanism has been described using molecular modelling and atomistic simulations. The most effective hAQP10 inhibitors are cyclometalated Au(III) C^N compounds known to efficiently react with cysteine residues leading to the formation of irreversible C-S bonds. Functional assays also demonstrate the irreversibility of the binding to hAQP10 by the organometallic complexes. The obtained computational results by metadynamics show that the local arylation of Cys209 in hAQP10 by one of the gold inhibitors is mapped into a global change of the overall free energy of glycerol translocation across the channel. Our study further pinpoints the need to understand the mechanism of glycerol and small molecule permeation as a combination of local structural motifs and global pore conformational changes, which are taking place on the scale of the translocation process and whose study, therefore, require sophisticated molecular dynamics strategies.
Collapse
Affiliation(s)
- Catarina Pimpão
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisboa, Portugal
| | - Darren Wragg
- Department of Chemistry, Technical University of Munich, 85747 Garching bei München, Germany
| | - Riccardo Bonsignore
- Department of Chemistry, Technical University of Munich, 85747 Garching bei München, Germany
| | - Brech Aikman
- Department of Chemistry, Technical University of Munich, 85747 Garching bei München, Germany
- School of Chemistry, Cardiff University, Main Building, Park Place, Cardiff CF10 3AT, UK
| | - Per Amstrup Pedersen
- Department of Biology, University of Copenhagen, Universitetsparken 13, DK-2100 Copenhagen OE, Denmark
| | - Stefano Leoni
- School of Chemistry, Cardiff University, Main Building, Park Place, Cardiff CF10 3AT, UK
| | - Graça Soveral
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisboa, Portugal
| | - Angela Casini
- Department of Chemistry, Technical University of Munich, 85747 Garching bei München, Germany
| |
Collapse
|
16
|
Zhou XQ, Carbo-Bague I, Siegler MA, Hilgendorf J, Basu U, Ott I, Liu R, Zhang L, Ramu V, IJzerman AP, Bonnet S. Rollover Cyclometalation vs Nitrogen Coordination in Tetrapyridyl Anticancer Gold(III) Complexes: Effect on Protein Interaction and Toxicity. JACS AU 2021; 1:380-395. [PMID: 34056633 PMCID: PMC8154207 DOI: 10.1021/jacsau.0c00104] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Indexed: 05/05/2023]
Abstract
In this work, a pair of gold(III) complexes derived from the analogous tetrapyridyl ligands H2biqbpy1 and H2biqbpy2 was prepared: the rollover, bis-cyclometalated [Au(biqbpy1)Cl ([1]Cl) and its isomer [Au(biqbpy2)Cl ([2]Cl). In [1]+, two pyridyl rings coordinate to the metal via a Au-C bond (C∧N∧N∧C coordination) and the two noncoordinated amine bridges of the ligand remain protonated, while in [2]+ all four pyridyl rings of the ligand coordinate to the metal via a Au-N bond (N∧N∧N∧N coordination), but both amine bridges are deprotonated. As a result, both complexes are monocationic, which allowed comparison of the sole effect of cyclometalation on the chemistry, protein interaction, and anticancer properties of the gold(III) compounds. Due to their identical monocationic charge and similar molecular shape, both complexes [1]Cl and [2]Cl displaced reference radioligand [3H]dofetilide equally well from cell membranes expressing the Kv11.1 (hERG) potassium channel, and more so than the tetrapyridyl ligands H2biqbpy1 and H2biqbpy2. By contrast, cyclometalation rendered [1]Cl coordinatively stable in the presence of biological thiols, while [2]Cl was reduced by a millimolar concentration of glutathione into metastable Au(I) species releasing the free ligand H2biqbpy2 and TrxR-inhibiting Au+ ions. The redox stability of [1]Cl dramatically decreased its thioredoxin reductase (TrxR) inhibition properties, compared to [2]Cl. On the other hand, unlike [2]Cl, [1]Cl aggregated into nanoparticles in FCS-containing medium, which resulted in much more efficient gold cellular uptake. [1]Cl had much more selective anticancer properties than [2]Cl and cisplatin, as it was almost 10 times more cytotoxic to human cancer cells (A549, A431, A375, and MCF7) than to noncancerous cells (MRC5). Mechanistic studies highlight the strikingly different mode of action of the two compounds: while for [1]Cl high gold cellular uptake, nuclear DNA damage, and interaction with hERG may contribute to cell killing, for [2]Cl extracellular reduction released TrxR-inhibiting Au+ ions that were taken up in minute amounts in the cytosol, and a toxic tetrapyridyl ligand also capable of binding to hERG. These results demonstrate that bis-cyclometalation is an appealing method to improve the redox stability of Au(III) compounds and to develop gold-based cytotoxic compounds that do not rely on TrxR inhibition to kill cancer cells.
Collapse
Affiliation(s)
- Xue-Quan Zhou
- Leiden
Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Imma Carbo-Bague
- Leiden
Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
- Department
of Chemistry, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia V5A 1S6, Canada
| | - Maxime A. Siegler
- Department
of Chemistry, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Jonathan Hilgendorf
- Leiden
Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Uttara Basu
- Institute
of Medicinal and Pharmaceutical Chemistry, Technische Universität Braunschweig, Beethovenstrasse 55, 38106 Braunschweig, Germany
| | - Ingo Ott
- Institute
of Medicinal and Pharmaceutical Chemistry, Technische Universität Braunschweig, Beethovenstrasse 55, 38106 Braunschweig, Germany
| | - Rongfang Liu
- Division
of Drug Discovery & Safety, Leiden Academic Centre for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands
| | - Liyan Zhang
- Leiden
Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Vadde Ramu
- Leiden
Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Adriaan P. IJzerman
- Division
of Drug Discovery & Safety, Leiden Academic Centre for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands
| | - Sylvestre Bonnet
- Leiden
Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| |
Collapse
|
17
|
Lv H, Li Y, Xue C, Dong N, Bi C, Shan A. Aquaporin: targets for dietary nutrients to regulate intestinal health. J Anim Physiol Anim Nutr (Berl) 2021; 106:167-180. [PMID: 33811387 DOI: 10.1111/jpn.13539] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 12/07/2020] [Accepted: 02/07/2021] [Indexed: 12/22/2022]
Abstract
Aquaporins (AQP) are a class of water channel membrane proteins that are widely expressed in the gut. The biological functions of aquaporins, which regulate the absorption and secretion of water molecules and small solutes, maintain the stable state of the intestine, regulate cell proliferation and migration, participate in the process of intestinal inflammation, and mediate tumorigenesis, demonstrate the physiological significance of these channels in intestinal health. The pathology of many intestinal diseases is associated with changes in the location and expression of aquaporins, such as intestinal infection, which can change the expression and distribution of AQPs in intestinal tissues/cells by affecting cytokines and chemokines. This can lead to various intestinal diseases such as diarrhoea, which also suggests the importance of aquaporins in the prevention and treatment of intestinal diseases. This review summarizes the relationship between aquaporins and intestinal physiology and diseases and focuses on drugs (such as plant extracts) or diets that can regulate intestinal health by regulating aquaporins. It provides a basis for establishing aquaporins as biomarkers and therapeutic targets for intestinal health.
Collapse
Affiliation(s)
- Hao Lv
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Ying Li
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Chenyu Xue
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Na Dong
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Chongpeng Bi
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Anshan Shan
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| |
Collapse
|
18
|
Calamita G, Delporte C. Involvement of aquaglyceroporins in energy metabolism in health and disease. Biochimie 2021; 188:20-34. [PMID: 33689852 DOI: 10.1016/j.biochi.2021.03.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/23/2021] [Accepted: 03/01/2021] [Indexed: 11/27/2022]
Abstract
Aquaglyceroporins are a group of the aquaporin (AQP) family of transmembrane water channels. While AQPs facilitate the passage of water, small solutes, and gases across biological membranes, aquaglyceroporins allow passage of water, glycerol, urea and some other solutes. Thanks to their glycerol permeability, aquaglyceroporins are involved in energy homeostasis. This review provides an overview of what is currently known concerning the functional implication and control of aquaglyceroporins in tissues involved in energy metabolism, i.e. liver, adipose tissue and endocrine pancreas. The expression, role and (dys)regulation of aquaglyceroporins in disorders affecting energy metabolism, and the potential relevance of aquaglyceroporins as drug targets to treat the alterations of the energy balance is also addressed.
Collapse
Affiliation(s)
- Giuseppe Calamita
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari "Aldo Moro", Bari, Italy
| | - Christine Delporte
- Laboratory of Pathophysiological and Nutritional Biochemistry, Université Libre de Bruxelles, Brussels, Belgium.
| |
Collapse
|
19
|
Sánchez Delgado GY, Arvellos JFA, Paschoal DFS, Dos Santos HF. Role of the Enzymatic Environment in the Reactivity of the Au III-C^N^C Anticancer Complexes. Inorg Chem 2021; 60:3181-3195. [PMID: 33600154 DOI: 10.1021/acs.inorgchem.0c03521] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The action mechanism of anticancer gold(III) complexes is a multi-step process and depends on their redox stability. First, the gold(III) complex undergoes a ligand exchange reaction in the presence of cellular thiols, such as those available in the active site of the enzyme TrxR, and then, the AuIII → AuI reduction occurs. Most experimental and theoretical studies describe these processes under chemical conditions without considering the enzyme structure effect. In the present study, molecular models are proposed for the [AuIII(C^N^C)(SHCys-R)]+ adduct, with the [AuIII(C^N^C)]+ moiety bonded to the Cys498 residue in the C-terminal arm of the TrxR. This one represents the product of the first ligand exchange reaction. Overall, our results suggest that the exchange of the auxiliary ligand (for instance, Cl- to S-R) plays a primary role in increasing the reduction potential, with the enzyme structure having a small effect. The parent compound [AuIII(C^N^C)Cl] has E° = -1.20 V, which enlarges to -0.72 V for [AuIII(C^N^C)CH3SH]+ and to -0.65 V for the largest model studied, Au-trx. In addition to the effect of the enzyme structure on the redox stability, we also analyze the Au transfer to the enzyme using a small peptide model (a tetramer). This reaction is dependent on the Cys497 protonation state. Thermodynamics and kinetic analysis suggests that the C^N^C ligand substitution by Cys497 is an exergonic process, with an energy barrier estimated at 20.2 kcal mol-1. The complete transfer of the Au ion to the enzyme's active site would lead to a total loss of enzyme activity, generating oxidative damage and, consequently, cancer cell death.
Collapse
Affiliation(s)
- Giset Y Sánchez Delgado
- NEQC: Núcleo de Estudos em Química Computacional, Department of Chemistry, Federal University of Juiz de Fora, Campus Universitário Martelos, 36.036-900 Juiz de Fora, Minas Gerais, Brazil
| | - Júlio F A Arvellos
- NEQC: Núcleo de Estudos em Química Computacional, Department of Chemistry, Federal University of Juiz de Fora, Campus Universitário Martelos, 36.036-900 Juiz de Fora, Minas Gerais, Brazil
| | - Diego F S Paschoal
- NQTCM: Núcleo de Química Teórica e Computacional de Macaé, Polo Ajuda, Universidade Federal do Rio de Janeiro, Campus UFRJ-Macaé, 27.971-525 Macaé, Rio de Janeiro, Brazil
| | - Hélio F Dos Santos
- NEQC: Núcleo de Estudos em Química Computacional, Department of Chemistry, Federal University of Juiz de Fora, Campus Universitário Martelos, 36.036-900 Juiz de Fora, Minas Gerais, Brazil
| |
Collapse
|
20
|
da Silva IV, Cardoso C, Martínez-Banaclocha H, Casini A, Pelegrín P, Soveral G. Aquaporin-3 is involved in NLRP3-inflammasome activation contributing to the setting of inflammatory response. Cell Mol Life Sci 2021; 78:3073-3085. [PMID: 33231721 PMCID: PMC11073090 DOI: 10.1007/s00018-020-03708-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 10/23/2020] [Accepted: 11/11/2020] [Indexed: 12/11/2022]
Abstract
Inflammasomes are large immune multiprotein complexes that tightly regulate the production of the pro-inflammatory cytokines, being dependent on cell regulatory volume mechanisms. Aquaporins (AQPs) are protein channels that facilitate the transport of water and glycerol (aquaglyceroporins) through membranes, essential for cell volume regulation. Although these membrane proteins are highly expressed in monocytes and macrophages, their role in the inflammatory process is still unclear. Here, we investigated the role of aquaglyceroporin AQP3 in NLRP3-inflammasome activation by complementary approaches based either on shRNA silencing or on AQP3 selective inhibition. The latter has been achieved using a reported potent gold-based inhibitor, Auphen. AQP3 inhibition or silencing partially blocked LPS-priming and decreased production of IL-6, proIL-1β, and TNF-α, suggesting the possible involvement of AQP3 in macrophage priming by Toll-like receptor 4 engagement. Moreover, AQP3-dependent cell reswelling increased IL-1β release through caspase-1 activation. NLRP3-inflammasome activation induced by reswelling, nigericin, and ATP was also blocked when AQP3 was inhibited or silenced. Altogether, these data point towards AQPs as potential players in the setting of the inflammatory response.
Collapse
Affiliation(s)
- Inês Vieira da Silva
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisboa, Portugal
- Department of Biochemistry and Human Biology, Faculty of Pharmacy, Universidade de Lisboa, 1649-003, Lisboa, Portugal
| | - Carlos Cardoso
- Department of Biochemistry and Human Biology, Faculty of Pharmacy, Universidade de Lisboa, 1649-003, Lisboa, Portugal
- Clinical Chemistry Laboratory, Dr. Joaquim Chaves, 1495-148, Algés, Portugal
| | - Helios Martínez-Banaclocha
- Molecular Inflammation Group, Biomedical Research Institute of Murcia (IMIB-Arrixaca), Hospital Clínico Universitario Virgen de La Arrixaca, Carretera Buenavista, 30120, Murcia, Spain
| | - Angela Casini
- Department of Chemistry, Technical University of Munich, Lichtenbergstr. 4, 85748, Garching b. München, Germany
| | - Pablo Pelegrín
- Molecular Inflammation Group, Biomedical Research Institute of Murcia (IMIB-Arrixaca), Hospital Clínico Universitario Virgen de La Arrixaca, Carretera Buenavista, 30120, Murcia, Spain.
| | - Graça Soveral
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisboa, Portugal.
- Department of Biochemistry and Human Biology, Faculty of Pharmacy, Universidade de Lisboa, 1649-003, Lisboa, Portugal.
| |
Collapse
|
21
|
Palermo G, Spinello A, Saha A, Magistrato A. Frontiers of metal-coordinating drug design. Expert Opin Drug Discov 2020; 16:497-511. [PMID: 33874825 DOI: 10.1080/17460441.2021.1851188] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Introduction: The occurrence of metal ions in biomolecules is required to exert vital cellular functions. Metal-containing biomolecules can be modulated by small-molecule inhibitors targeting their metal-moiety. As well, the discovery of cisplatin ushered the rational discovery of metal-containing-drugs. The use of both drug types exploiting metal-ligand interactions is well established to treat distinct pathologies. Therefore, characterizing and leveraging metal-coordinating drugs is a pivotal, yet challenging, part of medicinal chemistry.Area covered: Atomic-level simulations are increasingly employed to overcome the challenges met by traditional drug-discovery approaches and to complement wet-lab experiments in elucidating the mechanisms of drugs' action. Multiscale simulations, allow deciphering the mechanism of metal-binding inhibitors and metallo-containing-drugs, enabling a reliable description of metal-complexes in their biological environment. In this compendium, the authors review selected applications exploiting the metal-ligand interactions by focusing on understanding the mechanism and design of (i) inhibitors targeting iron and zinc-enzymes, and (ii) ruthenium and gold-based anticancer agents targeting the nucleosome and aquaporin protein, respectively.Expert opinion: The showcased applications exemplify the current role and the potential of atomic-level simulations and reveal how their synergic use with experiments can contribute to uncover fundamental mechanistic facets and exploit metal-ligand interactions in medicinal chemistry.
Collapse
Affiliation(s)
- Giulia Palermo
- Department of Bioengineering and Department of Chemistry, University of California Riverside, Riverside, United States
| | - Angelo Spinello
- National Research Council (CNR) of Italy, Institute of Material (IOM) @ International School for Advanced Studies (SISSA), Trieste, Italy
| | - Aakash Saha
- Department of Bioengineering, University of California Riverside, Riverside, United States
| | - Alessandra Magistrato
- National Research Council (CNR) of Italy, Institute of Material (IOM) @ International School for Advanced Studies (SISSA), Trieste, Italy
| |
Collapse
|
22
|
Wragg D, Leoni S, Casini A. Aquaporin-driven hydrogen peroxide transport: a case of molecular mimicry? RSC Chem Biol 2020; 1:390-394. [PMID: 34458769 PMCID: PMC8341912 DOI: 10.1039/d0cb00160k] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 11/18/2020] [Indexed: 01/13/2023] Open
Abstract
Aquaporins (AQPs) are membrane proteins that have evolved to control cellular water uptake and efflux, and as such are amongst the most ancient biological "devices" in cellular organisms. Recently, using metadynamics, we have shown that water nanoconfinement within aquaporin channels results into bidirectional water movement along single file chains, extending previous investigations. Here, the elusive mechanisms of H2O2 facilitated transport by the human 'peroxiporin' AQP3 has been unravelled via a combination of atomistic simulations, showing that while hydrogen peroxide is able to mimic water during AQP3 permeation, this comes at a certain energy expense due to the required conformational changes within the channel. Furthermore, the intrinsic water dynamics allows for host H2O2 molecule solvation and transport in both directions, highlighting the fundamental role of water nanoconfinement for successful transduction and molecular selection. Overall, the bidirectional nature of the water flux under equilibrium conditions along with the mimicking behavior of hydrogen peroxide during a conductance event introduce a new chemical paradigm never reported so far in any theoretical paper involving any aquaporin isoform.
Collapse
Affiliation(s)
- Darren Wragg
- Department of Chemistry, Technical University of Munich Lichtenbergstr. 4 85748 Garching Germany
| | - Stefano Leoni
- School of Chemistry, Cardiff University Park Place CF103AT Cardiff UK
| | - Angela Casini
- Department of Chemistry, Technical University of Munich Lichtenbergstr. 4 85748 Garching Germany
| |
Collapse
|
23
|
Minori K, Rosa LB, Bonsignore R, Casini A, Miguel DC. Comparing the Antileishmanial Activity of Gold(I) and Gold(III) Compounds in L. amazonensis and L. braziliensis in Vitro. ChemMedChem 2020; 15:2146-2150. [PMID: 32830445 PMCID: PMC7756297 DOI: 10.1002/cmdc.202000536] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Indexed: 12/13/2022]
Abstract
A series of mononuclear coordination or organometallic AuI /AuIII complexes (1-9) have been comparatively studied in vitro for their antileishmanial activity against promastigotes and amastigotes, the clinically relevant parasite form, of Leishmania amazonensis and Leishmania braziliensis. One of the cationic AuI bis-N-heterocyclic carbenes (3) has low EC50 values (ca. 4 μM) in promastigotes cells and no toxicity in host macrophages. Together with two other AuIII complexes (6 and 7), the compound is also extremely effective in intracellular amastigotes from L. amazonensis. Initial mechanistic studies include an evaluation of the gold complexes' effect on L. amazonensis' plasma membrane integrity.
Collapse
Affiliation(s)
- Karen Minori
- Department of Animal Biology, Biology InstituteUniversity of Campinas (UNICAMP)Rua Monteiro Lobato, 25513083-862.CampinasSPBrazil
| | - Letícia B. Rosa
- Department of Animal Biology, Biology InstituteUniversity of Campinas (UNICAMP)Rua Monteiro Lobato, 25513083-862.CampinasSPBrazil
| | - Riccardo Bonsignore
- Department of ChemistryTechnical University of Munich (TUM)Lichtenbergstraße 485748Garching b. MünchenGermany
| | - Angela Casini
- Department of ChemistryTechnical University of Munich (TUM)Lichtenbergstraße 485748Garching b. MünchenGermany
| | - Danilo C. Miguel
- Department of Animal Biology, Biology InstituteUniversity of Campinas (UNICAMP)Rua Monteiro Lobato, 25513083-862.CampinasSPBrazil
| |
Collapse
|
24
|
Janoš P, Spinello A, Magistrato A. All-atom simulations to studying metallodrugs/target interactions. Curr Opin Chem Biol 2020; 61:1-8. [PMID: 32781390 DOI: 10.1016/j.cbpa.2020.07.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 07/03/2020] [Accepted: 07/12/2020] [Indexed: 12/16/2022]
Abstract
Metallodrugs are extensively used to treat and diagnose distinct disease types. The unique physical-chemical properties of metal ions offer tantalizing opportunities to tailor effective scaffolds for selectively targeting specific biomolecules. Modern experimental techniques have collected a large body of structural data concerning the interactions of metallodrugs with their biomolecular targets, although being unable to exhaustively assess the molecular basis of their mechanism of action. In this scenario, the complementary use of accurate computational methods allows uncovering the minutiae of metallodrugs/targets interactions and their underlying mechanism of action at an atomic-level of detail. This knowledge is increasingly perceived as an invaluable requirement to rationally devise novel and selective metallodrugs. Building on literature studies, selected largely from the last 2 years, this compendium encompasses a cross-section of the current role, advances, and challenges met by computer simulations to decipher the mechanistic intricacies of prototypical metallodrugs.
Collapse
Affiliation(s)
- Pavel Janoš
- CNR-IOM c/o SISSA, Via Bonomea 265, 34136, Trieste, Italy
| | | | | |
Collapse
|
25
|
Neumann LSM, Dias AHS, Skaf MS. Molecular Modeling of Aquaporins from Leishmania major. J Phys Chem B 2020; 124:5825-5836. [PMID: 32551664 DOI: 10.1021/acs.jpcb.0c03550] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Aquaporins are membrane proteins responsible for permeating water, ions, dissolved gases, and other small molecular weight compounds through the protective cell membranes of living organisms. These proteins have been gaining increased importance as targets for treating a variety of parasitic diseases, since they control key physiological processes in the life cycle of parasitic protozoans, such as the uptake of nutrients, release of metabolites, and alleviation of osmotic stress. In this work, we use homology modeling to build three-dimensional structures for the four main aquaporins encoded and expressed by Leishmania major, a protozoan that causes leishmaniasis and affects millions of people worldwide. Physico-chemical properties of the proposed models for LmAQP1, LmAQPα, LmAQPβ, and LmAQPγ are then investigated using molecular dynamics simulations and the reference interaction site model (RISM) molecular theory of solvation. Pore characteristics, water permeation, and potential of mean force across the AQP channels for water, methanol, urea, ammonia, and carbon dioxide are examined and compared with results obtained for a protozoan (Plasmodium falciparum) aquaporin for which a crystal structure is available.
Collapse
Affiliation(s)
- Lucas S M Neumann
- Institute of Chemistry and Center for Computing in Engineering and Sciences, University of Campinas, Campinas, SP 13084-862, Brazil
| | - Artur H S Dias
- Institute of Chemistry and Center for Computing in Engineering and Sciences, University of Campinas, Campinas, SP 13084-862, Brazil
| | - Munir S Skaf
- Institute of Chemistry and Center for Computing in Engineering and Sciences, University of Campinas, Campinas, SP 13084-862, Brazil
| |
Collapse
|
26
|
Yadav DK, Kumar S, Choi EH, Chaudhary S, Kim MH. Computational Modeling on Aquaporin-3 as Skin Cancer Target: A Virtual Screening Study. Front Chem 2020; 8:250. [PMID: 32351935 PMCID: PMC7175779 DOI: 10.3389/fchem.2020.00250] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 03/17/2020] [Indexed: 12/25/2022] Open
Abstract
Aquaporin-3 (AQP3) is one of the aquaglyceroporins, which is expressed in the basolateral layer of the skin membrane. Studies have reported that human skin squamous cell carcinoma overexpresses AQP3 and inhibition of its function may alleviate skin tumorigenesis. In the present study, we have applied a virtual screening method that encompasses filters for physicochemical properties and molecular docking to select potential hit compounds that bind to the Aquaporin-3 protein. Based on molecular docking results, the top 20 hit compounds were analyzed for stability in the binding pocket using unconstrained molecular dynamics simulations and further evaluated for binding free energy. Furthermore, examined the ligand-unbinding pathway of the inhibitor from its bound form to explore possible routes for inhibitor approach to the ligand-binding site. With a good docking score, stability in the binding pocket, and free energy of binding, these hit compounds can be developed as Aquaporin-3 inhibitors in the near future.
Collapse
Affiliation(s)
- Dharmendra Kumar Yadav
- Gachon Institute of Pharmaceutical Science & Department of Pharmacy, College of Pharmacy, Gachon University, Incheon, South Korea
| | - Surendra Kumar
- Gachon Institute of Pharmaceutical Science & Department of Pharmacy, College of Pharmacy, Gachon University, Incheon, South Korea
| | - Eun-Ha Choi
- Plasma Bioscience Research Center/PDP Research Center, Kwangwoon University, Nowon-Gu, South Korea
| | - Sandeep Chaudhary
- Laboratory of Organic & Medicinal Chemistry, Department of Chemistry, Malaviya National Institute of Technology, Jaipur, India
| | - Mi-Hyun Kim
- Gachon Institute of Pharmaceutical Science & Department of Pharmacy, College of Pharmacy, Gachon University, Incheon, South Korea
| |
Collapse
|
27
|
A Novel Hybrid Nanosystem Integrating Cytotoxic and Magnetic Properties as a Tool to Potentiate Melanoma Therapy. NANOMATERIALS 2020; 10:nano10040693. [PMID: 32268611 PMCID: PMC7221742 DOI: 10.3390/nano10040693] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 03/27/2020] [Accepted: 03/31/2020] [Indexed: 12/11/2022]
Abstract
Cancer is a major health concern and the prognosis is often poor. Significant advances in nanotechnology are now driving a revolution in cancer detection and treatment. The goal of this study was to develop a novel hybrid nanosystem for melanoma treatment, integrating therapeutic and magnetic targeting modalities. Hence, we designed long circulating and pH-sensitive liposomes loading both dichloro(1,10-phenanthroline) copper (II) (Cuphen), a cytotoxic metallodrug, and iron oxide nanoparticles (IONPs). The synthetized IONPs were characterized by transmission electron microscopy and dynamic light scattering. Lipid-based nanoformulations were prepared by the dehydration rehydration method, followed by an extrusion step for reducing and homogenizing the mean size. Liposomes were characterized in terms of incorporation parameters and mean size. High Cuphen loadings were obtained and the presence of IONPs slightly reduced Cuphen incorporation parameters. Cuphen antiproliferative properties were preserved after association to liposomes and IONPs (at 2 mg/mL) did not interfere on cellular proliferation of murine and human melanoma cell lines. Moreover, the developed nanoformulations displayed magnetic properties. The absence of hemolytic activity for formulations under study demonstrated their safety for parenteral administration. In conclusion, a lipid-based nanosystem loading the cytotoxic metallodrug, Cuphen, and displaying magnetic properties was successfully designed.
Collapse
|
28
|
Pimpão C, da Silva IV, Mósca AF, Pinho JO, Gaspar MM, Gumerova NI, Rompel A, Aureliano M, Soveral G. The Aquaporin-3-Inhibiting Potential of Polyoxotungstates. Int J Mol Sci 2020; 21:E2467. [PMID: 32252345 PMCID: PMC7177757 DOI: 10.3390/ijms21072467] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 03/25/2020] [Accepted: 03/31/2020] [Indexed: 02/07/2023] Open
Abstract
Polyoxometalates (POMs) are of increasing interest due to their proven anticancer activities. Aquaporins (AQPs) were found to be overexpressed in tumors bringing particular attention to their inhibitors as anticancer drugs. Herein, we report for the first time the ability of polyoxotungstates (POTs), such as of Wells-Dawson P2W18, P2W12, and P2W15, and Preyssler P5W30 structures, to affect aquaporin-3 (AQP3) activity and impair melanoma cell migration. The tested POTs were revealed to inhibit AQP3 function with different effects, with P2W18, P2W12, and P5W30 being the most potent (50% inhibitory concentration (IC50) = 0.8, 2.8, and 3.2 µM), and P2W15 being the weakest (IC50 > 100 µM). The selectivity of P2W18 toward AQP3 was confirmed in yeast cells transformed with human aquaglyceroporins. The effect of P2W12 and P2W18 on melanoma cells that highly express AQP3 revealed an impairment of cell migration between 55% and 65% after 24 h, indicating that the anticancer properties of these compounds may in part be due to the blockage of AQP3-mediated permeability. Altogether, our data revealed that P2W18 strongly affects AQP3 activity and cancer cell growth, unveiling its potential as an anticancer drug against tumors where AQP3 is highly expressed.
Collapse
Affiliation(s)
- Catarina Pimpão
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal; (C.P.); (I.V.d.S.); (A.F.M.); (J.O.P.); (M.M.G.)
- Department of Biochemistry and Human Biology, Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | - Inês V. da Silva
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal; (C.P.); (I.V.d.S.); (A.F.M.); (J.O.P.); (M.M.G.)
- Department of Biochemistry and Human Biology, Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | - Andreia F. Mósca
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal; (C.P.); (I.V.d.S.); (A.F.M.); (J.O.P.); (M.M.G.)
- Department of Biochemistry and Human Biology, Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | - Jacinta O. Pinho
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal; (C.P.); (I.V.d.S.); (A.F.M.); (J.O.P.); (M.M.G.)
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | - Maria Manuela Gaspar
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal; (C.P.); (I.V.d.S.); (A.F.M.); (J.O.P.); (M.M.G.)
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | - Nadiia I. Gumerova
- Universität Wien, Fakultät für Chemie, Institut für Biophysikalische Chemie, 1090 Vienna, Austria; (N.I.G.); (A.R.)
| | - Annette Rompel
- Universität Wien, Fakultät für Chemie, Institut für Biophysikalische Chemie, 1090 Vienna, Austria; (N.I.G.); (A.R.)
| | - Manuel Aureliano
- Faculdade de Ciências e Tecnologia (FCT), CCMar, Universidade do Algarve, 8005-139 Faro, Portugal
| | - Graça Soveral
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal; (C.P.); (I.V.d.S.); (A.F.M.); (J.O.P.); (M.M.G.)
- Department of Biochemistry and Human Biology, Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| |
Collapse
|
29
|
Bonsignore R, Thomas SR, Klooster WT, Coles SJ, Jenkins RL, Bourissou D, Barone G, Casini A. Carbon-Phosphorus Coupling from C^N Cyclometalated Au III Complexes. Chemistry 2020; 26:4226-4231. [PMID: 31994237 PMCID: PMC7187188 DOI: 10.1002/chem.201905392] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 01/10/2020] [Indexed: 12/20/2022]
Abstract
With the aim of exploiting new organometallic species for cross-coupling reactions, we report here on the AuIII -mediated Caryl -P bond formation occurring upon reaction of C^N cyclometalated AuIII complexes with phosphines. The [Au(C^N)Cl2 ] complex 1 featuring the bidentate 2-benzoylpyridine (CCO N) scaffold was found to react with PTA (1,3,5-triaza-7-phosphaadamantane) under mild conditions, including in water, to afford the corresponding phosphonium 5 through C-P reductive elimination. A mechanism is proposed for the title reaction based on in situ 31 P{1 H} NMR and HR-ESI-MS analyses combined with DFT calculations. The C-P coupling has been generalized to other C^N cyclometalated AuIII complexes and other tertiary phosphines. Overall, this work provides new insights into the reactivity of cyclometalated AuIII compounds and establishes initial structure-activity relationships to develop AuIII -mediated C-P cross-coupling reactions.
Collapse
Affiliation(s)
- Riccardo Bonsignore
- School of ChemistryCardiff UniversityMain Building, Park PlaceCF10 3ATCardiffUK
| | - Sophie R. Thomas
- School of ChemistryCardiff UniversityMain Building, Park PlaceCF10 3ATCardiffUK
| | - Wim T. Klooster
- School of ChemistryUniversity of SouthamptonSouthamptonSO17 1BJUK
| | - Simon J. Coles
- School of ChemistryUniversity of SouthamptonSouthamptonSO17 1BJUK
| | - Robert L. Jenkins
- School of ChemistryCardiff UniversityMain Building, Park PlaceCF10 3ATCardiffUK
| | - Didier Bourissou
- CNRS/Université Paul SabatierLaboratoire Hétérochimie Fondamentale et Appliquée (LHFA, UMR 5069)118 Route de Narbonne31062Toulouse Cedex 09France
| | - Giampaolo Barone
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e FarmaceuticheUniversità degli Studi di PalermoViale delle Scienze, Edificio 1790128PalermoItaly
| | - Angela Casini
- School of ChemistryCardiff UniversityMain Building, Park PlaceCF10 3ATCardiffUK
- Department of ChemistryTechnical University of MunichLichtenbergstr. 485747GarchingGermany
| |
Collapse
|
30
|
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]
|
31
|
Oliveira Pinho J, Matias M, Gaspar MM. Emergent Nanotechnological Strategies for Systemic Chemotherapy against Melanoma. NANOMATERIALS (BASEL, SWITZERLAND) 2019; 9:E1455. [PMID: 31614947 PMCID: PMC6836019 DOI: 10.3390/nano9101455] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 10/04/2019] [Accepted: 10/10/2019] [Indexed: 12/24/2022]
Abstract
Melanoma is an aggressive form of skin cancer, being one of the deadliest cancers in the world. The current treatment options involve surgery, radiotherapy, targeted therapy, immunotherapy and the use of chemotherapeutic agents. Although the last approach is the most used, the high toxicity and the lack of efficacy in advanced stages of the disease have demanded the search for novel bioactive molecules and/or efficient drug delivery systems. The current review aims to discuss the most recent advances on the elucidation of potential targets for melanoma treatment, such as aquaporin-3 and tyrosinase. In addition, the role of nanotechnology as a valuable strategy to effectively deliver selective drugs is emphasized, either incorporating/encapsulating synthetic molecules or natural-derived compounds in lipid-based nanosystems such as liposomes. Nanoformulated compounds have been explored for their improved anticancer activity against melanoma and promising results have been obtained. Indeed, they displayed improved physicochemical properties and higher accumulation in tumoral tissues, which potentiated the efficacy of the compounds in pre-clinical experiments. Overall, these experiments opened new doors for the discovery and development of more effective drug formulations for melanoma treatment.
Collapse
Affiliation(s)
- Jacinta Oliveira Pinho
- Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal.
| | - Mariana Matias
- Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal.
| | - Maria Manuela Gaspar
- Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal.
| |
Collapse
|
32
|
Wenzel MN, Bonsignore R, Thomas SR, Bourissou D, Barone G, Casini A. Cyclometalated Au III Complexes for Cysteine Arylation in Zinc Finger Protein Domains: towards Controlled Reductive Elimination. Chemistry 2019; 25:7628-7634. [PMID: 30990916 PMCID: PMC6594228 DOI: 10.1002/chem.201901535] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Indexed: 12/14/2022]
Abstract
With the aim of exploiting the use of organometallic species for the efficient modification of proteins through C-atom transfer, the gold-mediated cysteine arylation through a reductive elimination process occurring from the reaction of cyclometalated AuIII C^N complexes with a zinc finger peptide (Cys2 His2 type) is here reported. Among the four selected AuIII cyclometalated compounds, the [Au(CCO N)Cl2 ] complex featuring the 2-benzoylpyridine (CCO N) scaffold was identified as the most prone to reductive elimination and Cys arylation in buffered aqueous solution (pH 7.4) at 37 °C by high-resolution LC electrospray ionization mass spectrometry. DFT and quantum mechanics/molecular mechanics (QM/MM) studies permitted to propose a mechanism for the title reaction that is in line with the experimental results. Overall, the results provide new insights into the reactivity of cytotoxic organogold compounds with biologically important zinc finger domains and identify initial structure-activity relationships to enable AuIII -catalyzed reductive elimination in aqueous media.
Collapse
Affiliation(s)
- Margot N. Wenzel
- School of ChemistryCardiff UniversityMain Building, Park PlaceCF10 3ATCardiffUK
| | - Riccardo Bonsignore
- School of ChemistryCardiff UniversityMain Building, Park PlaceCF10 3ATCardiffUK
| | - Sophie R. Thomas
- School of ChemistryCardiff UniversityMain Building, Park PlaceCF10 3ATCardiffUK
| | - Didier Bourissou
- CNRS/Université Paul SabatierLaboratoire Hétérochimie Fondamentale et Appliquée (LHFA, UMR 5069)118 Route de Narbonne31062Toulouse Cedex 09France
| | - Giampaolo Barone
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e FarmaceuticheUniversità di PalermoViale delle Scienze, Edificio 1790128PalermoItaly
| | - Angela Casini
- School of ChemistryCardiff UniversityMain Building, Park PlaceCF10 3ATCardiffUK
| |
Collapse
|
33
|
Wragg D, de Almeida A, Casini A, Leoni S. Unveiling the Mechanisms of Aquaglyceroporin‐3 Water and Glycerol Permeation by Metadynamics. Chemistry 2019; 25:8713-8718. [DOI: 10.1002/chem.201902121] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Indexed: 12/29/2022]
Affiliation(s)
- Darren Wragg
- School of ChemistryCardiff University Park Place CF10 3AT Cardiff UK
| | - Andreia de Almeida
- Tumour Micro Environment Group, Division of Cancer and GeneticsSchool of MedicineCardiff University Tenovus Building Cardiff CF14 4XN UK
| | - Angela Casini
- School of ChemistryCardiff University Park Place CF10 3AT Cardiff UK
| | - Stefano Leoni
- School of ChemistryCardiff University Park Place CF10 3AT Cardiff UK
| |
Collapse
|
34
|
Mazzei L, Wenzel MN, Cianci M, Palombo M, Casini A, Ciurli S. Inhibition Mechanism of Urease by Au(III) Compounds Unveiled by X-ray Diffraction Analysis. ACS Med Chem Lett 2019; 10:564-570. [PMID: 30996797 DOI: 10.1021/acsmedchemlett.8b00585] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 01/04/2019] [Indexed: 11/29/2022] Open
Abstract
The nickel-dependent enzyme urease is a virulence factor for a large number of critical human pathogens, making this enzyme a potential target of therapeutics for the treatment of resistant bacterial infections. In the search for novel urease inhibitors, five selected coordination and organometallic Au(III) compounds containing N∧N or C∧N and C∧N∧N ligands were tested for their inhibitory effects against Canavalia ensiformis (jack bean) urease. The results showed potent inhibition effects with IC50 values in the nanomolar range. The 2.14 Å resolution crystal structure of Sporosarcina pasteurii urease inhibited by the most effective Au(III) compound [Au(PbImMe)Cl2]PF6 (PbImMe = 1-methyl-2-(pyridin-2-yl)-benzimidazole) reveals the presence of two Au ions bound to the conserved triad αCys322/αHis323/αMet367. The binding of the Au ions to these residues blocks the movement of a flap, located at the edge of the active site channel and essential for enzyme catalysis, completely obliterating the catalytic activity of urease. Overall, the obtained results constitute the basis for the design of new gold complexes as selective urease inhibitors with future antibacterial applications.
Collapse
Affiliation(s)
- Luca Mazzei
- Laboratory of Bioinorganic Chemistry, Department of Pharmacy and Biotechnology, University of Bologna, Viale Giuseppe Fanin 40, I-40127 Bologna, Italy
| | - Margot N. Wenzel
- School of Chemistry, Cardiff University, Main Building, Park Place, CF10 3AT Cardiff, United Kingdom
| | - Michele Cianci
- Department of Agricultural, Food and Environmental Sciences, Marche Polytechnic University, Via Brecce Bianche, I-60131 Ancona, Italy
| | - Marta Palombo
- Laboratory of Bioinorganic Chemistry, Department of Pharmacy and Biotechnology, University of Bologna, Viale Giuseppe Fanin 40, I-40127 Bologna, Italy
| | - Angela Casini
- School of Chemistry, Cardiff University, Main Building, Park Place, CF10 3AT Cardiff, United Kingdom
| | - Stefano Ciurli
- Laboratory of Bioinorganic Chemistry, Department of Pharmacy and Biotechnology, University of Bologna, Viale Giuseppe Fanin 40, I-40127 Bologna, Italy
| |
Collapse
|
35
|
Aikman B, de Almeida A, Meier-Menches SM, Casini A. Aquaporins in cancer development: opportunities for bioinorganic chemistry to contribute novel chemical probes and therapeutic agents. Metallomics 2019; 10:696-712. [PMID: 29766198 DOI: 10.1039/c8mt00072g] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Aquaporins (AQPs) are membrane proteins allowing permeation of water, glycerol & hydrogen peroxide across biomembranes, and playing an important role in water homeostasis in different organs, exocrine gland secretion, urine concentration, skin moisturization, fat metabolism and neural signal transduction. Notably, a large number of studies showed that AQPs are closely associated with cancer biological functions and expressed in more than 20 human cancer cell types. Furthermore, AQP expression is positively correlated with tumour types, grades, proliferation, migration, angiogenesis, as well as tumour-associated oedema, rendering these membrane channels attractive as both diagnostic and therapeutic targets in cancer. Recent developments in the field of AQPs modulation have identified coordination metal-based complexes as potent and selective inhibitors of aquaglyceroporins, opening new avenues in the application of inorganic compounds in medicine and chemical biology. The present review is aimed at providing an overview on AQP structure and function, mainly in relation to cancer. In this context, the exploration of coordination metal compounds as possible inhibitors of aquaporins may open the way to novel chemical approaches to study AQP roles in tumour growth and potentially to new drug families. Thus, we describe recent results in the field and reflect upon the potential of inorganic chemistry in providing compounds to modulate the activity of "elusive" membrane targets as the aquaporins.
Collapse
Affiliation(s)
- Brech Aikman
- School of Chemistry, Cardiff University, Main Building, Park Place, Cardiff CF10 3AT, UK.
| | | | | | | |
Collapse
|
36
|
Wenzel MN, Mósca AF, Graziani V, Aikman B, Thomas SR, de Almeida A, Platts JA, Re N, Coletti C, Marrone A, Soveral G, Casini A. Insights into the Mechanisms of Aquaporin-3 Inhibition by Gold(III) Complexes: the Importance of Non-Coordinative Adduct Formation. Inorg Chem 2019; 58:2140-2148. [DOI: 10.1021/acs.inorgchem.8b03233] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Margot N. Wenzel
- School of Chemistry, Cardiff University, Main Building, Park Place, CF10 3AT Cardiff, United Kingdom
| | - Andreia F. Mósca
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisboa, Portugal
| | - Valentina Graziani
- Università“G. d’Annunzio” di Chieti-Pescara, Department of Pharmacy, Via dei Vestini 31, 66100 Chieti, Italy
| | - Brech Aikman
- School of Chemistry, Cardiff University, Main Building, Park Place, CF10 3AT Cardiff, United Kingdom
| | - Sophie R. Thomas
- School of Chemistry, Cardiff University, Main Building, Park Place, CF10 3AT Cardiff, United Kingdom
| | - Andreia de Almeida
- School of Chemistry, Cardiff University, Main Building, Park Place, CF10 3AT Cardiff, United Kingdom
- Tumour Microenvironment Group, Division of Cancer and Genetics, School of Medicine, Cardiff University, Tenovus Building, Cardiff CF14 4XN, United Kingdom
| | - James A. Platts
- School of Chemistry, Cardiff University, Main Building, Park Place, CF10 3AT Cardiff, United Kingdom
| | - Nazzareno Re
- Università“G. d’Annunzio” di Chieti-Pescara, Department of Pharmacy, Via dei Vestini 31, 66100 Chieti, Italy
| | - Cecilia Coletti
- Università“G. d’Annunzio” di Chieti-Pescara, Department of Pharmacy, Via dei Vestini 31, 66100 Chieti, Italy
| | - Alessandro Marrone
- Università“G. d’Annunzio” di Chieti-Pescara, Department of Pharmacy, Via dei Vestini 31, 66100 Chieti, Italy
| | - Graça Soveral
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisboa, Portugal
| | - Angela Casini
- School of Chemistry, Cardiff University, Main Building, Park Place, CF10 3AT Cardiff, United Kingdom
| |
Collapse
|
37
|
Ferraro G, Giorgio A, Mansour AM, Merlino A. Protein-mediated disproportionation of Au(i): insights from the structures of adducts of Au(iii) compounds bearingN,N-pyridylbenzimidazole derivatives with lysozyme. Dalton Trans 2019; 48:14027-14035. [DOI: 10.1039/c9dt02729g] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Structural data of protein/gold adducts suggest protein-mediated reduction of Au(iii) into Au(i) and disproportionation of Au(i) into Au(iii) and Au(0).
Collapse
Affiliation(s)
- Giarita Ferraro
- Department of Chemistry “Ugo Schiff”
- University of Florence
- Sesto Fiorentino
- Italy
| | - Anna Giorgio
- Department of Chemical Sciences
- University of Naples Federico II
- Complesso Universitario di Monte Sant'Angelo
- Naples
- Italy
| | | | - Antonello Merlino
- Department of Chemical Sciences
- University of Naples Federico II
- Complesso Universitario di Monte Sant'Angelo
- Naples
- Italy
| |
Collapse
|
38
|
Gold(III) Pyridine-Benzimidazole Complexes as Aquaglyceroporin Inhibitors and Antiproliferative Agents. INORGANICS 2018. [DOI: 10.3390/inorganics6040123] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Gold compounds have been proven to be novel and versatile tools for biological applications, including as anticancer agents. Recently, we explored the potential of Au(III) complexes with bi-dentate N-donor ligands as inhibitors of the membrane water and glycerol channels aquaporins (AQPs), involved in different physiological and pathophysiological pathways. Here, eight new Au(III) complexes featuring a pyridine-benzimidazole scaffold have been synthesized and characterized via different methods. The stability of all the compounds in aqueous solution and their reactivity with glutathione have been investigated by UV–visible spectroscopy. The Au(III) compounds, tested for their AQPs inhibition properties in human Red Blood Cells (hRBC), are potent and selective inhibitors of AQP3. Furthermore, the compounds’ antiproliferative effects have been studied in a small panel of human cancer cells expressing AQP3. The complexes show only very moderate anticancer effects in vitro and are mostly active against the melanoma A375 cells, with marked expression of AQP3 at the level of the nuclear membrane. In general, the AQP3 inhibition properties of these complexes hold promises to develop them as chemical probes to study the function of this protein isoform in biological systems.
Collapse
|
39
|
Calamita G, Perret J, Delporte C. Aquaglyceroporins: Drug Targets for Metabolic Diseases? Front Physiol 2018; 9:851. [PMID: 30042691 PMCID: PMC6048697 DOI: 10.3389/fphys.2018.00851] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 06/15/2018] [Indexed: 12/29/2022] Open
Abstract
Aquaporins (AQPs) are a family of transmembrane channel proteins facilitating the transport of water, small solutes, and gasses across biological membranes. AQPs are expressed in all tissues and ensure multiple roles under normal and pathophysiological conditions. Aquaglyceroporins are a subfamily of AQPs permeable to glycerol in addition to water and participate thereby to energy metabolism. This review focalizes on the present knowledge of the expression, regulation and physiological roles of AQPs in adipose tissue, liver and endocrine pancreas, that are involved in energy metabolism. In addition, the review aims at summarizing the involvement of AQPs in metabolic disorders, such as obesity, diabetes and liver diseases. Finally, challenges and recent advances related to pharmacological modulation of AQPs expression and function to control and treat metabolic diseases are discussed.
Collapse
Affiliation(s)
- Giuseppe Calamita
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari Aldo Moro, Bari, Italy
| | - Jason Perret
- Laboratory of Pathophysiological and Nutritional Biochemistry, Université Libre de Bruxelles, Brussels, Belgium
| | - Christine Delporte
- Laboratory of Pathophysiological and Nutritional Biochemistry, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
40
|
Bertrand B, Williams MRM, Bochmann M. Gold(III) Complexes for Antitumor Applications: An Overview. Chemistry 2018; 24:11840-11851. [DOI: 10.1002/chem.201800981] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 03/22/2018] [Indexed: 12/20/2022]
Affiliation(s)
- Benoît Bertrand
- School of ChemistryUniversity of East Anglia Norwich NR4 7TJ United Kingdom
- Sorbonne UniversitésUPMC Univ Paris 06CNRSInstitut Parisien de Chimie Moléculaire (IPCM) 4 Place Jussieu 75005 Paris France
| | | | - Manfred Bochmann
- School of ChemistryUniversity of East Anglia Norwich NR4 7TJ United Kingdom
| |
Collapse
|
41
|
Wenzel MN, Meier-Menches SM, Williams TL, Rämisch E, Barone G, Casini A. Selective targeting of PARP-1 zinc finger recognition domains with Au(iii) organometallics. Chem Commun (Camb) 2018; 54:611-614. [DOI: 10.1039/c7cc08406d] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Insights into gold finger formation by organometallics and implications for targeting pharmacologically relevant zinc-finger proteins.
Collapse
Affiliation(s)
| | | | | | - Eberard Rämisch
- Heraeus Deutschland GmbH & Co. KG
- GBU
- Heraeus Chemicals
- Germany
| | - Giampaolo Barone
- Dipartimento di Scienze e Tecnologie Biologiche
- Chimiche e Farmaceutiche
- Università di Palermo
- Viale delle Scienze
- Italy
| | - Angela Casini
- School of Chemistry
- Cardiff University
- Main Building
- Park Place
- UK
| |
Collapse
|
42
|
Graziani V, Marrone A, Re N, Coletti C, Platts JA, Casini A. A Multi-Level Theoretical Study to Disclose the Binding Mechanisms of Gold(III)-Bipyridyl Compounds as Selective Aquaglyceroporin Inhibitors. Chemistry 2017; 23:13802-13813. [DOI: 10.1002/chem.201703092] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Indexed: 11/11/2022]
Affiliation(s)
- Valentina Graziani
- Department of Pharmacy; Università “G d'Annunzio” di Chieti-Pescara; Via dei Vestini 31 66100 Chieti Italy
| | - Alessandro Marrone
- Department of Pharmacy; Università “G d'Annunzio” di Chieti-Pescara; Via dei Vestini 31 66100 Chieti Italy
| | - Nazzareno Re
- Department of Pharmacy; Università “G d'Annunzio” di Chieti-Pescara; Via dei Vestini 31 66100 Chieti Italy
| | - Cecilia Coletti
- Department of Pharmacy; Università “G d'Annunzio” di Chieti-Pescara; Via dei Vestini 31 66100 Chieti Italy
| | - James A. Platts
- School of Chemistry; Cardiff University, Park Place; Cardiff CF10 3AT UK
| | - Angela Casini
- School of Chemistry; Cardiff University, Park Place; Cardiff CF10 3AT UK
| |
Collapse
|
43
|
Tavares TT, Azevedo GC, Garcia A, Carpanez AG, Lewer PM, Paschoal D, Müller BL, Dos Santos HF, Matos RC, Silva H, Grazul RM, Fontes APS. Gold(I) complexes with aryl-thiosemicarbazones: Molecular modeling, synthesis, cytotoxicity and TrxR inhibition. Polyhedron 2017. [DOI: 10.1016/j.poly.2017.05.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|