1
|
Koyama S, Weber EL, Heinbockel T. Possible Combinatorial Utilization of Phytochemicals and Extracellular Vesicles for Wound Healing and Regeneration. Int J Mol Sci 2024; 25:10353. [PMID: 39408681 PMCID: PMC11476926 DOI: 10.3390/ijms251910353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/16/2024] [Accepted: 09/17/2024] [Indexed: 10/20/2024] Open
Abstract
Organ and tissue damage can result from injury and disease. How to facilitate regeneration from damage has been a topic for centuries, and still, we are trying to find agents to use for treatments. Two groups of biological substances are known to facilitate wound healing. Phytochemicals with bioactive properties form one group. Many phytochemicals have anti-inflammatory effects and enhance wound healing. Recent studies have described their effects at the gene and protein expression levels, highlighting the receptors and signaling pathways involved. The extremely large number of phytochemicals and the multiple types of receptors they activate suggest a broad range of applicability for their clinical use. The hydrophobic nature of many phytochemicals and the difficulty with chemical stabilization have been a problem. Recent developments in biotechnology and nanotechnology methods are enabling researchers to overcome these problems. The other group of biological substances is extracellular vesicles (EVs), which are now known to have important biological functions, including the improvement of wound healing. The proteins and nanoparticles contained in mammalian EVs as well as the specificity of the targets of microRNAs included in the EVs are becoming clear. Plant-derived EVs have been found to contain phytochemicals. The overlap in the wound-healing capabilities of both phytochemicals and EVs and the differences in their nature suggest the possibility of a combinatorial use of the two groups, which may enhance their effects.
Collapse
Affiliation(s)
- Sachiko Koyama
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Erin L. Weber
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| | - Thomas Heinbockel
- Department of Anatomy, College of Medicine, Howard University, Washington, DC 20059, USA
| |
Collapse
|
2
|
Jakova E, Aigbogun OP, Moutaoufik MT, Allen KJH, Munir O, Brown D, Taghibiglou C, Babu M, Phenix CP, Krol ES, Cayabyab FS. The Bifunctional Dimer Caffeine-Indan Attenuates α-Synuclein Misfolding, Neurodegeneration and Behavioral Deficits after Chronic Stimulation of Adenosine A1 Receptors. Int J Mol Sci 2024; 25:9386. [PMID: 39273333 PMCID: PMC11395333 DOI: 10.3390/ijms25179386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 08/26/2024] [Accepted: 08/28/2024] [Indexed: 09/15/2024] Open
Abstract
We previously found that chronic adenosine A1 receptor stimulation with N6-Cyclopentyladenosine increased α-synuclein misfolding and neurodegeneration in a novel α-synucleinopathy model, a hallmark of Parkinson's disease. Here, we aimed to synthesize a dimer caffeine-indan linked by a 6-carbon chain to cross the blood-brain barrier and tested its ability to bind α-synuclein, reducing misfolding, behavioral abnormalities, and neurodegeneration in our rodent model. Behavioral tests and histological stains assessed neuroprotective effects of the dimer compound. A rapid synthesis of the 18F-labeled analogue enabled Positron Emission Tomography and Computed Tomography imaging for biodistribution measurement. Molecular docking analysis showed that the dimer binds to α-synuclein N- and C-termini and the non-amyloid-β-component (NAC) domain, similar to 1-aminoindan, and this binding promotes a neuroprotective α-synuclein "loop" conformation. The dimer also binds to the orthosteric binding site for adenosine within the adenosine A1 receptor. Immunohistochemistry and confocal imaging showed the dimer abolished α-synuclein upregulation and aggregation in the substantia nigra and hippocampus, and the dimer mitigated cognitive deficits, anxiety, despair, and motor abnormalities. The 18F-labeled dimer remained stable post-injection and distributed in various organs, notably in the brain, suggesting its potential as a Positron Emission Tomography tracer for α-synuclein and adenosine A1 receptor in Parkinson's disease therapy.
Collapse
Affiliation(s)
- Elisabet Jakova
- Department of Surgery, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Omozojie P. Aigbogun
- Department of Chemistry, University of Saskatchewan, Saskatoon, SK S7N 5C9, Canada
| | | | - Kevin J. H. Allen
- Pharmaceutical and Nutrition Sciences Research Group, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Omer Munir
- Department of Anatomy, Physiology, Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Devin Brown
- Department of Chemistry, University of Saskatchewan, Saskatoon, SK S7N 5C9, Canada
| | - Changiz Taghibiglou
- Department of Anatomy, Physiology, Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Mohan Babu
- Department of Chemistry and Biochemistry, University of Regina, Regina, SK S4S 0A2, Canada
| | - Chris P. Phenix
- Department of Chemistry, University of Saskatchewan, Saskatoon, SK S7N 5C9, Canada
| | - Ed S. Krol
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | | |
Collapse
|
3
|
Asuku AO, Ayinla MT, Olajide TS, Oyerinde TO, Yusuf JA, Bayo-Olugbami AA, Fajemidagba GA. Coffee and Parkinson's disease. PROGRESS IN BRAIN RESEARCH 2024; 289:1-19. [PMID: 39168575 DOI: 10.1016/bs.pbr.2024.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
Parkinson's disease (PD) is a prevalent neurodegenerative disease marked by dopaminergic neuronal loss and misfolded alpha-synuclein (α-syn) accumulation, which results in both motor and cognitive symptoms. Its occurrence grows with age, with a larger prevalence among males. Despite substantial study, effective medicines to reduce or stop the progression of diseases remain elusive. Interest has grown in examining dietary components, such as caffeine present in coffee, for potential medicinal effects. Epidemiological studies imply a lower incidence of PD with coffee drinking, attributable to caffeine's neuroprotective abilities. Beyond caffeine, coffee constituent like chlorogenic acid and cafestol have anti-Parkinsonian benefits. Moreover, coffee use has been related with variations in gut microbiota composition, which may reduce intestinal inflammation and prevent protein misfolding in enteric nerves, perhaps through the microbiota-gut-brain axis. This review gives a summary of the neuroprotective effects of coffee, investigating both its motor and non-motor advantages in individuals with PD as well as in experimental models of PD. We reviewed some bioactive constituents of coffee, their respective interactions with misfolded α-syn accumulation, and its emerging mechanisms associated to the gut microbiome.
Collapse
Affiliation(s)
- Abraham Olufemi Asuku
- Bioresources Development Centre, National Biotechnology Research and Development Agency, Ogbomoso, Oyo, Nigeria; Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Kwara, Nigeria.
| | - Maryam Tayo Ayinla
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Kwara, Nigeria
| | - Tobiloba Samuel Olajide
- Laboratory for Experimental and Translational Neurobiology, University of Medical Sciences, Ondo, Ondo, Nigeria
| | - Toheeb O Oyerinde
- Laboratory for Experimental and Translational Neurobiology, University of Medical Sciences, Ondo, Ondo, Nigeria
| | - Joshua Ayodele Yusuf
- Department of Anatomy, Faculty of Basic Medical Sciences, College of Health Sciences, Ladoke Akintola University of Technology, Ogbomoso, Oyo, Nigeria
| | | | | |
Collapse
|
4
|
Ikizawa S, Hori T, Wijaya TN, Kono H, Bai Z, Kimizono T, Lu W, Tran DP, Kitao A. PaCS-Toolkit: Optimized Software Utilities for Parallel Cascade Selection Molecular Dynamics (PaCS-MD) Simulations and Subsequent Analyses. J Phys Chem B 2024; 128:3631-3642. [PMID: 38578072 PMCID: PMC11033871 DOI: 10.1021/acs.jpcb.4c01271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 03/26/2024] [Accepted: 03/26/2024] [Indexed: 04/06/2024]
Abstract
Parallel cascade selection molecular dynamics (PaCS-MD) is an enhanced conformational sampling method conducted as a "repetition of time leaps in parallel worlds", comprising cycles of multiple molecular dynamics (MD) simulations performed in parallel and selection of the initial structures of MDs for the next cycle. We developed PaCS-Toolkit, an optimized software utility enabling the use of different MD software and trajectory analysis tools to facilitate the execution of the PaCS-MD simulation and analyze the obtained trajectories, including the preparation for the subsequent construction of the Markov state model. PaCS-Toolkit is coded with Python, is compatible with various computing environments, and allows for easy customization by editing the configuration file and specifying the MD software and analysis tools to be used. We present the software design of PaCS-Toolkit and demonstrate applications of PaCS-MD variations: original targeted PaCS-MD to peptide folding; rmsdPaCS-MD to protein domain motion; and dissociation PaCS-MD to ligand dissociation from adenosine A2A receptor.
Collapse
Affiliation(s)
- Shinji Ikizawa
- School
of Life Science and Technology, Tokyo Institute
of Technology, 2-12-2 Ookayama, Meguro, Tokyo 152-8550, Japan
| | - Tatsuki Hori
- School
of Life Science and Technology, Tokyo Institute
of Technology, 2-12-2 Ookayama, Meguro, Tokyo 152-8550, Japan
| | - Tegar Nurwahyu Wijaya
- School
of Life Science and Technology, Tokyo Institute
of Technology, 2-12-2 Ookayama, Meguro, Tokyo 152-8550, Japan
- Department
of Chemistry, Universitas Pertamina, Jl. Teuku Nyak Arief, Simprug, Jakarta 12220, Indonesia
| | - Hiroshi Kono
- School
of Life Science and Technology, Tokyo Institute
of Technology, 2-12-2 Ookayama, Meguro, Tokyo 152-8550, Japan
| | - Zhen Bai
- School
of Life Science and Technology, Tokyo Institute
of Technology, 2-12-2 Ookayama, Meguro, Tokyo 152-8550, Japan
| | - Tatsuhiro Kimizono
- School
of Life Science and Technology, Tokyo Institute
of Technology, 2-12-2 Ookayama, Meguro, Tokyo 152-8550, Japan
| | - Wenbo Lu
- School
of Life Science and Technology, Tokyo Institute
of Technology, 2-12-2 Ookayama, Meguro, Tokyo 152-8550, Japan
| | - Duy Phuoc Tran
- School
of Life Science and Technology, Tokyo Institute
of Technology, 2-12-2 Ookayama, Meguro, Tokyo 152-8550, Japan
| | - Akio Kitao
- School
of Life Science and Technology, Tokyo Institute
of Technology, 2-12-2 Ookayama, Meguro, Tokyo 152-8550, Japan
| |
Collapse
|
5
|
Babou Kammoe RB, Sévigny J. Extracellular nucleotides in smooth muscle contraction. Biochem Pharmacol 2024; 220:116005. [PMID: 38142836 DOI: 10.1016/j.bcp.2023.116005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 12/18/2023] [Accepted: 12/19/2023] [Indexed: 12/26/2023]
Abstract
Extracellular nucleotides and nucleosides are crucial signalling molecules, eliciting diverse biological responses in almost all organs and tissues. These molecules exert their effects by activating specific nucleotide receptors, which are finely regulated by ectonucleotidases that break down their ligands. In this comprehensive review, we aim to elucidate the relevance of extracellular nucleotides as signalling molecules in the context of smooth muscle contraction, considering the modulatory influence of ectonucleotidases on this intricate process. Specifically, we provide a detailed examination of the involvement of extracellular nucleotides in the contraction of non-vascular smooth muscles, including those found in the urinary bladder, the airways, the reproductive system, and the gastrointestinal tract. Furthermore, we present a broader overview of the role of extracellular nucleotides in vascular smooth muscle contraction.
Collapse
Affiliation(s)
- Romuald Brice Babou Kammoe
- Centre de Recherche du CHU de Québec - Université Laval, Québec City, QC G1V 4G2, Canada; Département de microbiologie-infectiologie et d'immunologie, Faculté de Médecine, Université Laval, Québec City, QC G1V 0A6, Canada
| | - Jean Sévigny
- Centre de Recherche du CHU de Québec - Université Laval, Québec City, QC G1V 4G2, Canada; Département de microbiologie-infectiologie et d'immunologie, Faculté de Médecine, Université Laval, Québec City, QC G1V 0A6, Canada.
| |
Collapse
|
6
|
Kim G, Hou X, Byun WS, Kim G, Jarhad DB, Lee G, Hyun YE, Yu J, Lee CS, Qu S, Warnick E, Gao ZG, Kim JY, Ji S, Shin H, Choi JR, Jacobson KA, Lee HW, Lee SK, Jeong LS. Structure-Activity Relationship of Truncated 2,8-Disubstituted-Adenosine Derivatives as Dual A 2A/A 3 Adenosine Receptor Antagonists and Their Cancer Immunotherapeutic Activity. J Med Chem 2023; 66:12249-12265. [PMID: 37603705 PMCID: PMC10896643 DOI: 10.1021/acs.jmedchem.3c00806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2023]
Abstract
Based on hA2AAR structures, a hydrophobic C8-heteroaromatic ring in 5'-truncated adenosine analogues occupies the subpocket tightly, converting hA2AAR agonists into antagonists while maintaining affinity toward hA3AR. The final compounds of 2,8-disubstituted-N6-substituted 4'-thionucleosides, or 4'-oxo, were synthesized from d-mannose and d-erythrono-1,4-lactone, respectively, using a Pd-catalyst-controlled regioselective cross-coupling reaction. All tested compounds completely antagonized hA2AAR, including 5d with the highest affinity (Ki,A2A = 7.7 ± 0.5 nM). The hA2AAR-5d X-ray structure revealed that C8-heteroaromatic rings prevented receptor activation-associated conformational changes. However, the C8-substituted compounds still antagonized hA3AR. Structural SAR features and docking studies supported different binding modes at A2AAR and A3AR, elucidating pharmacophores for receptor activation and selectivity. Favorable pharmacokinetics were demonstrated, in which 5d displayed high oral absorption, moderate half-life, and bioavailability. Also, 5d significantly improved the antitumor effect of anti-PD-L1 in vivo. Overall, this study suggests that the novel dual A2AAR/A3AR nucleoside antagonists would be promising drug candidates for immune-oncology.
Collapse
Affiliation(s)
- Gibae Kim
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Xiyan Hou
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
- College of Life Science, Dalian Minzu University, Dalian 116600, People's Republic of China
| | - Woong Sub Byun
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
- Department of Chemical and Systems Biology, Chem-H and Stanford Cancer Institute, Stanford School of Medicine, Stanford University, Stanford, California 94305, United States
| | - Gyudong Kim
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
- College of Pharmacy & Research Institute of Drug Development, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Dnyandev B Jarhad
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Grim Lee
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Young Eum Hyun
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Jinha Yu
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, South Korea
| | - Chang Soo Lee
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Shuhao Qu
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Eugene Warnick
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes, and Digestive and Kidney Disease, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Zhan-Guo Gao
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes, and Digestive and Kidney Disease, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Ji Yong Kim
- Future Medicine Company Limited, Seoul 06665, Republic of Korea
| | - Seunghee Ji
- HK Inno.N Corporation, Seoul 04551, Republic of Korea
| | - Hyunwoo Shin
- HK Inno.N Corporation, Seoul 04551, Republic of Korea
| | | | - Kenneth A Jacobson
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes, and Digestive and Kidney Disease, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Hyuk Woo Lee
- Future Medicine Company Limited, Seoul 06665, Republic of Korea
| | - Sang Kook Lee
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Lak Shin Jeong
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
- Future Medicine Company Limited, Seoul 06665, Republic of Korea
| |
Collapse
|
7
|
Gutiérrez-Rodelo C, Ochoa-López A, Luis Balderas-López J, Reyes-Ramírez A, Millán-Pacheco C, Favela-Rosales F, Navarrete A. "Eritadenine as a regulator of anxiety Disorders: An experimental and docking Approach". Neurosci Lett 2023; 813:137413. [PMID: 37541318 DOI: 10.1016/j.neulet.2023.137413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 06/15/2023] [Accepted: 07/30/2023] [Indexed: 08/06/2023]
Abstract
Uncertainty persists regarding the specific chemical causal factors and their corresponding behavioral effects in anxiety disorders. Commonly employed first-line treatments for anxiety target G protein-coupled receptors (GPCRs), including inhibitors of monoaminergic systems. Alternatively, emerging natural bioactive strategies offer potential for mitigating adverse effects. Recent investigations have implicated adenosine in anxiety-triggering mechanisms, while eritadenine, an adenosine analog derived from Shiitake mushroom, has displayed promising attributes. This study explores eritadenine's potential as a bioactive substance for anxiety disorders in mice, employing behavioral tests, pentobarbital-sleep induction, and molecular docking. Behavioral test results reveal a pronounced anxiolytic and sedative-hypnotic pharmacological effect of eritadenine. Our findings suggest that eritadenine may modulate locomotor functions mediated by adenosine receptors, with a stronger affinity for binding to A2AAR over A1AR, thus eliciting these effects.
Collapse
Affiliation(s)
- Citlaly Gutiérrez-Rodelo
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad Universitaria, Coyoacán, Ciudad de México 04510, México.
| | - Aurora Ochoa-López
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad Universitaria, Coyoacán, Ciudad de México 04510, México
| | - José Luis Balderas-López
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad Universitaria, Coyoacán, Ciudad de México 04510, México
| | - Adelfo Reyes-Ramírez
- Unidad Multidisciplinaria de Investigación Experimental, Facultad de Estudios Superiores Zaragoza, Universidad Nacional Autónoma de México, Batalla 5 de mayo s/n esquina Fuerte de Loreto, Ejército de Oriente, Iztapalapa, Ciudad de México 09230, México
| | - Cesar Millán-Pacheco
- Facultad de Farmacia. Universidad Autónoma del Estado de Morelos, Cuernavaca Mor. C.P. 62209, México
| | - Fernando Favela-Rosales
- Departamento de Ciencias Básicas, Tecnológico Nacional de México - ITS Zacatecas Occidente, Sombrerete, Zacatecas 99102, México
| | - Andrés Navarrete
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad Universitaria, Coyoacán, Ciudad de México 04510, México.
| |
Collapse
|
8
|
Goßen J, Ribeiro RP, Bier D, Neumaier B, Carloni P, Giorgetti A, Rossetti G. AI-based identification of therapeutic agents targeting GPCRs: introducing ligand type classifiers and systems biology. Chem Sci 2023; 14:8651-8661. [PMID: 37592985 PMCID: PMC10430665 DOI: 10.1039/d3sc02352d] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Accepted: 07/20/2023] [Indexed: 08/19/2023] Open
Abstract
Identifying ligands targeting G protein coupled receptors (GPCRs) with novel chemotypes other than the physiological ligands is a challenge for in silico screening campaigns. Here we present an approach that identifies novel chemotype ligands by combining structural data with a random forest agonist/antagonist classifier and a signal-transduction kinetic model. As a test case, we apply this approach to identify novel antagonists of the human adenosine transmembrane receptor type 2A, an attractive target against Parkinson's disease and cancer. The identified antagonists were tested here in a radio ligand binding assay. Among those, we found a promising ligand whose chemotype differs significantly from all so-far reported antagonists, with a binding affinity of 310 ± 23.4 nM. Thus, our protocol emerges as a powerful approach to identify promising ligand candidates with novel chemotypes while preserving antagonistic potential and affinity in the nanomolar range.
Collapse
Affiliation(s)
- Jonas Goßen
- Institute for Computational Biomedicine (INM-9/IAS-5) Forschungszentrum Jülich Wilhelm-Johnen-Straße 52428 Jülich Germany
- Faculty of Mathematics, Computer Science and Natural Sciences RWTH Aachen University Aachen Germany
| | - Rui Pedro Ribeiro
- Institute for Computational Biomedicine (INM-9/IAS-5) Forschungszentrum Jülich Wilhelm-Johnen-Straße 52428 Jülich Germany
| | - Dirk Bier
- Institute of Neuroscience and Medicine, Nuclear Chemistry (INM-5), Forschungszentrum Jülich GmbH Wilhelm-Johnen-Straße 52428 Jülich Germany
| | - Bernd Neumaier
- Institute of Neuroscience and Medicine, Nuclear Chemistry (INM-5), Forschungszentrum Jülich GmbH Wilhelm-Johnen-Straße 52428 Jülich Germany
- Institute of Radiochemistry and Experimental Molecular Imaging, University of Cologne, Faculty of Medicine and University Hospital Cologne Kerpener Straße 62 50937 Cologne Germany
| | - Paolo Carloni
- Institute for Computational Biomedicine (INM-9/IAS-5) Forschungszentrum Jülich Wilhelm-Johnen-Straße 52428 Jülich Germany
- Faculty of Mathematics, Computer Science and Natural Sciences RWTH Aachen University Aachen Germany
- JARA-Institut Molecular Neuroscience and Neuroimaging (INM-11) Forschungszentrum Jülich Wilhelm-Johnen-Straße 52428 Jülich Germany
| | - Alejandro Giorgetti
- Institute for Computational Biomedicine (INM-9/IAS-5) Forschungszentrum Jülich Wilhelm-Johnen-Straße 52428 Jülich Germany
- Department of Biotechnology University of Verona Verona Italy
| | - Giulia Rossetti
- Institute for Computational Biomedicine (INM-9/IAS-5) Forschungszentrum Jülich Wilhelm-Johnen-Straße 52428 Jülich Germany
- Jülich Supercomputing Centre (JSC) Forschungszentrum Jülich Jülich Germany
- Department of Neurology University Hospital Aachen (UKA), RWTH Aachen University Aachen Germany
| |
Collapse
|
9
|
Paladines N, Dawson S, Ryan W, Serrano-Lopez R, Messer R, Huo Y, Cutler CW, Ramos-Junior ES, Morandini AC. Metabolic reprogramming through mitochondrial biogenesis drives adenosine anti-inflammatory effects: new mechanism controlling gingival fibroblast hyper-inflammatory state. Front Immunol 2023; 14:1148216. [PMID: 37350964 PMCID: PMC10282177 DOI: 10.3389/fimmu.2023.1148216] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 05/25/2023] [Indexed: 06/24/2023] Open
Abstract
Introduction Fibroblasts are the dominant stromal cells in the gingival lamina propria with a well-established relevance in regulation of inflammation, and in innate immunity. This is exemplified by their hypersecretion of CXCL8, enhancing leukocyte infiltration in chronic and sustained inflammatory conditions. We have previously shown adenosine to be a key metabolic nucleoside that regulates stromal inflammation, but the underlying mechanisms linking adenosine to the metabolic status of fibroblasts and to the resultant inflammatory response are unclear. This study examined, by seahorse real-time cell metabolic analysis, the bioenergetics of the stromal fibroblast response to extracellular adenosine and IL-1β, focusing on CXCL8 secretion by primary human gingival fibroblasts (HGF). Methods Markers of the glycolytic pathway and mitochondrial biogenesis were tracked through immunoblot. Further, the influence of adenosine on mitochondrial accumulation was measured by uptake of MitoTracker Red fluorescent probe and assessment of the role of FCCP (a mitochondrial uncoupler) in CXCL8 secretion and mitochondrial accumulation. Results Our results show that the anti-inflammatory response of HGF to extracellular adenosine, typified by reduced CXCL8 secretion, is mediated by mitochondrial oxidative phosphorylation, reflected in higher oxygen consumption rate (OCR). In the presence of IL-1β, adenosine-treated cells induced higher ATP production, basal respiration and proton leak compared to IL-1β without adenosine. Surprisingly, adenosine had no additional effect on the IL-1β-induced higher glycolysis rate demonstrated by the extracellular acidification rate (ECAR). In addition, the higher OCR in adenosine-stimulated cells was not due to the mitochondrial fuel dependency or capacity, but due to an increase in mitochondrial biogenesis and accumulation in the cells with concomitant decrease in mitophagy-required p-PINK1 marker. We detected the accumulation of functional mitochondria with increased activation of the AMPK/SIRT1/PGC-1α pathway. The adenosine-induced uptake of MitoTracker was abrogated by PGC-1α inhibition with SR-12898. In addition, the adenosine effects on reduced CXCL8 were ablated by treatment with FCCP, a potent uncoupler of mitochondrial oxidative phosphorylation. Conclusion Our findings reveal a key role for mitochondrial bioenergetics in regulation of CXCL8-mediated inflammation by HGF through the adenosine/AMPK/SIRT1/PGC-1α axis. Therapeutically targeting this pathway in gingival fibroblasts might be a promising future strategy to modulate stromal-mediated sustained hyper-inflammatory responses.
Collapse
Affiliation(s)
- Nathalie Paladines
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA, United States
| | - Shantiece Dawson
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA, United States
| | - Weston Ryan
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA, United States
| | - Rogelio Serrano-Lopez
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA, United States
| | - Regina Messer
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA, United States
| | - Yuqing Huo
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, United States
- Vascular Biology Center, Augusta University, Augusta, GA, United States
| | - Christopher W. Cutler
- Department of Periodontics, Dental College of Georgia, Augusta University, Augusta, GA, United States
| | - Erivan S. Ramos-Junior
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA, United States
| | - Ana Carolina Morandini
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA, United States
- Department of Periodontics, Dental College of Georgia, Augusta University, Augusta, GA, United States
| |
Collapse
|
10
|
Claff T, Schlegel JG, Voss JH, Vaaßen VJ, Weiße RH, Cheng RKY, Markovic-Mueller S, Bucher D, Sträter N, Müller CE. Crystal structure of adenosine A 2A receptor in complex with clinical candidate Etrumadenant reveals unprecedented antagonist interaction. Commun Chem 2023; 6:106. [PMID: 37264098 DOI: 10.1038/s42004-023-00894-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Accepted: 05/04/2023] [Indexed: 06/03/2023] Open
Abstract
The Gs protein-coupled adenosine A2A receptor (A2AAR) represents an emerging drug target for cancer immunotherapy. The clinical candidate Etrumadenant was developed as an A2AAR antagonist with ancillary blockade of the A2BAR subtype. It constitutes a unique chemotype featuring a poly-substituted 2-amino-4-phenyl-6-triazolylpyrimidine core structure. Herein, we report two crystal structures of the A2AAR in complex with Etrumadenant, obtained with differently thermostabilized A2AAR constructs. This led to the discovery of an unprecedented interaction, a hydrogen bond of T883.36 with the cyano group of Etrumadenant. T883.36 is mutated in most A2AAR constructs used for crystallization, which has prevented the discovery of its interactions. In-vitro characterization of Etrumadenant indicated low selectivity versus the A1AR subtype, which can be rationalized by the structural data. These results will facilitate the future design of AR antagonists with desired selectivity. Moreover, they highlight the advantages of the employed A2AAR crystallization construct that is devoid of ligand binding site mutations.
Collapse
Affiliation(s)
- Tobias Claff
- PharmaCenter Bonn & Pharmaceutical Institute, Department of Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53113, Bonn, Germany.
| | - Jonathan G Schlegel
- PharmaCenter Bonn & Pharmaceutical Institute, Department of Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53113, Bonn, Germany
| | - Jan H Voss
- PharmaCenter Bonn & Pharmaceutical Institute, Department of Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53113, Bonn, Germany
| | - Victoria J Vaaßen
- PharmaCenter Bonn & Pharmaceutical Institute, Department of Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53113, Bonn, Germany
| | - Renato H Weiße
- Institute of Bioanalytical Chemistry, Center for Biotechnology and Biomedicine, University of Leipzig, Deutscher Platz 5, 04103, Leipzig, Germany
| | | | | | - Denis Bucher
- leadXpro AG, PARK InnovAARE, 5234, Villigen, Switzerland
| | - Norbert Sträter
- Institute of Bioanalytical Chemistry, Center for Biotechnology and Biomedicine, University of Leipzig, Deutscher Platz 5, 04103, Leipzig, Germany
| | - Christa E Müller
- PharmaCenter Bonn & Pharmaceutical Institute, Department of Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53113, Bonn, Germany.
| |
Collapse
|
11
|
Kaipa JM, Krasnoselska G, Owens RJ, van den Heuvel J. Screening of Membrane Protein Production by Comparison of Transient Expression in Insect and Mammalian Cells. Biomolecules 2023; 13:biom13050817. [PMID: 37238687 DOI: 10.3390/biom13050817] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 05/04/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
Membrane proteins are difficult biomolecules to express and purify. In this paper, we compare the small-scale production of six selected eukaryotic integral membrane proteins in insect and mammalian cell expression systems using different techniques for gene delivery. The target proteins were C terminally fused to the green fluorescent marker protein GFP to enable sensitive monitoring. We show that the choice of expression systems makes a considerable difference to the yield and quality of the six selected membrane proteins. Virus-free transient gene expression (TGE) in insect High Five cells combined with solubilization in dodecylmaltoside plus cholesteryl hemisuccinate generated the most homogeneous samples for all six targets. Further, the affinity purification of the solubilized proteins using the Twin-Strep® tag improved protein quality in terms of yield and homogeneity compared to His-tag purification. TGE in High Five insect cells offers a fast and economically attractive alternative to the established methods that require either baculovirus construction and the infection of the insect cells or relatively expensive transient gene expression in mammalian cells for the production of integral membrane proteins.
Collapse
Affiliation(s)
| | - Ganna Krasnoselska
- Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3B, 18.5, 42, 2200 Copenhagen, Denmark
| | - Raymond J Owens
- Structural Biology Division, Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
- Rosalind Franklin Institute, Harwell Campus, Didcot OX11 0QX, UK
| | - Joop van den Heuvel
- Helmholtz Center for Infection Research, Department of Structure and Function of Proteins, Inhoffenstrasse 7, 38124 Braunschweig, Germany
| |
Collapse
|
12
|
Hok L, Vianello R. Selective Deuteration Improves the Affinity of Adenosine A 2A Receptor Ligands: A Computational Case Study with Istradefylline and Caffeine. J Chem Inf Model 2023; 63:3138-3149. [PMID: 37155356 DOI: 10.1021/acs.jcim.3c00424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
We used a range of computational techniques to assess the effect of selective C-H deuteration on the antagonist istradefylline affinity for the adenosine A2A receptor, which was discussed relative to its structural analogue caffeine, a well-known and likely the most widely used stimulant. The obtained results revealed that smaller caffeine shows high receptor flexibility and exchanges between two distinct poses, which agrees with crystallographic data. In contrast, the additional C8-trans-styryl fragment in istradefylline locks the ligand within a uniform binding pose, while contributing to the affinity through the C-H···π and π···π contacts with surface residues, which, together with its much lower hydration prior to binding, enhances the affinity over caffeine. In addition, the aromatic C8-unit shows a higher deuteration sensitivity over the xanthine part, so when both of its methoxy groups are d6-deuterated, the affinity improvement is -0.4 kcal mol-1, which surpasses the overall affinity gain of -0.3 kcal mol-1 in the perdeuterated d9-caffeine. Yet, the latter predicts around 1.7-fold potency increase, being relevant for its pharmaceutical implementations, and also those within the coffee and energy drink production industries. Still, the full potential of our strategy is achieved in polydeuterated d19-istradefylline, whose A2A affinity improves by -0.6 kcal mol-1, signifying a 2.8-fold potency increase that strongly promotes it as a potential synthetic target. This knowledge supports deuterium application in drug design, and while the literature already reports about over 20 deuterated drugs currently in the clinical development, it is easily foreseen that more examples will hit the market in the years to come. With this in mind, we propose that the devised computational methodology, involving the ONIOM division of the QM region for the ligand and the MM region for its environment, with an implicit quantization of nuclear motions relevant for the H/D exchange, allows fast and efficient estimates of the binding isotope effects in any biological system.
Collapse
Affiliation(s)
- Lucija Hok
- Laboratory for the Computational Design and Synthesis of Functional Materials, Division of Organic Chemistry and Biochemistry, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| | - Robert Vianello
- Laboratory for the Computational Design and Synthesis of Functional Materials, Division of Organic Chemistry and Biochemistry, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| |
Collapse
|
13
|
Matricon P, Nguyen AT, Vo DD, Baltos JA, Jaiteh M, Luttens A, Kampen S, Christopoulos A, Kihlberg J, May LT, Carlsson J. Structure-based virtual screening discovers potent and selective adenosine A 1 receptor antagonists. Eur J Med Chem 2023; 257:115419. [PMID: 37301076 DOI: 10.1016/j.ejmech.2023.115419] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 04/24/2023] [Accepted: 04/25/2023] [Indexed: 06/12/2023]
Abstract
Development of subtype-selective leads is essential in drug discovery campaigns targeting G protein-coupled receptors (GPCRs). Herein, a structure-based virtual screening approach to rationally design subtype-selective ligands was applied to the A1 and A2A adenosine receptors (A1R and A2AR). Crystal structures of these closely related subtypes revealed a non-conserved subpocket in the binding sites that could be exploited to identify A1R selective ligands. A library of 4.6 million compounds was screened computationally against both receptors using molecular docking and 20 A1R selective ligands were predicted. Of these, seven antagonized the A1R with micromolar activities and several compounds displayed slight selectivity for this subtype. Twenty-seven analogs of two discovered scaffolds were designed, resulting in antagonists with nanomolar potency and up to 76-fold A1R-selectivity. Our results show the potential of structure-based virtual screening to guide discovery and optimization of subtype-selective ligands, which could facilitate the development of safer drugs.
Collapse
Affiliation(s)
- Pierre Matricon
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, SE-751 24, Uppsala, Sweden
| | - Anh Tn Nguyen
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia
| | - Duc Duy Vo
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, SE-751 24, Uppsala, Sweden
| | - Jo-Anne Baltos
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia
| | - Mariama Jaiteh
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, SE-751 24, Uppsala, Sweden
| | - Andreas Luttens
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, SE-751 24, Uppsala, Sweden
| | - Stefanie Kampen
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, SE-751 24, Uppsala, Sweden
| | - Arthur Christopoulos
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia
| | - Jan Kihlberg
- Department of Chemistry - BMC, Uppsala University, SE-751 23, Uppsala, Sweden
| | - Lauren Therese May
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia.
| | - Jens Carlsson
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, SE-751 24, Uppsala, Sweden.
| |
Collapse
|
14
|
Xu G, Zhang S, Zheng L, Hu Z, Cheng L, Chen L, Li J, Shi Z. In silico identification of A1 agonists and A2a inhibitors in pain based on molecular docking strategies and dynamics simulations. Purinergic Signal 2023; 19:87-97. [PMID: 34677752 PMCID: PMC9984648 DOI: 10.1007/s11302-021-09808-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 07/05/2021] [Indexed: 12/19/2022] Open
Abstract
Most recently, the adenosine is considered as one of the most promising targets for treating pain, with few side effects. It exists in the central nervous system, and plays a key role in nociceptive afferent pathway. It is reported that the A1 receptor (A1R) could inhibit Ca2+ channels to reduce the pain like analgesic mechanism of morphine. And, A2a receptor (A2aR) was reported to enhance the accumulation of AMP (cAMP) and released peptides from sensory neurons, resulting in constitutive activation of pain. Much evidence showed that A1R and A2aR could be served as the interesting targets for the treatment of pain. Herein, virtual screening was utilized to identify the small molecule compounds towards A1R and A2aR, and top six molecules were considered as candidates via amber scores. The molecular dynamic (MD) simulations and molecular mechanics/generalized born surface area (MM/GBSA) were employed to further analyze the affinity and binding stability of the six molecules towards A1R and A2aR. Moreover, energy decomposition analysis showed significant residues in A1R and A2aR, including His1383, Phe1276, and Glu1277. It provided basics for discovery of novel agonists and antagonists. Finally, the agonists of A1R (ZINC19943625, ZINC13555217, and ZINC04698406) and inhibitors of A2aR (ZINC19370372, ZINC20176051, and ZINC57263068) were successfully recognized. Taken together, our discovered small molecules may serve as the promising candidate agents for future pain research.
Collapse
Affiliation(s)
- Guangya Xu
- Clinical Genetics Laboratory, Clinical Medical College & Affiliated Hospital & College of Basic Medicine & College of Food and Biological Engineering, Chengdu University, Chengdu, 610081, China
| | - Shutao Zhang
- Clinical Genetics Laboratory, Clinical Medical College & Affiliated Hospital & College of Basic Medicine & College of Food and Biological Engineering, Chengdu University, Chengdu, 610081, China
| | - Lulu Zheng
- Clinical Genetics Laboratory, Clinical Medical College & Affiliated Hospital & College of Basic Medicine & College of Food and Biological Engineering, Chengdu University, Chengdu, 610081, China
| | - Zhongjiao Hu
- Clinical Genetics Laboratory, Clinical Medical College & Affiliated Hospital & College of Basic Medicine & College of Food and Biological Engineering, Chengdu University, Chengdu, 610081, China.,School of Pharmacy, Zunyi Medical University, Zunyi, 563000, China
| | - Lijia Cheng
- Clinical Genetics Laboratory, Clinical Medical College & Affiliated Hospital & College of Basic Medicine & College of Food and Biological Engineering, Chengdu University, Chengdu, 610081, China
| | - Lvlin Chen
- Clinical Genetics Laboratory, Clinical Medical College & Affiliated Hospital & College of Basic Medicine & College of Food and Biological Engineering, Chengdu University, Chengdu, 610081, China
| | - Jun Li
- Clinical Genetics Laboratory, Clinical Medical College & Affiliated Hospital & College of Basic Medicine & College of Food and Biological Engineering, Chengdu University, Chengdu, 610081, China. .,Sichuan Wuyan Biotech Co. Ltd Company, Chengdu, 610041, China.
| | - Zheng Shi
- Clinical Genetics Laboratory, Clinical Medical College & Affiliated Hospital & College of Basic Medicine & College of Food and Biological Engineering, Chengdu University, Chengdu, 610081, China. .,School of Pharmacy, Zunyi Medical University, Zunyi, 563000, China.
| |
Collapse
|
15
|
Chen Y, Zhang J, Weng Y, Xu Y, Lu W, Liu W, Liu M, Hua T, Song G. Cryo-EM structure of the human adenosine A 2B receptor-G s signaling complex. SCIENCE ADVANCES 2022; 8:eadd3709. [PMID: 36563137 PMCID: PMC9788782 DOI: 10.1126/sciadv.add3709] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 11/22/2022] [Indexed: 06/03/2023]
Abstract
The human adenosine A2B receptor (A2BR) is a class A G protein-coupled receptor that is involved in several major physiological and pathological processes throughout the body. A2BR recognizes its ligands adenosine and NECA with relatively low affinity, but the detailed mechanism for its ligand recognition and signaling is still elusive. Here, we present two structures determined by cryo-electron microscopy of A2BR bound to its agonists NECA and BAY60-6583, each coupled to an engineered Gs protein. The structures reveal conserved orthosteric binding pockets with subtle differences, whereas the selectivity or specificity can mainly be attributed to regions extended from the orthosteric pocket. We also found that BAY60-6583 occupies a secondary pocket, where residues V2506.51 and N2737.36 were two key determinants for its selectivity against A2BR. This study offers a better understanding of ligand selectivity for the adenosine receptor family and provides a structural template for further development of A2BR ligands for related diseases.
Collapse
Affiliation(s)
- Ying Chen
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Jinyi Zhang
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yuan Weng
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Yueming Xu
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Weiqiang Lu
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Wei Liu
- Cancer Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Mingyao Liu
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Tian Hua
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Gaojie Song
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| |
Collapse
|
16
|
Ferré G, Anazia K, Silva LO, Thakur N, Ray AP, Eddy MT. Global insights into the fine tuning of human A 2AAR conformational dynamics in a ternary complex with an engineered G protein viewed by NMR. Cell Rep 2022; 41:111844. [PMID: 36543140 PMCID: PMC9832913 DOI: 10.1016/j.celrep.2022.111844] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 11/03/2022] [Accepted: 11/24/2022] [Indexed: 12/24/2022] Open
Abstract
G protein-coupled receptor (GPCR) conformational plasticity enables formation of ternary signaling complexes with intracellular proteins in response to binding extracellular ligands. We investigate the dynamic process of GPCR complex formation in solution with the human A2A adenosine receptor (A2AAR) and an engineered Gs protein, mini-Gs. 2D nuclear magnetic resonance (NMR) data with uniform stable isotope-labeled A2AAR enabled a global comparison of A2AAR conformations between complexes with an agonist and mini-Gs and with an agonist alone. The two conformations are similar and show subtle differences at the receptor intracellular surface, supporting a model whereby agonist binding alone is sufficient to populate a conformation resembling the active state. However, an A2AAR "hot spot" connecting the extracellular ligand-binding pocket to the intracellular surface is observed to be highly dynamic in the ternary complex, suggesting a mechanism for allosteric connection between the bound G protein and the drug-binding pocket involving structural plasticity of the "toggle switch" tryptophan.
Collapse
Affiliation(s)
- Guillaume Ferré
- Department of Chemistry, University of Florida, Gainesville, FL 32611, USA
| | - Kara Anazia
- Department of Chemistry, University of Florida, Gainesville, FL 32611, USA
| | - Larissa O Silva
- Department of Chemistry, University of Florida, Gainesville, FL 32611, USA
| | - Naveen Thakur
- Department of Chemistry, University of Florida, Gainesville, FL 32611, USA
| | - Arka P Ray
- Department of Chemistry, University of Florida, Gainesville, FL 32611, USA
| | - Matthew T Eddy
- Department of Chemistry, University of Florida, Gainesville, FL 32611, USA.
| |
Collapse
|
17
|
Preti B, Suchankova A, Deganutti G, Leuenberger M, Barkan K, Manulak I, Huang X, Carvalho S, Ladds G, Lochner M. Discovery and Structure-Activity Relationship Studies of Novel Adenosine A 1 Receptor-Selective Agonists. J Med Chem 2022; 65:14864-14890. [PMID: 36270633 PMCID: PMC9661479 DOI: 10.1021/acs.jmedchem.2c01414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Indexed: 11/29/2022]
Abstract
A series of benzyloxy and phenoxy derivatives of the adenosine receptor agonists N6-cyclopentyl adenosine (CPA) and N6-cyclopentyl 5'-N-ethylcarboxamidoadenosine (CP-NECA) were synthesized, and their potency and selectivity were assessed. We observed that the most potent were the compounds with a halogen in the meta position on the aromatic ring of the benzyloxy- or phenoxycyclopentyl substituent. In general, the NECA-based compounds displayed greater A1R selectivity than the adenosine-based compounds, with N6-2-(3-bromobenzyloxy)cyclopentyl-NECA and N6-2-(3-methoxyphenoxy)cyclopentyl-NECA showing ∼1500-fold improved A1R selectivity compared to NECA. In addition, we quantified the compounds' affinity and kinetics of binding at both human and rat A1R using a NanoBRET binding assay and found that the halogen substituent in the benzyloxy- or phenoxycyclopentyl moiety seems to confer high affinity for the A1R. Molecular modeling studies suggested a hydrophobic subpocket as contributing to the A1R selectivity displayed. We believe that the identified selective potent A1R agonists are valuable tool compounds for adenosine receptor research.
Collapse
Affiliation(s)
- Barbara Preti
- Institute
of Biochemistry and Molecular Medicine, University of Bern, Bühlstrasse 28, 3012Bern, Switzerland
| | - Anna Suchankova
- Department
of Pharmacology, University of Cambridge, Tennis Court Road, CambridgeCB2 1PD, U.K.
| | - Giuseppe Deganutti
- Centre
for Sport, Exercise and Life Sciences, Faculty of Health and Life
Sciences, Coventry University, CoventryCV1 5FB, U.K.
| | - Michele Leuenberger
- Institute
of Biochemistry and Molecular Medicine, University of Bern, Bühlstrasse 28, 3012Bern, Switzerland
| | - Kerry Barkan
- Department
of Pharmacology, University of Cambridge, Tennis Court Road, CambridgeCB2 1PD, U.K.
| | - Iga Manulak
- Department
of Pharmacology, University of Cambridge, Tennis Court Road, CambridgeCB2 1PD, U.K.
| | - Xianglin Huang
- Department
of Pharmacology, University of Cambridge, Tennis Court Road, CambridgeCB2 1PD, U.K.
| | - Sabrina Carvalho
- Department
of Pharmacology, University of Cambridge, Tennis Court Road, CambridgeCB2 1PD, U.K.
| | - Graham Ladds
- Department
of Pharmacology, University of Cambridge, Tennis Court Road, CambridgeCB2 1PD, U.K.
| | - Martin Lochner
- Institute
of Biochemistry and Molecular Medicine, University of Bern, Bühlstrasse 28, 3012Bern, Switzerland
| |
Collapse
|
18
|
Jacobson KA, Gao ZG, Matricon P, Eddy MT, Carlsson J. Adenosine A 2A receptor antagonists: from caffeine to selective non-xanthines. Br J Pharmacol 2022; 179:3496-3511. [PMID: 32424811 PMCID: PMC9251831 DOI: 10.1111/bph.15103] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 04/30/2020] [Accepted: 05/03/2020] [Indexed: 12/12/2022] Open
Abstract
A long evolution of knowledge of the psychostimulant caffeine led in the 1960s to another purine natural product, adenosine and its A2A receptor. Adenosine is a short-lived autocrine/paracrine mediator that acts pharmacologically at four different adenosine receptors in a manner opposite to the pan-antagonist caffeine and serves as an endogenous allostatic regulator. Although detrimental in the developing brain, caffeine appears to be cerebroprotective in aging. Moderate caffeine consumption in adults, except in pregnancy, may also provide benefit in pain, diabetes, and kidney and liver disorders. Inhibition of A2A receptors is one of caffeine's principal effects and we now understand this interaction at the atomic level. The A2A receptor has become a prototypical example of utilizing high-resolution structures of GPCRs for the rational design of chemically diverse drug molecules. The previous focus on discovery of selective A2A receptor antagonists for neurodegenerative diseases has expanded to include immunotherapy for cancer, and clinical trials have ensued. LINKED ARTICLES: This article is part of a themed issue on Structure Guided Pharmacology of Membrane Proteins (BJP 75th Anniversary). To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v179.14/issuetoc.
Collapse
Affiliation(s)
- Kenneth A. Jacobson
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Zhan-Guo Gao
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Pierre Matricon
- Department of Cell and Molecular Biology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Matthew T. Eddy
- Department of Chemistry, University of Florida, Gainesville, FL, USA
| | - Jens Carlsson
- Department of Cell and Molecular Biology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| |
Collapse
|
19
|
Atif M, Alsrhani A, Naz F, Ullah MI, Alameen AAM, Imran M, Ejaz H. Adenosine A 2A receptor as a potential target for improving cancer immunotherapy. Mol Biol Rep 2022; 49:10677-10687. [PMID: 35752699 DOI: 10.1007/s11033-022-07685-7] [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: 04/26/2022] [Accepted: 06/07/2022] [Indexed: 11/29/2022]
Abstract
The adenosine nucleoside performs a wide range of actions on various human tissues by activating four cell surface receptors. Adenosine A2A receptors (A2ARs) are widely expressed in the striatum, olfactory bulb, platelets, leukocytes, spleen, and thymus. They promote vasodilatation, platelet antiaggregatory effect, protection from ischemic damage, and regulation of sensorimotor neurons in basal ganglia. Adenosine signaling plays a vital part in modulating in vivo pathophysiological responses. A2ARs are potent negative regulators of the antitumor and proinflammatory actions of activated T cells. This axis offers several therapeutic targets, the most important of which are A2ARs, HIF-1α, and CD39/CD73. Downregulation of this axis increases the effectiveness of modern immunotherapeutic approaches against cancer, such as αCTLA-4/αPD-1. These discoveries have led to a promising novel role of antagonists of A2AR in blocking angiogenesis in immunotherapy of cancer. A small molecule, AZD4635, strongly inhibits A2AR, lowering cancer volume and increasing anticancer immunity. Deletion of A2AR with CRISPR/Cas9 in both human and murine CAR T cells produces a substantial increase in the efficiency of these cells. This review asserts that inhibition of the adenosinergic pathway can boost antitumor immunity, and this axis should be a target for future immunotherapeutic strategies.
Collapse
Affiliation(s)
- Muhammad Atif
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Al Jouf, 72388, Saudi Arabia
| | - Abdullah Alsrhani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Al Jouf, 72388, Saudi Arabia
| | - Farrah Naz
- Department of Pathology, Institute of Public Health, Lahore, Pakistan
| | - Muhammad Ikram Ullah
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Al Jouf, 72388, Saudi Arabia
| | - Ayman Ali Mohammed Alameen
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Al Jouf, 72388, Saudi Arabia
| | - Muhammad Imran
- Department of Food Science and Technology, University of Narowal, Narowal, Pakistan
| | - Hasan Ejaz
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Al Jouf, 72388, Saudi Arabia.
| |
Collapse
|
20
|
Bożyk A, Wojas-Krawczyk K, Krawczyk P, Milanowski J. Tumor Microenvironment—A Short Review of Cellular and Interaction Diversity. BIOLOGY 2022; 11:biology11060929. [PMID: 35741450 PMCID: PMC9220289 DOI: 10.3390/biology11060929] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 06/10/2022] [Accepted: 06/14/2022] [Indexed: 12/26/2022]
Abstract
The tumor microenvironment is a complex network of various interactions between immune cells and non-cellular components such as the extracellular matrix, exosomes and interleukins. Moreover, tumor heterogeneity and its constant modification may alter the immunophenotype and become responsible for its resistance regarding the therapies applied However, it should be remembered that in a strongly immunosuppressive neoplastic microenvironment, the immune system cells undergo reprogramming and most often cease to fulfill their original function. Therefore, understanding what happens within the tumor microenvironment, and which mechanisms are responsible for tumor development and progression should let us know how cancer could protect itself against the immune system. The presented review summarizes the latest information on the interactions between the tumor microenvironment and the cellular and non-cellular components, as well as their impact on cancer development, progression and immune system exhaustion.
Collapse
|
21
|
Claff T, Klapschinski TA, Tiruttani Subhramanyam UK, Vaaßen VJ, Schlegel JG, Vielmuth C, Voß JH, Labahn J, Müller CE. Single Stabilizing Point Mutation Enables High-Resolution Co-Crystal Structures of the Adenosine A 2A Receptor with Preladenant Conjugates. Angew Chem Int Ed Engl 2022; 61:e202115545. [PMID: 35174942 PMCID: PMC9310709 DOI: 10.1002/anie.202115545] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Indexed: 01/08/2023]
Abstract
The G protein-coupled adenosine A2A receptor (A2A AR) is an important new (potential) drug target in immuno-oncology, and for neurodegenerative diseases. Preladenant and its derivatives belong to the most potent A2A AR antagonists displaying exceptional selectivity. While crystal structures of the human A2A AR have been solved, mostly using the A2A -StaR2 protein that bears 9 point mutations, co-crystallization with Preladenant derivatives has so far been elusive. We developed a new A2A AR construct harboring a single point mutation (S913.39 K) which renders it extremely thermostable. This allowed the co-crystallization of two novel Preladenant derivatives, the polyethylene glycol-conjugated (PEGylated) PSB-2113, and the fluorophore-labeled PSB-2115. The obtained crystal structures (2.25 Å and 2.6 Å resolution) provide explanations for the high potency and selectivity of Preladenant derivatives. They represent the first crystal structures of a GPCR in complex with PEG- and fluorophore-conjugated ligands. The applied strategy is predicted to be applicable to further class A GPCRs.
Collapse
Affiliation(s)
- Tobias Claff
- Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53121, Bonn, Germany
| | - Tim A Klapschinski
- Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53121, Bonn, Germany
| | - Udaya K Tiruttani Subhramanyam
- Centre for Structural Systems Biology (CSSB), Notkestraße 85, 22607, Hamburg, Germany.,Research Centre Jülich, Institute of Complex Systems (IBI-7), Wilhelm-Johnen-Straße, 52425, Jülich, Germany
| | - Victoria J Vaaßen
- Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53121, Bonn, Germany
| | - Jonathan G Schlegel
- Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53121, Bonn, Germany
| | - Christin Vielmuth
- Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53121, Bonn, Germany
| | - Jan H Voß
- Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53121, Bonn, Germany
| | - Jörg Labahn
- Centre for Structural Systems Biology (CSSB), Notkestraße 85, 22607, Hamburg, Germany.,Research Centre Jülich, Institute of Complex Systems (IBI-7), Wilhelm-Johnen-Straße, 52425, Jülich, Germany
| | - Christa E Müller
- Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53121, Bonn, Germany
| |
Collapse
|
22
|
Zhu WM, Neuhaus A, Beard DJ, Sutherland BA, DeLuca GC. Neurovascular coupling mechanisms in health and neurovascular uncoupling in Alzheimer's disease. Brain 2022; 145:2276-2292. [PMID: 35551356 PMCID: PMC9337814 DOI: 10.1093/brain/awac174] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 05/05/2022] [Accepted: 05/07/2022] [Indexed: 11/25/2022] Open
Abstract
To match the metabolic demands of the brain, mechanisms have evolved to couple neuronal activity to vasodilation, thus increasing local cerebral blood flow and delivery of oxygen and glucose to active neurons. Rather than relying on metabolic feedback signals such as the consumption of oxygen or glucose, the main signalling pathways rely on the release of vasoactive molecules by neurons and astrocytes, which act on contractile cells. Vascular smooth muscle cells and pericytes are the contractile cells associated with arterioles and capillaries, respectively, which relax and induce vasodilation. Much progress has been made in understanding the complex signalling pathways of neurovascular coupling, but issues such as the contributions of capillary pericytes and astrocyte calcium signal remain contentious. Study of neurovascular coupling mechanisms is especially important as cerebral blood flow dysregulation is a prominent feature of Alzheimer’s disease. In this article we will discuss developments and controversies in the understanding of neurovascular coupling and finish by discussing current knowledge concerning neurovascular uncoupling in Alzheimer’s disease.
Collapse
Affiliation(s)
- Winston M Zhu
- Oxford Medical School, University of Oxford, Oxford, UK
| | - Ain Neuhaus
- Acute Stroke Programme, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Daniel J Beard
- Acute Stroke Programme, Radcliffe Department of Medicine, University of Oxford, Oxford, UK.,School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia
| | - Brad A Sutherland
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Australia
| | - Gabriele C DeLuca
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| |
Collapse
|
23
|
Suresh RR, Gao ZG, Salmaso V, Chen E, Campbell RG, Poe RB, Liston TE, Jacobson KA. Selective A 3 Adenosine Receptor Antagonist Radioligand for Human and Rodent Species. ACS Med Chem Lett 2022; 13:623-631. [PMID: 35450351 PMCID: PMC9014498 DOI: 10.1021/acsmedchemlett.1c00685] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 02/24/2022] [Indexed: 11/28/2022] Open
Abstract
The A3 adenosine receptor (A3AR) is a target for pain, ischemia, and inflammatory disease therapy. Among the ligand tools available are selective agonists and antagonists, including radioligands, but most high-affinity non-nucleoside antagonists are limited in selectivity to primate species. We have explored the structure-activity relationship of a previously reported A3AR antagonist DPTN 9 (N-[4-(3,5-dimethylphenyl)-5-(4-pyridyl)-1,3-thiazol-2-yl]nicotinamide) for radiolabeling, including 3-halo derivatives (3-iodo, MRS7907), and characterized 9 as a high -affinity radioligand [3H]MRS7799. A3AR K d values were (nM): 0.55 (human), 3.74 (mouse), and 2.80 (rat). An extended methyl acrylate (MRS8074, 19) maintained higher affinity (18.9 nM) than a 3-((5-chlorothiophen-2-yl)ethynyl) derivative 20. Compound 9 had an excellent brain distribution in rats (brain/plasma ratio ∼1). Receptor docking predicted its orthosteric site binding by engaging residues that were previously found to be essential for AR binding. Thus the new radioligand promises to be a useful species-general antagonist tracer for receptor characterization and drug discovery.
Collapse
Affiliation(s)
- R. Rama Suresh
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland 20892, United States
| | - Zhan-Guo Gao
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland 20892, United States
| | - Veronica Salmaso
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland 20892, United States
| | - Eric Chen
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland 20892, United States
| | - Ryan G. Campbell
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland 20892, United States
| | - Russell B. Poe
- Astrocyte Pharmaceuticals, Cambridge, Massachusetts 02142, United States
| | - Theodore E. Liston
- Astrocyte Pharmaceuticals, Cambridge, Massachusetts 02142, United States
| | - Kenneth A. Jacobson
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland 20892, United States
| |
Collapse
|
24
|
A 2A Adenosine Receptor Antagonists: Are Triazolotriazine and Purine Scaffolds Interchangeable? Molecules 2022; 27:molecules27082386. [PMID: 35458588 PMCID: PMC9032385 DOI: 10.3390/molecules27082386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 04/02/2022] [Accepted: 04/04/2022] [Indexed: 12/10/2022] Open
Abstract
The A2A adenosine receptor (A2AAR) is one of the four subtypes activated by nucleoside adenosine, and the molecules able to selectively counteract its action are attractive tools for neurodegenerative disorders. In order to find novel A2AAR ligands, two series of compounds based on purine and triazolotriazine scaffolds were synthesized and tested at ARs. Compound 13 was also tested in an in vitro model of neuroinflammation. Some compounds were found to possess high affinity for A2AAR, and it was observed that compound 13 exerted anti-inflammatory properties in microglial cells. Molecular modeling studies results were in good agreement with the binding affinity data and underlined that triazolotriazine and purine scaffolds are interchangeable only when 5- and 2-positions of the triazolotriazine moiety (corresponding to the purine 2- and 8-positions) are substituted.
Collapse
|
25
|
Mori A, Chen JF, Uchida S, Durlach C, King SM, Jenner P. The Pharmacological Potential of Adenosine A 2A Receptor Antagonists for Treating Parkinson's Disease. Molecules 2022; 27:2366. [PMID: 35408767 PMCID: PMC9000505 DOI: 10.3390/molecules27072366] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/30/2022] [Accepted: 03/31/2022] [Indexed: 02/07/2023] Open
Abstract
The adenosine A2A receptor subtype is recognized as a non-dopaminergic pharmacological target for the treatment of neurodegenerative disorders, notably Parkinson's disease (PD). The selective A2A receptor antagonist istradefylline is approved in the US and Japan as an adjunctive treatment to levodopa/decarboxylase inhibitors in adults with PD experiencing OFF episodes or a wearing-off phenomenon; however, the full potential of this drug class remains to be explored. In this article, we review the pharmacology of adenosine A2A receptor antagonists from the perspective of the treatment of both motor and non-motor symptoms of PD and their potential for disease modification.
Collapse
Affiliation(s)
- Akihisa Mori
- Kyowa Kirin Co., Ltd., Tokyo 100-0004, Japan; (A.M.); (S.U.)
| | - Jiang-Fan Chen
- Molecular Neuropharmacology Laboratory, Wenzhou Medical University, Wenzhou 325015, China;
| | - Shinichi Uchida
- Kyowa Kirin Co., Ltd., Tokyo 100-0004, Japan; (A.M.); (S.U.)
| | | | | | - Peter Jenner
- Institute of Pharmaceutical Science, Kings College London, London SE1 9NH, UK
| |
Collapse
|
26
|
Bedart C, Renault N, Chavatte P, Porcherie A, Lachgar A, Capron M, Farce A. SINAPs: A Software Tool for Analysis and Visualization of Interaction Networks of Molecular Dynamics Simulations. J Chem Inf Model 2022; 62:1425-1436. [PMID: 35239339 PMCID: PMC8966674 DOI: 10.1021/acs.jcim.1c00854] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
As long as the structural study of molecular mechanisms requires multiple molecular dynamics reflecting contrasted bioactive states, the subsequent analysis of molecular interaction networks remains a bottleneck to be fairly treated and requires a user-friendly 3D view of key interactions. Structural Interaction Network Analysis Protocols (SINAPs) is a proprietary python tool developed to (i) quickly solve key interactions able to distinguish two protein states, either from two sets of molecular dynamics simulations or from two crystallographic structures, and (ii) render a user-friendly 3D view of these key interactions through a plugin of UCSF Chimera, one of the most popular open-source viewing software for biomolecular systems. Through two case studies, glucose transporter-1 (GLUT-1) and A2A adenosine receptor (A2AR), SINAPs easily pinpointed key interactions observed experimentally and relevant for their bioactivities. This very effective tool was thus applied to identify the amino acids involved in the molecular enzymatic mechanisms ruling the activation of an immunomodulator drug candidate, P28 glutathione-S-transferase (P28GST). SINAPs is freely available at https://github.com/ParImmune/SINAPs.
Collapse
Affiliation(s)
- Corentin Bedart
- Univ.
Lille, Inserm, CHU Lille, U1286 - Infinite - Institute for Translational
Research in Inflammation, F-59000 Lille, France,Par’Immune,
Bio-incubateur Eurasanté, 70 rue du Dr. Yersin, 59120 Loos-Lez-Lille, France,
| | - Nicolas Renault
- Univ.
Lille, Inserm, CHU Lille, U1286 - Infinite - Institute for Translational
Research in Inflammation, F-59000 Lille, France
| | - Philippe Chavatte
- Univ.
Lille, Inserm, CHU Lille, U1286 - Infinite - Institute for Translational
Research in Inflammation, F-59000 Lille, France
| | - Adeline Porcherie
- Par’Immune,
Bio-incubateur Eurasanté, 70 rue du Dr. Yersin, 59120 Loos-Lez-Lille, France
| | - Abderrahim Lachgar
- Par’Immune,
Bio-incubateur Eurasanté, 70 rue du Dr. Yersin, 59120 Loos-Lez-Lille, France
| | - Monique Capron
- Univ.
Lille, Inserm, CHU Lille, U1286 - Infinite - Institute for Translational
Research in Inflammation, F-59000 Lille, France,Par’Immune,
Bio-incubateur Eurasanté, 70 rue du Dr. Yersin, 59120 Loos-Lez-Lille, France
| | - Amaury Farce
- Univ.
Lille, Inserm, CHU Lille, U1286 - Infinite - Institute for Translational
Research in Inflammation, F-59000 Lille, France,
| |
Collapse
|
27
|
Claff T, Klapschinski TA, Tiruttani Subhramanyam UK, Vaaßen VJ, Schlegel JG, Vielmuth C, Voß JH, Labahn J, Müller CE. Eine einzige stabilisierende Punktmutation ermöglicht hochaufgelöste Co‐Kristallstrukturen des Adenosin‐A
2A
‐Rezeptors mit Preladenant‐Konjugaten. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202115545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Tobias Claff
- Pharmaceutical Institute Pharmaceutical & Medicinal Chemistry University of Bonn An der Immenburg 4 53121 Bonn Deutschland
| | - Tim A. Klapschinski
- Pharmaceutical Institute Pharmaceutical & Medicinal Chemistry University of Bonn An der Immenburg 4 53121 Bonn Deutschland
| | - Udaya K. Tiruttani Subhramanyam
- Centre for Structural Systems Biology (CSSB) Notkestraße 85 22607 Hamburg Germany
- Research Centre Jülich Institute of Complex Systems (IBI-7) Wilhelm-Johnen-Straße 52425 Jülich Deutschland
| | - Victoria J. Vaaßen
- Pharmaceutical Institute Pharmaceutical & Medicinal Chemistry University of Bonn An der Immenburg 4 53121 Bonn Deutschland
| | - Jonathan G. Schlegel
- Pharmaceutical Institute Pharmaceutical & Medicinal Chemistry University of Bonn An der Immenburg 4 53121 Bonn Deutschland
| | - Christin Vielmuth
- Pharmaceutical Institute Pharmaceutical & Medicinal Chemistry University of Bonn An der Immenburg 4 53121 Bonn Deutschland
| | - Jan H. Voß
- Pharmaceutical Institute Pharmaceutical & Medicinal Chemistry University of Bonn An der Immenburg 4 53121 Bonn Deutschland
| | - Jörg Labahn
- Centre for Structural Systems Biology (CSSB) Notkestraße 85 22607 Hamburg Germany
- Research Centre Jülich Institute of Complex Systems (IBI-7) Wilhelm-Johnen-Straße 52425 Jülich Deutschland
| | - Christa E. Müller
- Pharmaceutical Institute Pharmaceutical & Medicinal Chemistry University of Bonn An der Immenburg 4 53121 Bonn Deutschland
| |
Collapse
|
28
|
Huang SK, Prosser RS. Dynamics and Mechanistic Underpinnings to Pharmacology of Class A GPCRs - An NMR Perspective. Am J Physiol Cell Physiol 2022; 322:C739-C753. [PMID: 35235425 DOI: 10.1152/ajpcell.00044.2022] [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] [Indexed: 01/14/2023]
Abstract
One-third of current pharmaceuticals target G protein-coupled receptors (GPCRs), the largest receptor superfamily in humans and mediators of diverse physiological processes. This review summarizes the recent progress in GPCR structural dynamics, focusing on class A receptors and insights derived from nuclear magnetic resonance (NMR) and other spectroscopic techniques. We describe the structural aspects of GPCR activation and the various pharmacological models that capture aspects of receptor signaling behaviour. Spectroscopic studies revealed that receptors and their signaling complexes are dynamic allosteric systems that sample multiple functional states under basal conditions. The distribution of states within the conformational ensemble and the kinetics of transitions between states are regulated through the binding of ligands, allosteric modulators, and the membrane environment. This ensemble view of GPCRs provides a mechanistic framework for understanding many of the pharmacological phenomena associated with receptor signaling, such as basal activity, efficacy, and functional bias.
Collapse
Affiliation(s)
- Shuya Kate Huang
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada.,Department of Chemical and Physical Sciences, University of Toronto Mississauga, Mississauga, Ontario, Canada
| | - R Scott Prosser
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada.,Department of Chemical and Physical Sciences, University of Toronto Mississauga, Mississauga, Ontario, Canada.,Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
29
|
Lambertucci C, Marucci G, Catarzi D, Colotta V, Francucci B, Spinaci A, Varano F, Volpini R. A2A Adenosine Receptor Antagonists and their Potential in Neurological Disorders. Curr Med Chem 2022; 29:4780-4795. [PMID: 35184706 DOI: 10.2174/0929867329666220218094501] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 12/11/2021] [Accepted: 12/18/2021] [Indexed: 11/22/2022]
Abstract
Endogenous nucleoside adenosine modulates a number of physiological effects through interaction with P1 purinergic receptors. All of them are G protein coupled receptors and, to date, four subtypes have been characterized and named A1, A2A, A2B, and A3. In recent years adenosine receptors, particularly the A2A subtype, have become attractive targets for the treatment of several neurodegenerative disorders, known to involve neuroinflammation, like Parkinson's and Alzheimer's diseases, multiple sclerosis and neuropsychiatric conditions. In fact, it has been demonstrated that inhibition of A2A adenosine receptors exerts neuroprotective effects counteracting neuroinflammatory processes and astroglial and microglial activation. The A2A adenosine receptor antagonist istradefylline, developed by Kyowa Hakko Kirin Inc., was approved in Japan as adjunctive therapy for the treatment of Parkinson's disease and very recently it was approved also by the US Food and Drug Administration. These findings pave the way for new therapeutic opportunities, so, in this review, a summary of the most relevant and promising A2A adenosine receptor antagonists will be presented along with their preclinical and clinical studies in neuroinflammation related diseases.
Collapse
Affiliation(s)
- Catia Lambertucci
- Medicinal Chemistry Unit, School of Pharmacy, University of Camerino, 62032 Camerino (MC), Italy
| | - Gabriella Marucci
- Medicinal Chemistry Unit, School of Pharmacy, University of Camerino, 62032 Camerino (MC), Italy
| | - Daniela Catarzi
- Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino, Sezione di Farmaceutica e Nutraceutica, Università degli Studi di Firenze, 50019 Sesto Fiorentino (FI), Italy
| | - Vittoria Colotta
- Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino, Sezione di Farmaceutica e Nutraceutica, Università degli Studi di Firenze, 50019 Sesto Fiorentino (FI), Italy
| | - Beatrice Francucci
- Medicinal Chemistry Unit, School of Pharmacy, University of Camerino, 62032 Camerino (MC), Italy
| | - Andrea Spinaci
- Medicinal Chemistry Unit, School of Pharmacy, University of Camerino, 62032 Camerino (MC), Italy
| | - Flavia Varano
- Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino, Sezione di Farmaceutica e Nutraceutica, Università degli Studi di Firenze, 50019 Sesto Fiorentino (FI), Italy
| | - Rosaria Volpini
- Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino, Sezione di Farmaceutica e Nutraceutica, Università degli Studi di Firenze, 50019 Sesto Fiorentino (FI), Italy
| |
Collapse
|
30
|
Mansoor S, Kayık G, Durdagi S, Sensoy O. Mechanistic insight into the impact of a bivalent ligand on the structure and dynamics of a GPCR oligomer. Comput Struct Biotechnol J 2022; 20:925-936. [PMID: 35242285 PMCID: PMC8861583 DOI: 10.1016/j.csbj.2022.01.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 12/25/2021] [Accepted: 01/17/2022] [Indexed: 12/25/2022] Open
Abstract
Development of effective bivalent ligands has become the focus of intensive research toward modulation of G protein-coupled receptor (GPCR) oligomers, particularly in the field of GPCR pharmacology. Experimental studies have shown that they increased binding affinity and signaling potency compared to their monovalent counterparts, yet underlying molecular mechanism remains elusive. To address this, we performed accelerated molecular dynamics simulations on bivalent-ligand bound Adenosine 2A receptor (A2AR) dimer in the context of a modeled tetramer, which consists of A2AR and dopamine 2 receptor (D2R) homodimers and their cognate G proteins. Our results demonstrate that bivalent ligand impacted interactions between pharmacophore groups and ligand binding residues, thus modulating allosteric communication network and water channel formed within the receptor. Moreover, it also strengthens contacts between receptor and G protein, by modulating the volume of ligand binding pocket and intracellular domain of the receptor. Importantly, we showed that impact evoked by the bivalent ligand on A2AR dimer was also transmitted to apo D2R, which is part of the neighboring D2R dimer. To the best of our knowledge, this is the first study that provides a mechanistic insight into the impact of a bivalent ligand on dynamics of a GPCR oligomer. Consequently, this will pave the way for development of effective ligands for modulation of GPCR oligomers and hence treatment of crucial diseases such as Parkinson's disease and cancer.
Collapse
Affiliation(s)
- Samman Mansoor
- School of Engineering and Natural Sciences, Department of Biomedical Engineering and Bioinformatics, Istanbul Medipol University, Istanbul 34810, Turkey
| | - Gülru Kayık
- Computational Biology and Molecular Simulations Laboratory, Department of Biophysics, School of Medicine, Bahcesehir University, Istanbul, Turkey
| | - Serdar Durdagi
- Computational Biology and Molecular Simulations Laboratory, Department of Biophysics, School of Medicine, Bahcesehir University, Istanbul, Turkey
| | - Ozge Sensoy
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciencesand Technologies (SABITA), Istanbul Medipol University, 34810 Istanbul, Turkey
- School of Engineering and Natural Sciences, Department of Computer Engineering, Istanbul Medipol University, Turkey
| |
Collapse
|
31
|
Kotulová J, Hajdúch M, Džubák P. Current Adenosinergic Therapies: What Do Cancer Cells Stand to Gain and Lose? Int J Mol Sci 2021; 22:12569. [PMID: 34830449 PMCID: PMC8617980 DOI: 10.3390/ijms222212569] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 11/18/2021] [Accepted: 11/18/2021] [Indexed: 12/12/2022] Open
Abstract
A key objective in immuno-oncology is to reactivate the dormant immune system and increase tumour immunogenicity. Adenosine is an omnipresent purine that is formed in response to stress stimuli in order to restore physiological balance, mainly via anti-inflammatory, tissue-protective, and anti-nociceptive mechanisms. Adenosine overproduction occurs in all stages of tumorigenesis, from the initial inflammation/local tissue damage to the precancerous niche and the developed tumour, making the adenosinergic pathway an attractive but challenging therapeutic target. Many current efforts in immuno-oncology are focused on restoring immunosurveillance, largely by blocking adenosine-producing enzymes in the tumour microenvironment (TME) and adenosine receptors on immune cells either alone or combined with chemotherapy and/or immunotherapy. However, the effects of adenosinergic immunotherapy are not restricted to immune cells; other cells in the TME including cancer and stromal cells are also affected. Here we summarise recent advancements in the understanding of the tumour adenosinergic system and highlight the impact of current and prospective immunomodulatory therapies on other cell types within the TME, focusing on adenosine receptors in tumour cells. In addition, we evaluate the structure- and context-related limitations of targeting this pathway and highlight avenues that could possibly be exploited in future adenosinergic therapies.
Collapse
Affiliation(s)
| | | | - Petr Džubák
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University Olomouc, 779 00 Olomouc, Czech Republic; (J.K.); (M.H.)
| |
Collapse
|
32
|
Saini A, Patel R, Gaba S, Singh G, Gupta GD, Monga V. Adenosine receptor antagonists: Recent advances and therapeutic perspective. Eur J Med Chem 2021; 227:113907. [PMID: 34695776 DOI: 10.1016/j.ejmech.2021.113907] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 10/05/2021] [Accepted: 10/05/2021] [Indexed: 12/14/2022]
Abstract
Adenosine is an endogenous purine-based nucleoside expressed nearly in all body tissues. It regulates various body functions by activating four G-protein coupled receptors, A1, A2A, A2B, and A3. These receptors are widely acknowledged as drug targets for treating different neurological, metabolic, and inflammatory diseases. Although numerous adenosine receptor inhibitors have been developed worldwide, achieving target selectivity is still a big hurdle in drug development. However, the identification of specific radioligands-based affinity assay, fluorescent ligands, and MS-based ligand assay have contributed to the development of selective and potent adenosine ligands. In recent years various small heterocyclic-based molecules have shown some promising results. Istradefylline has been approved for treating Parkinson's in Japan, while preladenant, tozadenant, CVT-6883, MRS-1523, and many more are under different phases of clinical development. The present review is focused on the quest to develop potent and selective adenosine inhibitors from 2013 to early 2021 by various research groups. The review also highlights their biological activity, selectivity, structure-activity relationship, molecular docking, and mechanistic studies. A special emphsesis on drug designing strategies has been also given the manuscript. The comprehensive compilation of research work carried out in the field will provide inevitable scope for designing and developing novel adenosine inhibitors with improved selectivity and efficacy.
Collapse
Affiliation(s)
- Anjali Saini
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga, 142001, Punjab, India
| | - Rajiv Patel
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga, 142001, Punjab, India
| | - Sobhi Gaba
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga, 142001, Punjab, India
| | - Gurpreet Singh
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga, 142001, Punjab, India.
| | - G D Gupta
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga, 142001, Punjab, India
| | - Vikramdeep Monga
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga, 142001, Punjab, India.
| |
Collapse
|
33
|
Sadgrove NJ. The ‘bald’ phenotype (androgenetic alopecia) is caused by the high glycaemic, high cholesterol and low mineral ‘western diet’. Trends Food Sci Technol 2021. [DOI: 10.1016/j.tifs.2021.06.056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
34
|
Bernardo A, De Nuccio C, Visentin S, Martire A, Minghetti L, Popoli P, Ferrante A. Myelin Defects in Niemann-Pick Type C Disease: Mechanisms and Possible Therapeutic Perspectives. Int J Mol Sci 2021; 22:ijms22168858. [PMID: 34445564 PMCID: PMC8396228 DOI: 10.3390/ijms22168858] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/11/2021] [Accepted: 08/16/2021] [Indexed: 12/25/2022] Open
Abstract
Niemann–Pick type C (NPC) disease is a wide-spectrum clinical condition classified as a neurovisceral disorder affecting mainly the liver and the brain. It is caused by mutations in one of two genes, NPC1 and NPC2, coding for proteins located in the lysosomes. NPC proteins are deputed to transport cholesterol within lysosomes or between late endosome/lysosome systems and other cellular compartments, such as the endoplasmic reticulum and plasma membrane. The first trait of NPC is the accumulation of unesterified cholesterol and other lipids, like sphingosine and glycosphingolipids, in the late endosomal and lysosomal compartments, which causes the blockade of autophagic flux and the impairment of mitochondrial functions. In the brain, the main consequences of NPC are cerebellar neurodegeneration, neuroinflammation, and myelin defects. This review will focus on myelin defects and the pivotal importance of cholesterol for myelination and will offer an overview of the molecular targets and the pharmacological strategies so far proposed, or an object of clinical trials for NPC. Finally, it will summarize recent data on a new and promising pharmacological perspective involving A2A adenosine receptor stimulation in genetic and pharmacological NPC dysmyelination models.
Collapse
Affiliation(s)
- Antonietta Bernardo
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (A.B.); (S.V.); (A.M.); (P.P.)
| | - Chiara De Nuccio
- Research Coordination and Support Service, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (C.D.N.); (L.M.)
| | - Sergio Visentin
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (A.B.); (S.V.); (A.M.); (P.P.)
| | - Alberto Martire
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (A.B.); (S.V.); (A.M.); (P.P.)
| | - Luisa Minghetti
- Research Coordination and Support Service, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (C.D.N.); (L.M.)
| | - Patrizia Popoli
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (A.B.); (S.V.); (A.M.); (P.P.)
| | - Antonella Ferrante
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (A.B.); (S.V.); (A.M.); (P.P.)
- Correspondence: ; Tel.: +39-06-49902050
| |
Collapse
|
35
|
Kampen S, Duy Vo D, Zhang X, Panel N, Yang Y, Jaiteh M, Matricon P, Svenningsson P, Brea J, Loza MI, Kihlberg J, Carlsson J. Structure‐Guided Design of G‐Protein‐Coupled Receptor Polypharmacology. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202101478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Stefanie Kampen
- Science for Life Laboratory Department of Cell and Molecular Biology Uppsala University 75124 Uppsala Sweden
| | - Duc Duy Vo
- Science for Life Laboratory Department of Cell and Molecular Biology Uppsala University 75124 Uppsala Sweden
| | - Xiaoqun Zhang
- Department of Clinical Neuroscience Karolinska Institute 17177 Stockholm Sweden
| | - Nicolas Panel
- Science for Life Laboratory Department of Cell and Molecular Biology Uppsala University 75124 Uppsala Sweden
| | - Yunting Yang
- Department of Clinical Neuroscience Karolinska Institute 17177 Stockholm Sweden
| | - Mariama Jaiteh
- Science for Life Laboratory Department of Cell and Molecular Biology Uppsala University 75124 Uppsala Sweden
| | - Pierre Matricon
- Science for Life Laboratory Department of Cell and Molecular Biology Uppsala University 75124 Uppsala Sweden
| | - Per Svenningsson
- Department of Clinical Neuroscience Karolinska Institute 17177 Stockholm Sweden
| | - Jose Brea
- USEF Screening Platform-BioFarma Research Group Centre for Research in Molecular Medicine and Chronic Diseases University of Santiago de Compostela 15706 Santiago de Compostela Spain
| | - Maria Isabel Loza
- USEF Screening Platform-BioFarma Research Group Centre for Research in Molecular Medicine and Chronic Diseases University of Santiago de Compostela 15706 Santiago de Compostela Spain
| | - Jan Kihlberg
- Department of Chemistry-BMC Uppsala University 75123 Uppsala Sweden
| | - Jens Carlsson
- Science for Life Laboratory Department of Cell and Molecular Biology Uppsala University 75124 Uppsala Sweden
| |
Collapse
|
36
|
Kampen S, Duy Vo D, Zhang X, Panel N, Yang Y, Jaiteh M, Matricon P, Svenningsson P, Brea J, Loza MI, Kihlberg J, Carlsson J. Structure-Guided Design of G-Protein-Coupled Receptor Polypharmacology. Angew Chem Int Ed Engl 2021; 60:18022-18030. [PMID: 33904641 PMCID: PMC8456950 DOI: 10.1002/anie.202101478] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Indexed: 12/29/2022]
Abstract
Many diseases are polygenic and can only be treated efficiently with drugs that modulate multiple targets. However, rational design of compounds with multi-target profiles is rarely pursued because it is considered too difficult, in particular if the drug must enter the central nervous system. Here, a structure-based strategy to identify dual-target ligands of G-protein-coupled receptors is presented. We use this approach to design compounds that both antagonize the A2A adenosine receptor and activate the D2 dopamine receptor, which have excellent potential as antiparkinson drugs. Atomic resolution models of the receptors guided generation of a chemical library with compounds designed to occupy orthosteric and secondary binding pockets in both targets. Structure-based virtual screens identified ten compounds, of which three had affinity for both targets. One of these scaffolds was optimized to nanomolar dual-target activity and showed the predicted pharmacodynamic effect in a rat model of Parkinsonism.
Collapse
Affiliation(s)
- Stefanie Kampen
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, 75124, Uppsala, Sweden
| | - Duc Duy Vo
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, 75124, Uppsala, Sweden
| | - Xiaoqun Zhang
- Department of Clinical Neuroscience, Karolinska Institute, 17177, Stockholm, Sweden
| | - Nicolas Panel
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, 75124, Uppsala, Sweden
| | - Yunting Yang
- Department of Clinical Neuroscience, Karolinska Institute, 17177, Stockholm, Sweden
| | - Mariama Jaiteh
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, 75124, Uppsala, Sweden
| | - Pierre Matricon
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, 75124, Uppsala, Sweden
| | - Per Svenningsson
- Department of Clinical Neuroscience, Karolinska Institute, 17177, Stockholm, Sweden
| | - Jose Brea
- USEF Screening Platform-BioFarma Research Group, Centre for Research in Molecular Medicine and Chronic Diseases, University of Santiago de Compostela, 15706, Santiago, de Compostela, Spain
| | - Maria Isabel Loza
- USEF Screening Platform-BioFarma Research Group, Centre for Research in Molecular Medicine and Chronic Diseases, University of Santiago de Compostela, 15706, Santiago, de Compostela, Spain
| | - Jan Kihlberg
- Department of Chemistry-BMC, Uppsala University, 75123, Uppsala, Sweden
| | - Jens Carlsson
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, 75124, Uppsala, Sweden
| |
Collapse
|
37
|
Jamieson CS, Misa J, Tang Y, Billingsley JM. Biosynthesis and synthetic biology of psychoactive natural products. Chem Soc Rev 2021; 50:6950-7008. [PMID: 33908526 PMCID: PMC8217322 DOI: 10.1039/d1cs00065a] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Psychoactive natural products play an integral role in the modern world. The tremendous structural complexity displayed by such molecules confers diverse biological activities of significant medicinal value and sociocultural impact. Accordingly, in the last two centuries, immense effort has been devoted towards establishing how plants, animals, and fungi synthesize complex natural products from simple metabolic precursors. The recent explosion of genomics data and molecular biology tools has enabled the identification of genes encoding proteins that catalyze individual biosynthetic steps. Once fully elucidated, the "biosynthetic pathways" are often comparable to organic syntheses in elegance and yield. Additionally, the discovery of biosynthetic enzymes provides powerful catalysts which may be repurposed for synthetic biology applications, or implemented with chemoenzymatic synthetic approaches. In this review, we discuss the progress that has been made toward biosynthetic pathway elucidation amongst four classes of psychoactive natural products: hallucinogens, stimulants, cannabinoids, and opioids. Compounds of diverse biosynthetic origin - terpene, amino acid, polyketide - are identified, and notable mechanisms of key scaffold transforming steps are highlighted. We also provide a description of subsequent applications of the biosynthetic machinery, with an emphasis placed on the synthetic biology and metabolic engineering strategies enabling heterologous production.
Collapse
Affiliation(s)
- Cooper S Jamieson
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA, USA.
| | - Joshua Misa
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA, USA.
| | - Yi Tang
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA, USA. and Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA, USA.
| | - John M Billingsley
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA, USA. and Invizyne Technologies, Inc., Monrovia, CA, USA
| |
Collapse
|
38
|
Pan S, Liang S, Wang X. ADORA1 promotes nasopharyngeal carcinoma cell progression through regulation of PI3K/AKT/GSK-3β/β-catenin signaling. Life Sci 2021; 278:119581. [PMID: 33961854 DOI: 10.1016/j.lfs.2021.119581] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 04/11/2021] [Accepted: 04/26/2021] [Indexed: 11/18/2022]
Abstract
AIMS For most human cancers, the expression pattern and biological function of ADORA1 (Adenosine A1 Receptor) are largely unknown. This study has been designed to explore the clinical significance and the mechanism of ADORA1 in nasopharyngeal carcinoma (NPC) cells. MATERIALS AND METHODS The level of ADORA1 in NPC and its adjacent tissues was analyzed by IHC, real-time PCR and western blotting. MTT and colony formation assays were used to determine the cell viability post ADORA1 overexpression or knockdown. Wound-healing assay and Transwell assay were used to analyze the effect of ADORA1 on migration and invasion. Moreover, the effect of ADORA1 on tumor growth was also studied in vivo by using xenograft mouse model. The regulation of ADORA1 on PI3K/AKT/GSK-3β/β-catenin pathway was determined by western blotting and TOP-Flash luciferase assay. KEY FINDINGS Primary NPC exhibits overexpression of ADORA1, which is related to the overexpression of its mRNA. Ectopic expression of ADORA1 promotes the proliferation, invasion and migration in NPC cells. The apoptosis, however, is suppressed. ADORA1 silencing was found to exert opposite effects in in vitro studies and produced a significant inhibitory effect on murine xenograft tumor growth in vivo experiments. Besides, ADORA1 also triggers the PI3K/AKT/GSK-3β/β-catenin intracellular oncogenic pathway for signal transduction. Inhibition of this pathway by PI3K inhibitor LY294002 obstructed the impact of ADORA1 on tumor development in cells with ADORA1-overexpression. SIGNIFICANCE ADORA1 has been identified as an important oncoprotein, promoting tumor cell proliferation via PI3K/AKT/GSK-3β/β-catenin signaling pathway in NPC.
Collapse
Affiliation(s)
- Suming Pan
- Department of Radiation Oncology, Yue Bei People's Hospital, Shaoguan, Guangdong, China.
| | - Sixian Liang
- Department of Radiation Oncology, Yue Bei People's Hospital, Shaoguan, Guangdong, China
| | - Xianyan Wang
- Department of Radiation Oncology, Yue Bei People's Hospital, Shaoguan, Guangdong, China
| |
Collapse
|
39
|
Do HN, Akhter S, Miao Y. Pathways and Mechanism of Caffeine Binding to Human Adenosine A 2A Receptor. Front Mol Biosci 2021; 8:673170. [PMID: 33987207 PMCID: PMC8111288 DOI: 10.3389/fmolb.2021.673170] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 03/24/2021] [Indexed: 11/13/2022] Open
Abstract
Caffeine (CFF) is a common antagonist to the four subtypes of adenosine G-protein-coupled receptors (GPCRs), which are critical drug targets for treating heart failure, cancer, and neurological diseases. However, the pathways and mechanism of CFF binding to the target receptors remain unclear. In this study, we have performed all-atom-enhanced sampling simulations using a robust Gaussian-accelerated molecular dynamics (GaMD) method to elucidate the binding mechanism of CFF to human adenosine A2A receptor (A2AAR). Multiple 500–1,000 ns GaMD simulations captured both binding and dissociation of CFF in the A2AAR. The GaMD-predicted binding poses of CFF were highly consistent with the x-ray crystal conformations with a characteristic hydrogen bond formed between CFF and residue N6.55 in the receptor. In addition, a low-energy intermediate binding conformation was revealed for CFF at the receptor extracellular mouth between ECL2 and TM1. While the ligand-binding pathways of the A2AAR were found similar to those of other class A GPCRs identified from previous studies, the ECL2 with high sequence divergence serves as an attractive target site for designing allosteric modulators as selective drugs of the A2AAR.
Collapse
Affiliation(s)
- Hung N Do
- Center for Computational Biology and Department of Molecular Biosciences, University of Kansas, Lawrence, KS, United States
| | - Sana Akhter
- Center for Computational Biology and Department of Molecular Biosciences, University of Kansas, Lawrence, KS, United States
| | - Yinglong Miao
- Center for Computational Biology and Department of Molecular Biosciences, University of Kansas, Lawrence, KS, United States
| |
Collapse
|
40
|
Atif M, Alsrhani A, Naz F, Imran M, Imran M, Ullah MI, Alameen AAM, Gondal TA, Raza Q. Targeting Adenosine Receptors in Neurological Diseases. Cell Reprogram 2021; 23:57-72. [PMID: 33861641 DOI: 10.1089/cell.2020.0087] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Adenosine plays a significant role in neurotransmission process by controlling the blood pressure, while adenosine triphosphate (ATP) acts as a neuromodulator and neurotransmitter and by activation of P2 receptors, regulates the contractility of the heart. Adenosine signaling is essential in the process of regeneration by regulating proliferation, differentiation, and apoptosis of stem cells. In this review, we have selected neurological disorders (Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, multiple sclerosis, and epilepsy) with clinical trials using antagonists and epigenetic tools targeting adenosine receptor as a therapeutic approach in the treatment of these disorders. Promising results have been reported from many clinical trials. It has been found that higher expression levels of A2A and P2X7 receptors in neurological disorders further complicate the disease condition. Therefore, modulations of these receptors by using antagonists of these receptors or SAM (S-adenosylmethionine) therapy as an epigenetic tool could be useful in reversing the complications of these disorders. Finally, we suggest that modulation of adenosine receptors in neurological disorders can increase the regenerative phase by increasing the rate of proliferation and differentiation in the damaged tissues.
Collapse
Affiliation(s)
- Muhmmad Atif
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia
| | - Abdullah Alsrhani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia
| | - Farrah Naz
- Department of Microbiology, Government College University, Faisalabad, Pakistan
| | - Muhammad Imran
- University Institute of Diet and Nutritional Sciences, Faculty of Allied Health Sciences, The University of Lahore, Lahore, Pakistan
| | - Muhammad Imran
- Department of Microbiology, University of Health Sciences, Lahore, Pakistan
| | - Muhammad Ikram Ullah
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia
| | - Ayman A M Alameen
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia.,Department of Chemical Pathology, Faculty of Medical Laboratory Sciences, University of Khartoum, Khartoum, Sudan
| | - Tanweer Aslam Gondal
- School of Exercise and Nutrition, Faculty of Health, Deakin University, Victoria, Australia
| | - Qaisar Raza
- Department of Clinical Nutrition, NUR International University, Lahore, Pakistan
| |
Collapse
|
41
|
Ahmad T, Linares J, Valencia DN, Agarwal A. Severe Shivering as an Adverse Effect of Regadenoson Myocardial Perfusion Imaging. Cureus 2021; 13:e14091. [PMID: 33927915 PMCID: PMC8075772 DOI: 10.7759/cureus.14091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Regadenoson myocardial perfusion imaging (MPI) is a widely used screening study for patients with an intermediate pretest probability of coronary artery disease (CAD). Via selective agonism of the adenosine A2A receptor, regadenoson can induce coronary steal, revealing stenotic vessel territory through transient ischemia. Common side effects of this medication include chest pain, shortness of breath, nausea, vomiting, atrioventricular block, seizure, and allergic reactions. Here we present a case of severe shivering and chest tightness after the administration of regadenoson, along with a physiologic explanation and treatment.
Collapse
Affiliation(s)
- Tanjeev Ahmad
- Department of Internal Medicine, Division of Cardiovascular Medicine, Boonshoft School of Medicine, Wright State University, Dayton, USA
| | - Juan Linares
- Department of Internal Medicine, Division of Cardiovascular Medicine, Kettering Medical Center, Dayton, USA.,Department of Internal Medicine, Division of Cardiovascular Medicine, Boonshoft School of Medicine, Wright State University, Dayton, USA.,Department of Cardiology, Dayton Veterans Affairs Medical Center, Dayton, USA
| | - Damian N Valencia
- Department of Internal Medicine, Division of Cardiovascular Medicine, Kettering Medical Center, Dayton, USA.,Department of Internal Medicine, Division of Cardiovascular Medicine, Boonshoft School of Medicine, Wright State University, Dayton, USA.,Department of Cardiology, Dayton Veterans Affairs Medical Center, Dayton, USA
| | - Ajay Agarwal
- Department of Cardiology, Dayton Veterans Affairs Medical Center, Dayton, USA.,Department of Internal Medicine, Division of Interventional Cardiology, Boonshoft School of Medicine, Wright State University, Dayton, USA
| |
Collapse
|
42
|
Kalash L, Winfield I, Safitri D, Bermudez M, Carvalho S, Glen R, Ladds G, Bender A. Structure-based identification of dual ligands at the A 2AR and PDE10A with anti-proliferative effects in lung cancer cell-lines. J Cheminform 2021; 13:17. [PMID: 33658076 PMCID: PMC7927403 DOI: 10.1186/s13321-021-00492-5] [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: 12/09/2020] [Accepted: 02/01/2021] [Indexed: 11/10/2022] Open
Abstract
Enhanced/prolonged cAMP signalling has been suggested as a suppressor of cancer proliferation. Interestingly, two key modulators that elevate cAMP, the A2A receptor (A2AR) and phosphodiesterase 10A (PDE10A), are differentially co-expressed in various types of non-small lung cancer (NSCLC) cell-lines. Thus, finding dual-target compounds, which are simultaneously agonists at the A2AR whilst also inhibiting PDE10A, could be a novel anti-proliferative approach. Using ligand- and structure-based modelling combined with MD simulations (which identified Val84 displacement as a novel conformational descriptor of A2AR activation), a series of known PDE10A inhibitors were shown to dock to the orthosteric site of the A2AR. Subsequent in-vitro analysis confirmed that these compounds bind to the A2AR and exhibit dual-activity at both the A2AR and PDE10A. Furthermore, many of the compounds exhibited promising anti-proliferative effects upon NSCLC cell-lines, which directly correlated with the expression of both PDE10A and the A2AR. Thus, we propose a structure-based methodology, which has been validated in in-vitro binding and functional assays, and demonstrated a promising therapeutic value.
Collapse
Affiliation(s)
- Leen Kalash
- Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Lensfield Road, CB21EW, Cambridge, UK
- GlaxoSmithKline, Gunnels Wood Road, Hertfordshire, SG1 2NY, Stevenage, UK
| | - Ian Winfield
- Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Lensfield Road, CB21EW, Cambridge, UK
- Department of Pharmacology, University of Cambridge, Tennis Court Road, CB2 1PD, Cambridge, UK
| | - Dewi Safitri
- Department of Pharmacology, University of Cambridge, Tennis Court Road, CB2 1PD, Cambridge, UK
- Pharmacology and Clinical Pharmacy Research Group, School of Pharmacy, Bandung Institute of Technology, 40132, Bandung, Indonesia
| | - Marcel Bermudez
- Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Lensfield Road, CB21EW, Cambridge, UK
- Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Straße 2 und 4, 14195, Berlin, Germany
| | - Sabrina Carvalho
- Department of Pharmacology, University of Cambridge, Tennis Court Road, CB2 1PD, Cambridge, UK
| | - Robert Glen
- Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Lensfield Road, CB21EW, Cambridge, UK
- Department of Metabolism Digestion and Reproduction, Faculty of Medicine, Imperial College London, SW7 2AZ, London, UK
| | - Graham Ladds
- Department of Pharmacology, University of Cambridge, Tennis Court Road, CB2 1PD, Cambridge, UK.
| | - Andreas Bender
- Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Lensfield Road, CB21EW, Cambridge, UK.
| |
Collapse
|
43
|
Giuliani AL, Sarti AC, Di Virgilio F. Ectonucleotidases in Acute and Chronic Inflammation. Front Pharmacol 2021; 11:619458. [PMID: 33613285 PMCID: PMC7887318 DOI: 10.3389/fphar.2020.619458] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 12/21/2020] [Indexed: 12/16/2022] Open
Abstract
Ectonucleotidases are extracellular enzymes with a pivotal role in inflammation that hydrolyse extracellular purine and pyrimidine nucleotides, e.g., ATP, UTP, ADP, UDP, AMP and NAD+. Ectonucleotidases, expressed by virtually all cell types, immune cells included, either as plasma membrane-associated or secreted enzymes, are classified into four main families: 1) nucleoside triphosphate diphosphohydrolases (NTPDases), 2) nicotinamide adenine dinucleotide glycohydrolase (NAD glycohydrolase/ADP-ribosyl cyclase/cyclic ADP-ribose hydrolase 1), 3) ecto-5′-nucleotidase (NT5E), and 4) ecto-nucleotide pyrophosphatase/phosphodiesterases (NPPs). Concentration of ATP, UTP and NAD+ can be increased in the extracellular space thanks to un-regulated, e.g., cell damage or cell death, or regulated processes. Regulated processes include secretory exocytosis, connexin or pannexin hemichannels, ATP binding cassette (ABC) transporters, calcium homeostasis modulator (CALMH) channels, the ATP-gated P2X7 receptor, maxi-anion channels (MACs) and volume regulated ion channels (VRACs). Hydrolysis of extracellular purine nucleotides generates adenosine, an important immunosuppressant. Extracellular nucleotides and nucleosides initiate or dampen inflammation via P2 and P1 receptors, respectively. All these agents, depending on their level of expression or activation and on the agonist concentration, are potent modulators of inflammation and key promoters of host defences, immune cells activation, pathogen clearance, tissue repair and regeneration. Thus, their knowledge is of great importance for a full understanding of the pathophysiology of acute and chronic inflammatory diseases. A selection of these pathologies will be briefly discussed here.
Collapse
Affiliation(s)
- Anna Lisa Giuliani
- Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Alba Clara Sarti
- Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Francesco Di Virgilio
- Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| |
Collapse
|
44
|
Spinozzi E, Baldassarri C, Acquaticci L, Del Bello F, Grifantini M, Cappellacci L, Riccardo P. Adenosine receptors as promising targets for the management of ocular diseases. Med Chem Res 2021; 30:353-370. [PMID: 33519168 PMCID: PMC7829661 DOI: 10.1007/s00044-021-02704-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 12/01/2020] [Indexed: 12/15/2022]
Abstract
The ocular drug discovery arena has undergone a significant improvement in the last few years culminating in the FDA approvals of 8 new drugs. However, despite a large number of drugs, generics, and combination products available, it remains an urgent need to find breakthrough strategies and therapies for tackling ocular diseases. Targeting the adenosinergic system may represent an innovative strategy for discovering new ocular therapeutics. This review focused on the recent advance in the field and described the numerous nucleoside and non-nucleoside modulators of the four adenosine receptors (ARs) used as potential tools or clinical drug candidates.
Collapse
Affiliation(s)
- Eleonora Spinozzi
- School of Pharmacy Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, 62032 Camerino, Italy
| | - Cecilia Baldassarri
- School of Pharmacy Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, 62032 Camerino, Italy
| | - Laura Acquaticci
- School of Pharmacy Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, 62032 Camerino, Italy
| | - Fabio Del Bello
- School of Pharmacy Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, 62032 Camerino, Italy
| | - Mario Grifantini
- School of Pharmacy Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, 62032 Camerino, Italy
| | - Loredana Cappellacci
- School of Pharmacy Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, 62032 Camerino, Italy
| | - Petrelli Riccardo
- School of Pharmacy Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, 62032 Camerino, Italy
| |
Collapse
|
45
|
Therapeutic Path to Double Knockout: Investigating the Selective Dual-Inhibitory Mechanisms of Adenosine Receptors A1 and A2 by a Novel Methoxy-Substituted Benzofuran Derivative in the Treatment of Parkinson's Disease. Cell Biochem Biophys 2020; 79:25-36. [PMID: 33222095 DOI: 10.1007/s12013-020-00957-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/05/2020] [Indexed: 10/22/2022]
Abstract
The dual inhibition of adenosine receptors A1 (A1 AR) and A2 (A2A AR) has been considered as an efficient strategy in the treatment of Parkinson's disease (PD). This led to the recent development of a series of methoxy-substituted benzofuran derivatives among which compound 3j exhibited dual-inhibitory potencies in the micromolar range. Therefore, in this study, we seek to resolve the mechanisms by which this novel compound elicits its selective dual targeting against A1 AR and A2A AR. Unique to the binding of 3j in both proteins, from our findings, is the ring-ring interaction elicited by A1Phe275 (→ A2Phe170) with the benzofuran ring of the compound. As observed, this π-stacking interaction contributes notably to the stability of 3j at the active sites of A1 and A2A AR. Besides, conserved active site residues in the proteins such as A1Ala170 (→ A2Ala65), A1Ile173 (→ A2Ile68), A1Val191 (→ A2Val86), A1Leu192 (→ A2Leu87), A1Ala195 (→ A2Ala90), A1Met284 (→ A2Met179), A1Tyr375 (→ A2Tyr369), A1Ile378 (→ A2Ile372), and A1His382 (→ A2His376) were commonly involved with other ring substituents which further complement the dual binding and stability of 3j. This reflects a similar interaction mechanism that involved aromatic (π) interactions. Consequentially, vdW energies contributed immensely to the dual binding of the compound, which culminated in high ΔGbinds that were homogenous in both proteins. Furthermore, 3j commonly disrupted the stable and compact conformation of A1 and A2A AR, coupled with their active sites where Cα deviations were relatively high. Ligand mobility analysis also revealed that both compounds exhibited a similar motion pattern at the active site of the proteins relative to their optimal dual binding. We believe that findings from this study with significantly aid the structure-based design of highly selective dual-inhibitors of A1 and A2A AR.
Collapse
|
46
|
Kumari S, Carmona AV, Tiwari AK, Trippier PC. Amide Bond Bioisosteres: Strategies, Synthesis, and Successes. J Med Chem 2020; 63:12290-12358. [PMID: 32686940 DOI: 10.1021/acs.jmedchem.0c00530] [Citation(s) in RCA: 254] [Impact Index Per Article: 63.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The amide functional group plays a key role in the composition of biomolecules, including many clinically approved drugs. Bioisosterism is widely employed in the rational modification of lead compounds, being used to increase potency, enhance selectivity, improve pharmacokinetic properties, eliminate toxicity, and acquire novel chemical space to secure intellectual property. The introduction of a bioisostere leads to structural changes in molecular size, shape, electronic distribution, polarity, pKa, dipole or polarizability, which can be either favorable or detrimental to biological activity. This approach has opened up new avenues in drug design and development resulting in more efficient drug candidates introduced onto the market as well as in the clinical pipeline. Herein, we review the strategic decisions in selecting an amide bioisostere (the why), synthetic routes to each (the how), and success stories of each bioisostere (the implementation) to provide a comprehensive overview of this important toolbox for medicinal chemists.
Collapse
Affiliation(s)
- Shikha Kumari
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Angelica V Carmona
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Amit K Tiwari
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, The University of Toledo, Toledo, Ohio 43614, United States
| | - Paul C Trippier
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States.,Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States.,UNMC Center for Drug Discovery, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| |
Collapse
|
47
|
The Effect of Caffeine on the Risk and Progression of Parkinson's Disease: A Meta-Analysis. Nutrients 2020; 12:nu12061860. [PMID: 32580456 PMCID: PMC7353179 DOI: 10.3390/nu12061860] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/20/2020] [Accepted: 06/22/2020] [Indexed: 12/21/2022] Open
Abstract
Coffee and caffeine are speculated to be associated with the reduced risk of Parkinson's disease (PD). The present study aimed to investigate the disease-modifying potential of caffeine on PD, either for healthy people or patients, through a meta-analysis. The electronic databases were searched using terms related to PD and coffee and caffeinated food products. Articles were included only upon fulfillment of clear diagnostic criteria for PD and details regarding their caffeine content. Reference lists of relevant articles were reviewed to identify eligible studies not shortlisted using these terms. In total, the present study enrolled 13 studies, nine were categorized into a healthy cohort and the rest into a PD cohort. The individuals in the healthy cohort with regular caffeine consumption had a significantly lower risk of PD during follow-up evaluation (hazard ratio (HR) = 0.797, 95% CI = 0.748-0.849, p < 0.001). The outcomes of disease progression in PD cohorts included dyskinesia, motor fluctuation, symptom onset, and levodopa initiation. Individuals consuming caffeine presented a significantly lower rate of PD progression (HR = 0.834, 95% CI = 0.707-0.984, p = 0.03). In conclusion, caffeine modified disease risk and progression in PD, among both healthy individuals or those with PD. Potential biological benefits, such as those obtained from adenosine 2A receptor antagonism, may require further investigation for designing new drugs.
Collapse
|
48
|
Lecas L, Hartmann L, Caro L, Mohamed-Bouteben S, Raingeval C, Krimm I, Wagner R, Dugas V, Demesmay C. Miniaturized weak affinity chromatography for ligand identification of nanodiscs-embedded G-protein coupled receptors. Anal Chim Acta 2020; 1113:26-35. [DOI: 10.1016/j.aca.2020.03.062] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 03/27/2020] [Accepted: 03/31/2020] [Indexed: 12/14/2022]
|
49
|
De Filippo E, Hinz S, Pellizzari V, Deganutti G, El-Tayeb A, Navarro G, Franco R, Moro S, Schiedel AC, Müller CE. A2A and A2B adenosine receptors: The extracellular loop 2 determines high (A2A) or low affinity (A2B) for adenosine. Biochem Pharmacol 2020; 172:113718. [DOI: 10.1016/j.bcp.2019.113718] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 11/13/2019] [Indexed: 12/21/2022]
|
50
|
Ballante F, Rudling A, Zeifman A, Luttens A, Vo DD, Irwin JJ, Kihlberg J, Brea J, Loza MI, Carlsson J. Docking Finds GPCR Ligands in Dark Chemical Matter. J Med Chem 2019; 63:613-620. [DOI: 10.1021/acs.jmedchem.9b01560] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Flavio Ballante
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, BMC, Box 596, SE-751 24 Uppsala, Sweden
| | - Axel Rudling
- Department of Biochemistry and Biophysics, Stockholm University, SE-106 91 Stockholm, Sweden
| | - Alexey Zeifman
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, BMC, Box 596, SE-751 24 Uppsala, Sweden
- Department of Biochemistry and Biophysics, Stockholm University, SE-106 91 Stockholm, Sweden
| | - Andreas Luttens
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, BMC, Box 596, SE-751 24 Uppsala, Sweden
| | - Duy Duc Vo
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, BMC, Box 596, SE-751 24 Uppsala, Sweden
| | - John J. Irwin
- Department of Pharmaceutical Chemistry, University of California, San Francisco, Byers Hall, 1700 4th Street, San Francisco, California 94158-2330, United States
| | - Jan Kihlberg
- Department of Chemistry-BMC, Uppsala University, Box 576, SE-751 23 Uppsala, Sweden
| | - Jose Brea
- Innopharma Screening Platform-BioFarma Research Group, Centre for Research in Molecular Medicine and Chronic Diseases, University of Santiago de Compostela, 15706 Santiago de Compostela, Spain
| | - Maria Isabel Loza
- Innopharma Screening Platform-BioFarma Research Group, Centre for Research in Molecular Medicine and Chronic Diseases, University of Santiago de Compostela, 15706 Santiago de Compostela, Spain
| | - Jens Carlsson
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, BMC, Box 596, SE-751 24 Uppsala, Sweden
| |
Collapse
|