1
|
Laketa D, Lavrnja I. Extracellular Purine Metabolism-Potential Target in Multiple Sclerosis. Mol Neurobiol 2024; 61:8361-8386. [PMID: 38499905 DOI: 10.1007/s12035-024-04104-9] [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: 07/26/2023] [Accepted: 03/07/2024] [Indexed: 03/20/2024]
Abstract
The purinergic signaling system comprises a complex network of extracellular purines and purine-metabolizing ectoenzymes, nucleotide and nucleoside receptors, ATP release channels, and nucleoside transporters. Because of its immunomodulatory function, this system is critically involved in the pathogenesis of multiple sclerosis (MS) and its best-characterized animal model, experimental autoimmune encephalomyelitis (EAE). MS is a chronic neuroinflammatory demyelinating and neurodegenerative disease with autoimmune etiology and great heterogeneity, mostly affecting young adults and leading to permanent disability. In MS/EAE, alterations were detected in almost all components of the purinergic signaling system in both peripheral immune cells and central nervous system (CNS) glial cells, which play an important role in the pathogenesis of the disease. A decrease in extracellular ATP levels and an increase in its downstream metabolites, particularly adenosine and inosine, were frequently observed at MS, indicating a shift in metabolism toward an anti-inflammatory environment. Accordingly, upregulation of the major ectonucleotidase tandem CD39/CD73 was detected in the blood cells and CNS of relapsing-remitting MS patients. Based on the postulated role of A2A receptors in the transition from acute to chronic neuroinflammation, the association of variants of the adenosine deaminase gene with the severity of MS, and the beneficial effects of inosine treatment in EAE, the adenosinergic system emerged as a promising target in neuroinflammation. More recently, several publications have identified ADP-dependent P2Y12 receptors and the major extracellular ADP producing enzyme nucleoside triphosphate diphosphohydrolase 2 (NTPDase2) as novel potential targets in MS.
Collapse
Affiliation(s)
- Danijela Laketa
- Department of General Physiology and Biophysics, Institute for Physiology and Biochemistry "Ivan Djaja", Faculty of Biology, University of Belgrade, Studentski Trg 3, Belgrade, Republic of Serbia.
| | - Irena Lavrnja
- Institute for Biological Research, Sinisa Stankovic" - National Institute of the Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, Belgrade, Republic of Serbia
| |
Collapse
|
2
|
Martínez-Gallego I, Rodríguez-Moreno A. Adenosine and Cortical Plasticity. Neuroscientist 2024:10738584241236773. [PMID: 38497585 DOI: 10.1177/10738584241236773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Brain plasticity is the ability of the nervous system to change its structure and functioning in response to experiences. These changes occur mainly at synaptic connections, and this plasticity is named synaptic plasticity. During postnatal development, environmental influences trigger changes in synaptic plasticity that will play a crucial role in the formation and refinement of brain circuits and their functions in adulthood. One of the greatest challenges of present neuroscience is to try to explain how synaptic connections change and cortical maps are formed and modified to generate the most suitable adaptive behavior after different external stimuli. Adenosine is emerging as a key player in these plastic changes at different brain areas. Here, we review the current knowledge of the mechanisms responsible for the induction and duration of synaptic plasticity at different postnatal brain development stages in which adenosine, probably released by astrocytes, directly participates in the induction of long-term synaptic plasticity and in the control of the duration of plasticity windows at different cortical synapses. In addition, we comment on the role of the different adenosine receptors in brain diseases and on the potential therapeutic effects of acting via adenosine receptors.
Collapse
Affiliation(s)
- Irene Martínez-Gallego
- Laboratory of Cellular Neuroscience and Plasticity, Department of Physiology, Anatomy and Cell Biology, University Pablo de Olavide, Seville, Spain
| | - Antonio Rodríguez-Moreno
- Laboratory of Cellular Neuroscience and Plasticity, Department of Physiology, Anatomy and Cell Biology, University Pablo de Olavide, Seville, Spain
| |
Collapse
|
3
|
Song X, Kirtipal N, Lee S, Malý P, Bharadwaj S. Current therapeutic targets and multifaceted physiological impacts of caffeine. Phytother Res 2023; 37:5558-5598. [PMID: 37679309 DOI: 10.1002/ptr.8000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 08/04/2023] [Accepted: 08/17/2023] [Indexed: 09/09/2023]
Abstract
Caffeine, which shares consubstantial structural similarity with purine adenosine, has been demonstrated as a nonselective adenosine receptor antagonist for eliciting most of the biological functions at physiologically relevant dosages. Accumulating evidence supports caffeine's beneficial effects against different disorders, such as total cardiovascular diseases and type 2 diabetes. Conversely, paradoxical effects are also linked to caffeine ingestion in humans including hypertension-hypotension and tachycardia-bradycardia. These observations suggest the association of caffeine action with its ingested concentration and/or concurrent interaction with preferential molecular targets to direct explicit events in the human body. Thus, a coherent analysis of the functional targets of caffeine, relevant to normal physiology, and disease pathophysiology, is required to understand the pharmacology of caffeine. This review provides a broad overview of the experimentally validated targets of caffeine, particularly those of therapeutic interest, and the impacts of caffeine on organ-specific physiology and pathophysiology. Overall, the available empirical and epidemiological evidence supports the dose-dependent functional activities of caffeine and advocates for further studies to get insights into the caffeine-induced changes under specific conditions, such as asthma, DNA repair, and cancer, in view of its therapeutic applications.
Collapse
Affiliation(s)
- Xinjie Song
- Zhejiang Provincial Key Lab for Chemical and Biological Processing Technology of Farm Product, School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou, China
| | - Nikhil Kirtipal
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Sunjae Lee
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Petr Malý
- Laboratory of Ligand Engineering, Institute of Biotechnology of the Czech Academy of Sciences v.v.i, BIOCEV Research Center, Vestec, Czech Republic
| | - Shiv Bharadwaj
- Laboratory of Ligand Engineering, Institute of Biotechnology of the Czech Academy of Sciences v.v.i, BIOCEV Research Center, Vestec, Czech Republic
| |
Collapse
|
4
|
Zabihian A, Sayyad FZ, Hashemi SM, Shami Tanha R, Hooshmand M, Gharaghani S. DEDTI versus IEDTI: efficient and predictive models of drug-target interactions. Sci Rep 2023; 13:9238. [PMID: 37286613 DOI: 10.1038/s41598-023-36438-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 06/03/2023] [Indexed: 06/09/2023] Open
Abstract
Drug repurposing is an active area of research that aims to decrease the cost and time of drug development. Most of those efforts are primarily concerned with the prediction of drug-target interactions. Many evaluation models, from matrix factorization to more cutting-edge deep neural networks, have come to the scene to identify such relations. Some predictive models are devoted to the prediction's quality, and others are devoted to the efficiency of the predictive models, e.g., embedding generation. In this work, we propose new representations of drugs and targets useful for more prediction and analysis. Using these representations, we propose two inductive, deep network models of IEDTI and DEDTI for drug-target interaction prediction. Both of them use the accumulation of new representations. The IEDTI takes advantage of triplet and maps the input accumulated similarity features into meaningful embedding corresponding vectors. Then, it applies a deep predictive model to each drug-target pair to evaluate their interaction. The DEDTI directly uses the accumulated similarity feature vectors of drugs and targets and applies a predictive model on each pair to identify their interactions. We have done a comprehensive simulation on the DTINet dataset as well as gold standard datasets, and the results show that DEDTI outperforms IEDTI and the state-of-the-art models. In addition, we conduct a docking study on new predicted interactions between two drug-target pairs, and the results confirm acceptable drug-target binding affinity between both predicted pairs.
Collapse
Affiliation(s)
- Arash Zabihian
- Laboratory of Bioinformatics and Drug Design (LBD), Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
- Department of Bioinformatics, Kish International Campus, University of Tehran, Kish, Iran
| | - Faeze Zakaryapour Sayyad
- Department of Computer Science and Information Technology, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan, Iran
| | - Seyyed Morteza Hashemi
- Department of Computer Science and Information Technology, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan, Iran
| | - Reza Shami Tanha
- Department of Computer Science and Information Technology, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan, Iran
| | - Mohsen Hooshmand
- Department of Computer Science and Information Technology, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan, Iran.
| | - Sajjad Gharaghani
- Laboratory of Bioinformatics and Drug Design (LBD), Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran.
| |
Collapse
|
5
|
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
|
6
|
Chang H, Huo M, Zhang Q, Zhou M, Zhang Y, Si Y, Zhang D, Guo Y, Fang Y. Flexible needle-type Microbiosensor for real-time monitoring traditional acupuncture-mediated adenosine release In vivo. Biosens Bioelectron 2023; 235:115383. [PMID: 37207583 DOI: 10.1016/j.bios.2023.115383] [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: 02/15/2023] [Revised: 04/19/2023] [Accepted: 05/08/2023] [Indexed: 05/21/2023]
Abstract
Rapid adenosine (ADO) signaling, on the time frame of seconds, regulates physiological and pathological processes, including the therapeutic efficacy of acupuncture. Nevertheless, standard monitoring strategies are limited by poor temporal resolution. Herein, an implantable needle-type microsensor capable of monitoring ADO release in vivo in response to acupuncture in real time has been developed. Electrocatalytic Prussian Blue nanoparticles, an immobilized multienzyme system, and a permselective poly-o-phenylenediamine-based membrane were used for the sequential modification of the sensing region of the electrode. The resultant sensor can perform amperometric measurements of ADO levels in response to a very low level of applied potential (-0.05 V vs Ag/AgCl). This microsensor also functioned across a broad linear range (0-50 μM) and exhibited good sensitivity (1.1 nA/μM) with a rapid response time of under 5 s. Importantly, the sensor also exhibited good reproducibility and high selectivity. For in vivo animal studies, the microsensor was employed for the continuous assessment of instantaneous ADO release at the ST36 (Zusanli) acupoint when this acupoint was subjected to twirling-rotating acupuncture manipulation. Benefiting from superior sensor in vivo performance and stability, the positive correlation between the variability in acupuncture-induced ADO release and the stimulus intensity levels that affect the clinical benefit can be demonstrated for the first time. Overall, these results highlight a powerful approach to analyzing the in vivo physiological effects of acupuncture, expanding application realm of micro-nano sensor technology on a fast time scale.
Collapse
Affiliation(s)
- Hongen Chang
- Research Center of Experimental Acupuncture Science, College of Acumox and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China
| | - Mingzhu Huo
- Research Center of Experimental Acupuncture Science, College of Acumox and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China
| | - Qingxiang Zhang
- Research Center of Experimental Acupuncture Science, College of Acumox and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China
| | - Mengmeng Zhou
- Research Center of Experimental Acupuncture Science, College of Acumox and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China
| | - Youlin Zhang
- Research Center of Experimental Acupuncture Science, College of Acumox and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China
| | - Yuxin Si
- Research Center of Experimental Acupuncture Science, College of Acumox and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China
| | - Di Zhang
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Haihe Laboratory of Modern Chinese Medicine, Tianjin, 301617, PR China.
| | - Yi Guo
- Research Center of Experimental Acupuncture Science, College of Acumox and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China.
| | - Yuxin Fang
- Research Center of Experimental Acupuncture Science, College of Acumox and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China.
| |
Collapse
|
7
|
Liu X, Ye K, van Vlijmen HWT, IJzerman AP, van Westen GJP. DrugEx v3: scaffold-constrained drug design with graph transformer-based reinforcement learning. J Cheminform 2023; 15:24. [PMID: 36803659 PMCID: PMC9940339 DOI: 10.1186/s13321-023-00694-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 02/06/2023] [Indexed: 02/22/2023] Open
Abstract
Rational drug design often starts from specific scaffolds to which side chains/substituents are added or modified due to the large drug-like chemical space available to search for novel drug-like molecules. With the rapid growth of deep learning in drug discovery, a variety of effective approaches have been developed for de novo drug design. In previous work we proposed a method named DrugEx, which can be applied in polypharmacology based on multi-objective deep reinforcement learning. However, the previous version is trained under fixed objectives and does not allow users to input any prior information (i.e. a desired scaffold). In order to improve the general applicability, we updated DrugEx to design drug molecules based on scaffolds which consist of multiple fragments provided by users. Here, a Transformer model was employed to generate molecular structures. The Transformer is a multi-head self-attention deep learning model containing an encoder to receive scaffolds as input and a decoder to generate molecules as output. In order to deal with the graph representation of molecules a novel positional encoding for each atom and bond based on an adjacency matrix was proposed, extending the architecture of the Transformer. The graph Transformer model contains growing and connecting procedures for molecule generation starting from a given scaffold based on fragments. Moreover, the generator was trained under a reinforcement learning framework to increase the number of desired ligands. As a proof of concept, the method was applied to design ligands for the adenosine A2A receptor (A2AAR) and compared with SMILES-based methods. The results show that 100% of the generated molecules are valid and most of them had a high predicted affinity value towards A2AAR with given scaffolds.
Collapse
Affiliation(s)
- Xuhan Liu
- grid.5132.50000 0001 2312 1970Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Einsteinweg 55, Leiden, The Netherlands
| | - Kai Ye
- grid.43169.390000 0001 0599 1243School of Electrics and Information Engineering, Xi’an Jiaotong University, 28 XianningW Rd, Xi’an, China
| | - Herman W. T. van Vlijmen
- grid.5132.50000 0001 2312 1970Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Einsteinweg 55, Leiden, The Netherlands ,grid.419619.20000 0004 0623 0341Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Adriaan P. IJzerman
- grid.5132.50000 0001 2312 1970Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Einsteinweg 55, Leiden, The Netherlands
| | - Gerard J. P. van Westen
- grid.5132.50000 0001 2312 1970Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Einsteinweg 55, Leiden, The Netherlands
| |
Collapse
|
8
|
Iser IC, Vedovatto S, Oliveira FD, Beckenkamp LR, Lenz G, Wink MR. The crossroads of adenosinergic pathway and epithelial-mesenchymal plasticity in cancer. Semin Cancer Biol 2022; 86:202-213. [PMID: 35779713 DOI: 10.1016/j.semcancer.2022.06.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 06/24/2022] [Accepted: 06/26/2022] [Indexed: 10/31/2022]
Abstract
Epithelial-mesenchymal transition (EMT) is a key mechanism related to tumor progression, invasion, metastasis, resistance to therapy and poor prognosis in several types of cancer. However, targeting EMT or partial-EMT, as well as the molecules involved in this process, has remained a challenge. Recently, the CD73 enzyme, which hydrolyzes AMP to produce adenosine (ADO), has been linked to the EMT process. This relationship is not only due to the production of the immunosuppressant ADO but also to its role as a receptor for extracellular matrix proteins, being involved in cell adhesion and migration. This article reviews the crosstalk between the adenosinergic pathway and the EMT program and the impact of this interrelation on cancer development and progression. An in silico analysis of RNAseq datasets showed that several tumor types have a significant correlation between an EMT score and NT5E (CD73) and ENTPD1 (CD39) expressions, with the strongest correlations in prostate adenocarcinoma. Furthermore, it is evident that the cooperation between EMT and adenosinergic pathway in tumor progression is context and tumor-dependent. The increased knowledge about this topic will help broaden the view to explore new treatments and therapies for different types of cancer.
Collapse
Affiliation(s)
- Isabele Cristiana Iser
- Department of Basics Health Sciences and Laboratory of Cell Biology, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
| | - Samlai Vedovatto
- Department of Biophysics and Center of Biotechnology, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Fernanda Dittrich Oliveira
- Department of Biophysics and Center of Biotechnology, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Liziane Raquel Beckenkamp
- Department of Basics Health Sciences and Laboratory of Cell Biology, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
| | - Guido Lenz
- Department of Biophysics and Center of Biotechnology, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Márcia Rosângela Wink
- Department of Basics Health Sciences and Laboratory of Cell Biology, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil.
| |
Collapse
|
9
|
Xiang Q, Zhao Y, Lin J, Jiang S, Li W. Epigenetic modifications in spinal ligament aging. Ageing Res Rev 2022; 77:101598. [PMID: 35218968 DOI: 10.1016/j.arr.2022.101598] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 02/16/2022] [Accepted: 02/21/2022] [Indexed: 02/07/2023]
Abstract
Spinal stenosis is a common degenerative spine disorder in the aged population and the spinal ligament aging is a main contributor to this chronic disease. However, the underlying mechanisms of spinal ligament aging remain unclear. Epigenetics is the study of heritable and reversible changes in the function of a gene or genome that occur without any alteration in the primary DNA sequence. Epigenetic alterations have been demonstrated to play crucial roles in age-related diseases and conditions, and they are recently studied as biomarkers and therapeutic targets in the field of cancer research. The main epigenetic modifications, including DNA methylation alteration, histone modifications as well as dysregulated noncoding RNA modulation, have all been implicated in spinal ligament aging diseases. DNA methylation modulates the expression of critical genes including WNT5A, GDNF, ACSM5, miR-497 and miR-195 during spinal ligament degeneration. Histone modifications widely affect gene expression and obvious histone modification abnormalities have been found in spinal ligament aging. MicroRNAs (miRNAs), long noncoding RNAs (lncRNAs) and circular RNAs (circRNAs) exert crucial regulating effects on spinal ligament aging conditions via targeting various osteogenic or fibrogenic differentiation related genes. To our knowledge, there is no systematic review yet to summarize the involvement of epigenetic mechanisms of spinal ligament aging in degenerative spinal diseases. In this study, we systematically discussed the different epigenetic modifications and their potential functions in spinal ligament aging process.
Collapse
|
10
|
Pacini ESA, Satori NA, Jackson EK, Godinho RO. Extracellular cAMP-Adenosine Pathway Signaling: A Potential Therapeutic Target in Chronic Inflammatory Airway Diseases. Front Immunol 2022; 13:866097. [PMID: 35479074 PMCID: PMC9038211 DOI: 10.3389/fimmu.2022.866097] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 03/21/2022] [Indexed: 12/25/2022] Open
Abstract
Adenosine is a purine nucleoside that, via activation of distinct G protein-coupled receptors, modulates inflammation and immune responses. Under pathological conditions and in response to inflammatory stimuli, extracellular ATP is released from damaged cells and is metabolized to extracellular adenosine. However, studies over the past 30 years provide strong evidence for another source of extracellular adenosine, namely the “cAMP-adenosine pathway.” The cAMP-adenosine pathway is a biochemical mechanism mediated by ATP-binding cassette transporters that facilitate cAMP efflux and by specific ectoenzymes that convert cAMP to AMP (ecto-PDEs) and AMP to adenosine (ecto-nucleotidases such as CD73). Importantly, the cAMP-adenosine pathway is operative in many cell types, including those of the airways. In airways, β2-adrenoceptor agonists, which are used as bronchodilators for treatment of asthma and chronic respiratory diseases, stimulate cAMP efflux and thus trigger the extracellular cAMP-adenosine pathway leading to increased concentrations of extracellular adenosine in airways. In the airways, extracellular adenosine exerts pro-inflammatory effects and induces bronchoconstriction in patients with asthma and chronic obstructive pulmonary diseases. These considerations lead to the hypothesis that the cAMP-adenosine pathway attenuates the efficacy of β2-adrenoceptor agonists. Indeed, our recent findings support this view. In this mini-review, we will highlight the potential role of the extracellular cAMP-adenosine pathway in chronic respiratory inflammatory disorders, and we will explore how extracellular cAMP could interfere with the regulatory effects of intracellular cAMP on airway smooth muscle and innate immune cell function. Finally, we will discuss therapeutic possibilities targeting the extracellular cAMP-adenosine pathway for treatment of these respiratory diseases.
Collapse
Affiliation(s)
- Enio Setsuo Arakaki Pacini
- Division of Cellular Pharmacology, Department of Pharmacology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Naiara Ayako Satori
- Division of Cellular Pharmacology, Department of Pharmacology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Edwin Kerry Jackson
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Rosely Oliveira Godinho
- Division of Cellular Pharmacology, Department of Pharmacology, Universidade Federal de São Paulo, São Paulo, Brazil
- *Correspondence: Rosely Oliveira Godinho,
| |
Collapse
|
11
|
Sancho M, Klug NR, Mughal A, Koide M, Huerta de la Cruz S, Heppner TJ, Bonev AD, Hill-Eubanks D, Nelson MT. Adenosine signaling activates ATP-sensitive K + channels in endothelial cells and pericytes in CNS capillaries. Sci Signal 2022; 15:eabl5405. [PMID: 35349300 DOI: 10.1126/scisignal.abl5405] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The dense network of capillaries composed of capillary endothelial cells (cECs) and pericytes lies in close proximity to all neurons, ideally positioning it to sense neuron- and glial-derived compounds that enhance regional and global cerebral perfusion. The membrane potential (VM) of vascular cells serves as the physiological bridge that translates brain activity into vascular function. In other beds, the ATP-sensitive K+ (KATP) channel regulates VM in vascular smooth muscle, which is absent in the capillary network. Here, with transgenic mice that expressed a dominant-negative mutant of the pore-forming Kir6.1 subunit specifically in brain cECs or pericytes, we demonstrated that KATP channels were present in both cell types and robustly controlled VM. We further showed that the signaling nucleotide adenosine acted through A2A receptors and the Gαs/cAMP/PKA pathway to activate capillary KATP channels. Moreover, KATP channel stimulation in vivo increased cerebral blood flow (CBF), an effect that was blunted by expression of the dominant-negative Kir6.1 mutant in either capillary cell type. These findings establish an important role for KATP channels in cECs and pericytes in the regulation of CBF.
Collapse
Affiliation(s)
- Maria Sancho
- Department of Pharmacology, University of Vermont, Burlington, VT 05405-0068, USA
| | - Nicholas R Klug
- Department of Pharmacology, University of Vermont, Burlington, VT 05405-0068, USA
| | - Amreen Mughal
- Department of Pharmacology, University of Vermont, Burlington, VT 05405-0068, USA
| | - Masayo Koide
- Department of Pharmacology, University of Vermont, Burlington, VT 05405-0068, USA.,Vermont Center for Cardiovascular and Brain Health, Larner College of Medicine, University of Vermont, Burlington, VT 05405-0068, USA
| | | | - Thomas J Heppner
- Department of Pharmacology, University of Vermont, Burlington, VT 05405-0068, USA
| | - Adrian D Bonev
- Department of Pharmacology, University of Vermont, Burlington, VT 05405-0068, USA
| | - David Hill-Eubanks
- Department of Pharmacology, University of Vermont, Burlington, VT 05405-0068, USA
| | - Mark T Nelson
- Department of Pharmacology, University of Vermont, Burlington, VT 05405-0068, USA.,Vermont Center for Cardiovascular and Brain Health, Larner College of Medicine, University of Vermont, Burlington, VT 05405-0068, USA.,Division of Cardiovascular Sciences, University of Manchester, Manchester M13 9PL, UK
| |
Collapse
|
12
|
Adenosine Receptor Signaling in Diseases with Focus on Cancer. JORJANI BIOMEDICINE JOURNAL 2022. [DOI: 10.52547/jorjanibiomedj.10.1.41] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
|
13
|
Kotulova J, Lonova K, Kubickova A, Vrbkova J, Kourilova P, Hajduch M, Dzubak P. 2‑Cl‑IB‑MECA regulates the proliferative and drug resistance pathways, and facilitates chemosensitivity in pancreatic and liver cancer cell lines. Int J Mol Med 2022; 49:31. [PMID: 35039871 PMCID: PMC8788926 DOI: 10.3892/ijmm.2022.5086] [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: 10/11/2021] [Accepted: 12/14/2021] [Indexed: 12/24/2022] Open
Abstract
Specific A3 adenosine receptor (A3AR) agonist, 2-chloro-N6-(3-iodobenzyl)-5′-N-methylcarboxamidoadenosine (2-Cl-IB-MECA), demonstrates anti-proliferative effects on various types of tumor. In the present study, the cytotoxicity of 2-Cl-IB-MECA was analyzed in a panel of tumor and non-tumor cell lines and its anticancer mechanisms in JoPaca-1 pancreatic and Hep-3B hepatocellular carcinoma cell lines were also investigated. Initially, decreased tumor cell proliferation, cell accumulation in the G1 phase and inhibition of DNA and RNA synthesis was found. Furthermore, western blot analysis showed decreased protein expression level of β-catenin, patched1 (Ptch1) and glioma-associated oncogene homolog zinc finger protein 1 (Gli1), which are components of the Wnt/β-catenin and Sonic hedgehog/Ptch/Gli transduction pathways. In concordance with these findings, the protein expression levels of cyclin D1 and c-Myc were reduced. Using a luciferase assay, it was revealed for the first time a decrease in β-catenin transcriptional activity, as an early event following 2-Cl-IB-MECA treatment. In addition, the protein expression levels of multidrug resistance-associated protein 1 and P-glycoprotein (P-gp) were reduced and the P-gp xenobiotic efflux function was also reduced. Next, the enhancing effects of 2-Cl-IB-MECA on the cytotoxicity of conventional chemotherapy was investigated. It was found that 2-Cl-IB-MECA enhanced carboplatin and doxorubicin cytotoxic effects in the JoPaca-1 and Hep-3B cell lines, and a greater synergy was found in the highly tumorigenic JoPaca-1 cell line. This provides a novel in vitro rationale for the utiliza- tion of 2-Cl-IB-MECA in combination with chemotherapeutic agents, not only for hepatocellular carcinoma, but also for pancreatic cancer. Other currently used conventional chemo- therapeutics, fluorouracil and gemcitabine, showed synergy only when combined with high doses of 2-Cl-IB-MECA. Notably, experiments with A3AR-specific antagonist, N-[9-Chloro-2-(2-furanyl)(1,2,4)-triazolo(1,5-c)quinazolin-5-yl] benzene acetamide, revealed that 2-Cl-IB-MECA had antitumor effects via both A3AR-dependent and -independent pathways. In conclusion, the present study identified novel antitumor mechanisms of 2-Cl-IB-MECA in pancreatic and hepatocellular carcinoma in vitro that further underscores the importance of A3AR agonists in cancer therapy.
Collapse
Affiliation(s)
- Jana Kotulova
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University Olomouc, 77900 Olomouc, Czech Republic
| | - Katerina Lonova
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University Olomouc, 77900 Olomouc, Czech Republic
| | - Agata Kubickova
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University Olomouc, 77900 Olomouc, Czech Republic
| | - Jana Vrbkova
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University Olomouc, 77900 Olomouc, Czech Republic
| | - Pavla Kourilova
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University Olomouc, 77900 Olomouc, Czech Republic
| | - Marian Hajduch
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University Olomouc, 77900 Olomouc, Czech Republic
| | - Petr Dzubak
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University Olomouc, 77900 Olomouc, Czech Republic
| |
Collapse
|
14
|
Matthee C, Terre'Blanche G, Janse van Rensburg HD, Aucamp J, Legoabe LJ. Chalcone-inspired rA 1 /A 2A adenosine receptor ligands: Ring closure as an alternative to a reactive substructure. Chem Biol Drug Des 2021; 99:416-437. [PMID: 34878728 DOI: 10.1111/cbdd.13999] [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: 09/07/2021] [Revised: 10/21/2021] [Accepted: 10/30/2021] [Indexed: 11/30/2022]
Abstract
Over the past few years, great progress has been made in the development of high-affinity adenosine A1 and/or A2A receptor antagonists-promising agents for the potential treatment of Parkinson's disease. Unfortunately, many of these compounds raise structure-related concerns. The present study investigated the effect of ring closures on the rA1 /A2A affinity of compounds containing a highly reactive α,β-unsaturated carbonyl system, hence providing insight into the potential of heterocycles to address these concerns. A total of 12 heterocyclic compounds were synthesised and evaluated in silico and in vitro. The test compounds performed well upon qualitative assessment of drug-likeness and were generally found to be free from potentially problematic fragments. Most also showed low/weak cytotoxicity. Results from radioligand binding experiments confirm that heterocycles (particularly 2-substituted 3-cyanopyridines) can replace the promiscuous α,β-unsaturated ketone functional group without compromising A1 /A2A affinity. Structure-activity relationships highlighted the importance of hydrogen bonds in binding to the receptors of interest. Compounds 3c (rA1 Ki = 16 nM; rA2A Ki = 65 nM) and 8a (rA1 Ki = 102 nM; rA2A Ki = 37 nM), which both act as A1 antagonists, showed significant dual A1 /A2A affinity and may, therefore, inspire further investigation into heterocycles as potentially safe and potent adenosine receptor antagonists.
Collapse
Affiliation(s)
- Chrisna Matthee
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom, South Africa
| | - Gisella Terre'Blanche
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom, South Africa.,Pharmaceutical Chemistry, School of Pharmacy, North-West University, Potchefstroom, South Africa
| | | | - Janine Aucamp
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom, South Africa
| | - Lesetja J Legoabe
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom, South Africa
| |
Collapse
|
15
|
Blockade of adenosine A 2A receptor alleviates cognitive dysfunction after chronic exposure to intermittent hypoxia in mice. Exp Neurol 2021; 350:113929. [PMID: 34813840 DOI: 10.1016/j.expneurol.2021.113929] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 10/25/2021] [Accepted: 11/16/2021] [Indexed: 11/22/2022]
Abstract
Obstructive sleep apnea-hypopnea syndrome (OSAHS) is widely known for its multiple systems damage, especially neurocognitive deficits in children. Since their discovery, adenosine A2A receptors (A2ARs) have been considered as key elements in signaling pathways mediating neurodegenerative diseases such as Huntington's and Alzheimer's, as well as cognitive function regulation. Herein, we investigated A2AR role in cognitive impairment induced by chronic intermittent hypoxia (CIH). Mice were exposed to CIH 7 h every day for 4 weeks, and intraperitoneally injected with A2AR agonist CGS21680 or A2AR antagonist SCH58261 half an hour before IH exposure daily. The 8-arm radial arm maze was utilized to assess spatial memory after CIH exposures.To validate findings using pharmacology, the impact of intermittent hypoxia was investigated in A2AR knockout mice. CIH-induced memory dysfunction was manifested by increased error rates in the radial arm maze test. The behavioral changes were associated with hippocampal pathology, neuronal apoptosis, and synaptic plasticity impairment. The stimulation of adenosine A2AR exacerbated memory impairment with more serious neuropathological damage, attenuated long-term potentiation (LTP), syntaxin down-regulation, and increased BDNF protein. Moreover, apoptosis-promoting protein cleaved caspase-3 was upregulated while anti-apoptotic protein Bcl-2 was downregulated. Consistent with these findings, A2AR inhibition with SCH58261 and A2AR deletion exhibited the opposite result. Overall, these findings suggest that A2AR plays a critical role in CIH-induced impairment of learning and memory by accelerating hippocampal neuronal apoptosis and reducing synaptic plasticity. Blockade of adenosine A2A receptor alleviates cognitive dysfunction after chronic exposure to intermittent hypoxia in mice.
Collapse
|
16
|
Liu X, Ye K, van Vlijmen HWT, Emmerich MTM, IJzerman AP, van Westen GJP. DrugEx v2: de novo design of drug molecules by Pareto-based multi-objective reinforcement learning in polypharmacology. J Cheminform 2021; 13:85. [PMID: 34772471 PMCID: PMC8588612 DOI: 10.1186/s13321-021-00561-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 10/12/2021] [Indexed: 12/03/2022] Open
Abstract
In polypharmacology drugs are required to bind to multiple specific targets, for example to enhance efficacy or to reduce resistance formation. Although deep learning has achieved a breakthrough in de novo design in drug discovery, most of its applications only focus on a single drug target to generate drug-like active molecules. However, in reality drug molecules often interact with more than one target which can have desired (polypharmacology) or undesired (toxicity) effects. In a previous study we proposed a new method named DrugEx that integrates an exploration strategy into RNN-based reinforcement learning to improve the diversity of the generated molecules. Here, we extended our DrugEx algorithm with multi-objective optimization to generate drug-like molecules towards multiple targets or one specific target while avoiding off-targets (the two adenosine receptors, A1AR and A2AAR, and the potassium ion channel hERG in this study). In our model, we applied an RNN as the agent and machine learning predictors as the environment. Both the agent and the environment were pre-trained in advance and then interplayed under a reinforcement learning framework. The concept of evolutionary algorithms was merged into our method such that crossover and mutation operations were implemented by the same deep learning model as the agent. During the training loop, the agent generates a batch of SMILES-based molecules. Subsequently scores for all objectives provided by the environment are used to construct Pareto ranks of the generated molecules. For this ranking a non-dominated sorting algorithm and a Tanimoto-based crowding distance algorithm using chemical fingerprints are applied. Here, we adopted GPU acceleration to speed up the process of Pareto optimization. The final reward of each molecule is calculated based on the Pareto ranking with the ranking selection algorithm. The agent is trained under the guidance of the reward to make sure it can generate desired molecules after convergence of the training process. All in all we demonstrate generation of compounds with a diverse predicted selectivity profile towards multiple targets, offering the potential of high efficacy and low toxicity.
Collapse
Affiliation(s)
- Xuhan Liu
- Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Kai Ye
- School of Electronics and Information Engineering, Xi'an Jiaotong University, 28 Xianning W Rd, Xi'an, China
| | - Herman W T van Vlijmen
- Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Einsteinweg 55, 2333 CC, Leiden, The Netherlands.,Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340, Beerse, Belgium
| | - Michael T M Emmerich
- Leiden Institute of Advanced Computer Science, Niels Bohrweg 1, 2333 CA, Leiden, The Netherlands
| | - Adriaan P IJzerman
- Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Gerard J P van Westen
- Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Einsteinweg 55, 2333 CC, Leiden, The Netherlands.
| |
Collapse
|
17
|
Acute Effects of High Doses of Caffeine on Bar Velocity during the Bench Press Throw in Athletes Habituated to Caffeine: A Randomized, Double-Blind and Crossover Study. J Clin Med 2021; 10:jcm10194380. [PMID: 34640398 PMCID: PMC8509759 DOI: 10.3390/jcm10194380] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/21/2021] [Accepted: 09/23/2021] [Indexed: 11/16/2022] Open
Abstract
Chronic intake of caffeine may produce a reduction in the potential performance benefits obtained with the acute intake of this substance. For this reason, athletes habituated to caffeine often use high doses of caffeine (≥9 mg/kg) to overcome tolerance to caffeine ergogenicity due to chronic intake. The main objective of the current investigation was to evaluate the effects of high caffeine doses on bar velocity during an explosive bench press throw in athletes habituated to caffeine. Twelve resistance-trained athletes, with a moderate-to-high chronic intake of caffeine (~5.3 mg/kg/day) participated in a randomized double-blind and randomized experimental design. Each participant performed three identical experimental sessions 60 min after the intake of a placebo (PLAC) or after the intake of 9 (CAF-9) or 12 mg/kg (CAF-12) of caffeine. In each experimental session, the athletes performed five sets of two repetitions of the bench press throw exercise with a load equivalent to 30% of their one-repetition maximum. In comparison to PLAC, the intake of caffeine increased peak and mean velocity (p < 0.01) during the five sets of the bench press throw exercise. There were no significant differences in peak and mean bar velocity between the two doses of caffeine (CAF-9 vs. CAF-12; p = 0.91, = 0.96, respectively). The ingestion of high doses of caffeine was effective in producing an increase in mean and peak bar velocity during the bench press throw in a group of habitual caffeine users. However, using CAF-12 did not offer additional benefits for performance with respect to CAF-9.
Collapse
|
18
|
Matthee C, Terre'Blanche G, Legoabe LJ, Janse van Rensburg HD. Exploration of chalcones and related heterocycle compounds as ligands of adenosine receptors: therapeutics development. Mol Divers 2021; 26:1779-1821. [PMID: 34176057 DOI: 10.1007/s11030-021-10257-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 06/15/2021] [Indexed: 12/20/2022]
Abstract
Adenosine receptors (ARs) are ubiquitously distributed throughout the mammalian body where they are involved in an extensive list of physiological and pathological processes that scientists have only begun to decipher. Resultantly, AR agonists and antagonists have been the focus of multiple drug design and development programmes within the past few decades. Considered to be a privileged scaffold in medicinal chemistry, the chalcone framework has attracted a substantial amount of interest in this regard. Due to the potential liabilities associated with its structure, however, it has become necessary to explore other potentially promising compounds, such as heterocycles, which have successfully been obtained from chalcone precursors in the past. This review aims to summarise the emerging therapeutic importance of adenosine receptors and their ligands, especially in the central nervous system (CNS), while highlighting chalcone and heterocyclic derivatives as promising AR ligand lead compounds.
Collapse
Affiliation(s)
- Chrisna Matthee
- Pharmaceutical Chemistry, School of Pharmacy, North-West University, Private Bag X6001, Potchefstroom, 2520, North West, South Africa
| | - Gisella Terre'Blanche
- Pharmaceutical Chemistry, School of Pharmacy, North-West University, Private Bag X6001, Potchefstroom, 2520, North West, South Africa.,Centre of Excellence for Pharmaceutical Sciences, School of Pharmacy, North-West University, Private Bag X6001, Potchefstroom, 2520, North West, South Africa
| | - Lesetja J Legoabe
- Centre of Excellence for Pharmaceutical Sciences, School of Pharmacy, North-West University, Private Bag X6001, Potchefstroom, 2520, North West, South Africa
| | - Helena D Janse van Rensburg
- Pharmaceutical Chemistry, School of Pharmacy, North-West University, Private Bag X6001, Potchefstroom, 2520, North West, South Africa.
| |
Collapse
|
19
|
Zefferino R, Piccoli C, Di Gioia S, Capitanio N, Conese M. How Cells Communicate with Each Other in the Tumor Microenvironment: Suggestions to Design Novel Therapeutic Strategies in Cancer Disease. Int J Mol Sci 2021; 22:ijms22052550. [PMID: 33806300 PMCID: PMC7961918 DOI: 10.3390/ijms22052550] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 02/25/2021] [Accepted: 02/26/2021] [Indexed: 02/06/2023] Open
Abstract
Connexin- and pannexin (Panx)-formed hemichannels (HCs) and gap junctions (GJs) operate an interaction with the extracellular matrix and GJ intercellular communication (GJIC), and on account of this they are involved in cancer onset and progression towards invasiveness and metastatization. When we deal with cancer, it is not correct to omit the immune system, as well as neglecting its role in resisting or succumbing to formation and progression of incipient neoplasia until the formation of micrometastasis, nevertheless what really occurs in the tumor microenvironment (TME), which are the main players and which are the tumor or body allies, is still unclear. The goal of this article is to discuss how the pivotal players act, which can enhance or contrast cancer progression during two important process: "Activating Invasion and Metastasis" and the "Avoiding Immune Destruction", with a particular emphasis on the interplay among GJIC, Panx-HCs, and the purinergic system in the TME without disregarding the inflammasome and cytokines thereof derived. In particular, the complex and contrasting roles of Panx1/P2X7R signalosome in tumor facilitation and/or inhibition is discussed in regard to the early/late phases of the carcinogenesis. Finally, considering this complex interplay in the TME between cancer cells, stromal cells, immune cells, and focusing on their means of communication, we should be capable of revealing harmful messages that help the cancer growth and transform them in body allies, thus designing novel therapeutic strategies to fight cancer in a personalized manner.
Collapse
Affiliation(s)
- Roberto Zefferino
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (S.D.G.); (M.C.)
- Correspondence: ; Tel.: +39-0881-884673
| | - Claudia Piccoli
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy; (C.P.); (N.C.)
| | - Sante Di Gioia
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (S.D.G.); (M.C.)
| | - Nazzareno Capitanio
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy; (C.P.); (N.C.)
| | - Massimo Conese
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (S.D.G.); (M.C.)
| |
Collapse
|
20
|
Szopa A, Socała K, Serefko A, Doboszewska U, Wróbel A, Poleszak E, Wlaź P. Purinergic transmission in depressive disorders. Pharmacol Ther 2021; 224:107821. [PMID: 33607148 DOI: 10.1016/j.pharmthera.2021.107821] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 12/11/2020] [Indexed: 12/13/2022]
Abstract
Purinergic signaling involves the actions of purine nucleotides and nucleosides (such as adenosine) at P1 (adenosine), P2X, and P2Y receptors. Here, we present recent data contributing to a comprehensive overview of the association between purinergic signaling and depression. We start with background information on adenosine production and metabolism, followed by a detailed characterization of P1 and P2 receptors, with an emphasis on their expression and function in the brain as well as on their ligands. We provide data suggestive of altered metabolism of adenosine in depressed patients, which might be regarded as a disease biomarker. We then turn to considerable amount of preclinical/behavioral data obtained with the aid of the forced swim test, tail suspension test, learned helplessness model, or unpredictable chronic mild stress model and genetic activation/inactivation of P1 or P2 receptors as well as nonselective or selective ligands of P1 or P2 receptors. We also aimed to discuss the reason underlying discrepancies observed in such studies.
Collapse
Affiliation(s)
- Aleksandra Szopa
- Department of Applied and Social Pharmacy, Laboratory of Preclinical Testing, Medical University of Lublin, Chodźki 1, PL 20-093 Lublin, Poland.
| | - Katarzyna Socała
- Department of Animal Physiology and Pharmacology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Akademicka 19, PL 20-033 Lublin, Poland
| | - Anna Serefko
- Department of Applied and Social Pharmacy, Laboratory of Preclinical Testing, Medical University of Lublin, Chodźki 1, PL 20-093 Lublin, Poland
| | - Urszula Doboszewska
- Department of Animal Physiology and Pharmacology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Akademicka 19, PL 20-033 Lublin, Poland
| | - Andrzej Wróbel
- Second Department of Gynecology, Medical University of Lublin, Jaczewskiego 8, PL 20-090 Lublin, Poland
| | - Ewa Poleszak
- Department of Applied and Social Pharmacy, Laboratory of Preclinical Testing, Medical University of Lublin, Chodźki 1, PL 20-093 Lublin, Poland.
| | - Piotr Wlaź
- Department of Animal Physiology and Pharmacology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Akademicka 19, PL 20-033 Lublin, Poland.
| |
Collapse
|
21
|
Wang X, van Westen GJP, Heitman LH, IJzerman AP. G protein-coupled receptors expressed and studied in yeast. The adenosine receptor as a prime example. Biochem Pharmacol 2020; 187:114370. [PMID: 33338473 DOI: 10.1016/j.bcp.2020.114370] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 12/11/2020] [Accepted: 12/11/2020] [Indexed: 11/25/2022]
Abstract
G protein-coupled receptors (GPCRs) are the largest class of membrane proteins with around 800 members in the human genome/proteome. Extracellular signals such as hormones and neurotransmitters regulate various biological processes via GPCRs, with GPCRs being the bodily target of 30-40% of current drugs on the market. Complete identification and understanding of GPCR functionality will provide opportunities for novel drug discovery. Yeast expresses three different endogenous GPCRs regulating pheromone and sugar sensing, with the pheromone pathway offering perspectives for the characterization of heterologous GPCR signaling. Moreover, yeast offers a ''null" background for studies on mammalian GPCRs, including GPCR activation and signaling, ligand identification, and characterization of disease-related mutations. This review focuses on modifications of the yeast pheromone signaling pathway for functional GPCR studies, and on opportunities and usage of the yeast system as a platform for human GPCR studies. Finally, this review discusses in some further detail studies of adenosine receptors heterologously expressed in yeast, and what Geoff Burnstock thought of this approach.
Collapse
Affiliation(s)
- Xuesong Wang
- Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Gerard J P van Westen
- Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Laura H Heitman
- Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Einsteinweg 55, 2333 CC Leiden, The Netherlands; Oncode Institute, Leiden, The Netherlands
| | - Adriaan P IJzerman
- Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| |
Collapse
|
22
|
Mohd Nasir NA, Agarwal R, Krasilnikova A, Sheikh Abdul Kadir SH, Iezhitsa I. Effect of trans-resveratrol on dexamethasone-induced changes in the expression of MMPs by human trabecular meshwork cells: Involvement of adenosine A 1 receptors and NFkB. Eur J Pharmacol 2020; 887:173431. [PMID: 32758568 DOI: 10.1016/j.ejphar.2020.173431] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 07/25/2020] [Accepted: 07/26/2020] [Indexed: 01/02/2023]
Abstract
Intraocular pressure (IOP) lowering in glaucomatous eyes is currently achieved mainly by improved aqueous outflow via alternate drainage pathways. However, the focus is now shifting to trabecular meshwork (TM), the site or major pathological changes including increased extracellular matrix (ECM) deposition and reduced matrix metalloproteinases (MMPs) secretion by TM cells. Trans-resveratrol was previously shown to lower IOP and reduce ECM deposition; however, the mechanisms of action remain unclear. Therefore, we determined the effect of trans-resveratrol on MMP-2 and -9 expression by human TM cells (HTMCs) in the presence of dexamethasone and whether it also affects adenosine A1 receptors (A1AR) expression and nuclear factor kappa B (NFkB) activation. We observed that trans-resveratrol, 12.5 μM, increased MMP-2 and -9 protein expression by HTMCs despite exposure to dexamethasone (1.89- and 1.53-fold, respectively; P < 0.001). Further it was observed that trans-resveratrol increases A1AR expression in HTMC in the presence of dexamethasone (1.55-fold; P < 0.01). Trans-resveratrol also increased NFkB activation in the presence of dexamethasone and A1AR antagonist (P < 0.01 versus dexamethasone group). These effects of trans-resveratrol were associated with increased MMP -2 and -9 expression. It could be concluded that trans-resveratrol prevents dexamethasone-induced reduction in MMP-2 and -9 secretion by NFkB activation in HTMCs. This effect of trans-resveratrol is likely to involve increased A1AR expression.
Collapse
Affiliation(s)
- Normie Aida Mohd Nasir
- Center for Neuroscience Research (NeuRon), Faculty of Medicine, Universiti Teknologi MARA (UiTM), Sungai Buloh Campus, Selangor, Malaysia; Institute of Medical Molecular Biotechnology (IMMB), Universiti Teknologi MARA (UiTM), Sungai Buloh Campus, Selangor, Malaysia
| | - Renu Agarwal
- School of Medicine, International Medical University, Bukit Jalil, 57000, Kuala Lumpur, Malaysia.
| | - Anna Krasilnikova
- Faculty of Medicine, Universiti Teknologi MARA (UiTM), Sungai Buloh Campus, Selangor, Malaysia; Volgograd State Medical University, Department of Clinical Pharmacology & Intensive Care, Volgograd, Russia
| | - Siti Hamimah Sheikh Abdul Kadir
- Faculty of Medicine, Universiti Teknologi MARA (UiTM), Sungai Buloh Campus, Selangor, Malaysia; Institute of Medical Molecular Biotechnology (IMMB), Universiti Teknologi MARA (UiTM), Sungai Buloh Campus, Selangor, Malaysia
| | - Igor Iezhitsa
- School of Medicine, International Medical University, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| |
Collapse
|
23
|
Pieterse L, van der Walt MM, Terre'Blanche G. C2-substituted quinazolinone derivatives exhibit A 1 and/or A 2A adenosine receptor affinities in the low micromolar range. Bioorg Med Chem Lett 2020; 30:127274. [PMID: 32631506 DOI: 10.1016/j.bmcl.2020.127274] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 05/11/2020] [Accepted: 05/16/2020] [Indexed: 12/20/2022]
Abstract
Antagonists of the adenosine receptors (A1 and A2A subtypes) are widely researched as potential drug candidates for their role in Parkinson's disease-related cognitive deficits (A1 subtype), motor dysfunction (A2A subtype) and to exhibit neuroprotective properties (A2A subtype). Previously the benzo-α-pyrone based derivative, 3-phenyl-1H-2-benzopyran-1-one, was found to display both A1 and A2A adenosine receptor affinity in the low micromolar range. Prompted by this, the α-pyrone core was structurally modified to explore related benzoxazinone and quinazolinone homologues previously unknown as adenosine receptor antagonists. Overall, the C2-substituted quinazolinone analogues displayed superior A1 and A2A adenosine receptor affinity over their C2-substituted benzoxazinone homologues. The benzoxazinones were devoid of A2A adenosine receptor binding, with only two compounds displaying A1 adenosine receptor affinity. In turn, the quinazolinones displayed varying degrees of affinity (low micromolar range) towards the A1 and A2A adenosine receptor subtypes. The highest A1 adenosine receptor affinity and selectivity were favoured by methyl para-substitution of phenyl ring B (A1Ki = 2.50 μM). On the other hand, 3,4-dimethoxy substitution of phenyl ring B afforded the best A2A adenosine receptor binding (A2AKi = 2.81 μM) among the quinazolinones investigated. In conclusion, the quinazolinones are ideal lead compounds for further structural optimization to gain improved adenosine receptor affinity, which may find therapeutic relevance in Parkinson's disease-associated cognitive deficits and motor dysfunctions as well as exerting neuroprotective properties.
Collapse
Affiliation(s)
- Lianie Pieterse
- Centre of Excellence for Pharmaceutical Sciences, School of Pharmacy, North-West University, Private Bag X6001, Potchefstroom 2520, South Africa
| | - Mietha M van der Walt
- Centre of Excellence for Pharmaceutical Sciences, School of Pharmacy, North-West University, Private Bag X6001, Potchefstroom 2520, South Africa; Human Metabolomics, Faculty of Natural and Agricultural Science, North-West University, Private Bag X6001, Box 269, Potchefstroom 2531, South Africa.
| | - Gisella Terre'Blanche
- Centre of Excellence for Pharmaceutical Sciences, School of Pharmacy, North-West University, Private Bag X6001, Potchefstroom 2520, South Africa; Pharmaceutical Chemistry, School of Pharmacy, North-West University, Private Bag X6001, Potchefstroom 2520, South Africa
| |
Collapse
|
24
|
Prevention of Oxygen-Induced Inflammatory Lung Injury by Caffeine in Neonatal Rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:3840124. [PMID: 32831996 PMCID: PMC7429812 DOI: 10.1155/2020/3840124] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/15/2020] [Accepted: 07/18/2020] [Indexed: 12/26/2022]
Abstract
Background Preterm birth implies an array of respiratory diseases including apnea of prematurity and bronchopulmonary dysplasia (BPD). Caffeine has been introduced to treat apneas but also appears to reduce rates of BPD. Oxygen is essential when treating preterm infants with respiratory problems but high oxygen exposure aggravates BPD. This experimental study is aimed at investigating the action of caffeine on inflammatory response and cell death in pulmonary tissue in a hyperoxia-based model of BPD in the newborn rat. Material/Methods. Lung injury was induced by hyperoxic exposure with 80% oxygen for three (P3) or five (P5) postnatal days with or without recovery in ambient air until postnatal day 15 (P15). Newborn Wistar rats were treated with PBS or caffeine (10 mg/kg) every two days beginning at the day of birth. The effects of caffeine on hyperoxic-induced pulmonary inflammatory response were examined at P3 and P5 immediately after oxygen exposure or after recovery in ambient air (P15) by immunohistological staining and analysis of lung homogenates by ELISA and qPCR. Results Treatment with caffeine significantly attenuated changes in hyperoxia-induced cell death and apoptosis-associated factors. There was a significant decrease in proinflammatory mediators and redox-sensitive transcription factor NFκB in the hyperoxia-exposed lung tissue of the caffeine-treated group compared to the nontreated group. Moreover, treatment with caffeine under hyperoxia modulated the transcription of the adenosine receptor (Adora)1. Caffeine induced pulmonary chemokine and cytokine transcription followed by immune cell infiltration of alveolar macrophages as well as increased adenosine receptor (Adora1, 2a, and 2b) expression. Conclusions The present study investigating the impact of caffeine on the inflammatory response, pulmonary cell degeneration and modulation of adenosine receptor expression, provides further evidence that caffeine acts as an antioxidative and anti-inflammatory drug for experimental oxygen-mediated lung injury. Experimental studies may broaden the understanding of therapeutic use of caffeine in modulating detrimental mechanisms involved in BPD development.
Collapse
|
25
|
Characterization of cancer-related somatic mutations in the adenosine A2B receptor. Eur J Pharmacol 2020; 880:173126. [DOI: 10.1016/j.ejphar.2020.173126] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 04/16/2020] [Accepted: 04/20/2020] [Indexed: 01/10/2023]
|
26
|
Adenosine Receptor Agonists Increase the Inhibition of Platelet Function by P2Y 12 Antagonists in a cAMP- and Calcium-Dependent Manner. Pharmaceuticals (Basel) 2020; 13:ph13080177. [PMID: 32752089 PMCID: PMC7464091 DOI: 10.3390/ph13080177] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/30/2020] [Accepted: 07/30/2020] [Indexed: 12/22/2022] Open
Abstract
We have shown previously that platelet activity can be lowered through the simultaneous inhibition of P2Y12 receptor and activation of adenosine receptors (AR). This work explores this concept by testing the antiplatelet potential of nine AR agonists in combination with P2Y12 receptor antagonists—cangrelor and prasugrel metabolite. A panel of in vitro methods was used to assess platelet viability, P-selectin expression, GPIIb-IIIa activation, fibrinogen binding, calcium ion mobilization, VASP-P level, and cAMP formation, utilizing whole blood or isolated platelets from healthy volunteers. The AR agonists demonstrated anti-platelet effects, but stimulated signaling pathways to varying degrees. AR agonists and P2Y12 antagonists reduced expression of both P-selectin and the activated form of GPIIb-IIIa on platelets; however, the combined systems (AR agonist + P2Y12 antagonist) demonstrated stronger effects. The antiplatelet effects of AR when combined with P2Y12 were more pronounced with regard to exogenous fibrinogen binding and calcium mobilization. The cAMP levels in both resting and ADPactivated platelets were increased by AR agonist treatment, and more so when combined with P2Y12 inhibitor. In conclusion, as AR agonists are fast-acting compounds, the methods detecting early activation events are more suitable for assessing their antiplatelet action. The exogenous fibrinogen binding, calcium mobilisation and cAMP level turned out to be sensitive markers for detecting the inhibition caused by AR agonists alone or in combination with P2Y12 receptor antagonists.
Collapse
|
27
|
Borodovsky A, Barbon CM, Wang Y, Ye M, Prickett L, Chandra D, Shaw J, Deng N, Sachsenmeier K, Clarke JD, Linghu B, Brown GA, Brown J, Congreve M, Cheng RK, Dore AS, Hurrell E, Shao W, Woessner R, Reimer C, Drew L, Fawell S, Schuller AG, Mele DA. Small molecule AZD4635 inhibitor of A 2AR signaling rescues immune cell function including CD103 + dendritic cells enhancing anti-tumor immunity. J Immunother Cancer 2020; 8:jitc-2019-000417. [PMID: 32727810 PMCID: PMC7394305 DOI: 10.1136/jitc-2019-000417] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/24/2020] [Indexed: 02/04/2023] Open
Abstract
Accumulation of extracellular adenosine within the microenvironment is a strategy exploited by tumors to escape detection by the immune system. Adenosine signaling through the adenosine 2A receptor (A2AR) on immune cells elicits a range of immunosuppressive effects which promote tumor growth and limit the efficacy of immune checkpoint inhibitors. Preclinical data with A2AR inhibitors have demonstrated tumor regressions in mouse models by rescuing T cell function; however, the mechanism and role on other immune cells has not been fully elucidated.
Collapse
Affiliation(s)
| | | | - Yanjun Wang
- Bioscience, AstraZeneca R&D Boston, Waltham, Massachusetts, USA
| | - Minwei Ye
- Bioscience, AstraZeneca R&D Boston, Waltham, Massachusetts, USA
| | - Laura Prickett
- Bioscience, AstraZeneca R&D Boston, Waltham, Massachusetts, USA
| | - Dinesh Chandra
- Bioscience, AstraZeneca R&D Boston, Waltham, Massachusetts, USA
| | - Joseph Shaw
- Discovery Sciences, AstraZeneca PLC, Cambridge, Cambridgeshire, UK
| | - Nanhua Deng
- Bioscience, AstraZeneca R&D Boston, Waltham, Massachusetts, USA
| | - Kris Sachsenmeier
- Translational Medicine, AstraZeneca R&D Boston, Waltham, Massachusetts, USA
| | - James D Clarke
- Drug Metabolism and Pharamcokinetics, AstraZeneca, Cambridge, Cambridgeshire, UK
| | - Bolan Linghu
- Translational Medicine, AstraZeneca R&D Boston, Waltham, Massachusetts, USA
| | - Giles A Brown
- Discovery, Omass Technologies Ltd, Oxford, United Kingdom
| | - James Brown
- Heptares Therapeutics, Welwyn Garden City, California, USA
| | - Miles Congreve
- Heptares Therapeutics, Welwyn Garden City, California, USA
| | | | - Andrew S Dore
- Heptares Therapeutics, Welwyn Garden City, California, USA
| | - Edward Hurrell
- Heptares Therapeutics, Welwyn Garden City, California, USA
| | - Wenlin Shao
- Oncology, AstraZeneca R&D Boston, Waltham, Massachusetts, USA
| | - Richard Woessner
- Pharmacology, Blueprint Medicines, Cambridge, Massachusetts, USA
| | - Corinne Reimer
- Bioscience, AstraZeneca R&D Boston, Waltham, Massachusetts, USA
| | - Lisa Drew
- Bioscience, AstraZeneca R&D Boston, Waltham, Massachusetts, USA
| | - Stephen Fawell
- Bioscience, AstraZeneca R&D Boston, Waltham, Massachusetts, USA
| | | | - Deanna A Mele
- Bioscience, AstraZeneca R&D Boston, Waltham, Massachusetts, USA
| |
Collapse
|
28
|
Mallo-Abreu A, Prieto-Díaz R, Jespers W, Azuaje J, Majellaro M, Velando C, García-Mera X, Caamaño O, Brea J, Loza MI, Gutiérrez-de-Terán H, Sotelo E. Nitrogen-Walk Approach to Explore Bioisosteric Replacements in a Series of Potent A 2B Adenosine Receptor Antagonists. J Med Chem 2020; 63:7721-7739. [PMID: 32573250 DOI: 10.1021/acs.jmedchem.0c00564] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
A systematic exploration of bioisosteric replacements for furan and thiophene cores in a series of potent A2BAR antagonists has been carried out using the nitrogen-walk approach. A collection of 42 novel alkyl 4-substituted-2-methyl-1,4-dihydrobenzo[4,5]imidazo[1,2-a]pyrimidine-3-carboxylates, which contain 18 different pentagonal heterocyclic frameworks at position 4, was synthesized and evaluated. This study enabled the identification of new ligands that combine remarkable affinity (Ki < 30 nM) and exquisite selectivity. The structure-activity relationship (SAR) trends identified were substantiated by a molecular modeling study, based on a receptor-driven docking model and including a systematic free energy perturbation (FEP) study. Preliminary evaluation of the CYP3A4 and CYP2D6 inhibitory activity in optimized ligands evidenced weak and negligible activity, respectively. The stereospecific interaction between hA2BAR and the eutomer of the most attractive novel antagonist (S)-18g (Ki = 3.66 nM) was validated.
Collapse
Affiliation(s)
| | | | - Willem Jespers
- Department of Cell and Molecular Biology, Uppsala University, Uppsala SE 75124, Sweden
| | | | | | | | | | | | - José Brea
- Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - María I Loza
- Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | | | | |
Collapse
|
29
|
Załuski M, Schabikowski J, Jaśko P, Bryła A, Olejarz-Maciej A, Kaleta M, Głuch-Lutwin M, Brockmann A, Hinz S, Zygmunt M, Kuder K, Latacz G, Vielmuth C, Müller CE, Kieć-Kononowicz K. 8-Benzylaminoxanthine scaffold variations for selective ligands acting on adenosine A 2A receptors. Design, synthesis and biological evaluation. Bioorg Chem 2020; 101:104033. [PMID: 32629282 DOI: 10.1016/j.bioorg.2020.104033] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 06/01/2020] [Accepted: 06/15/2020] [Indexed: 11/24/2022]
Abstract
A library of 34 novel compounds based on a xanthine scaffold was explored in biological studies for interaction with adenosine receptors (ARs). Structural modifications of the xanthine core were introduced in the 8-position (benzylamino and benzyloxy substitution) as well as at N1, N3, and N7 (small alkyl residues), thereby improving affinity and selectivity for the A2A AR. The compounds were characterized by radioligand binding assays, and our study resulted in the development of the potent A2A AR ligands including 8-((6-chloro-2-fluoro-3-methoxybenzyl)amino)-1-ethyl-3,7-dimethyl-3,7-dihydro-1H-purine-2,6-dione (12d; Ki human A2AAR: 68.5 nM) and 8-((2-chlorobenzyl)amino)-1-ethyl-3,7-dimethyl-3,7-dihydro-1H-purine-2,6-dione (12h; Ki human A2AAR: 71.1 nM). Moreover, dual A1/A2AAR ligands were identified in the group of 1,3-diethyl-7-methylxanthine derivatives. Compound 14b displayed Ki values of 52.2 nM for the A1AR and 167 nM for the A2AAR. Selected A2AAR ligands were further evaluated as inactive for inhibition of monoamine oxidase A, B and isoforms of phosphodiesterase-4B1, -10A, which represent classical targets for xanthine derivatives. Therefore, the developed 8-benzylaminoxanthine scaffold seems to be highly selective for AR activity and relevant for potent and selective A2A ligands. Compound 12d with high selectivity for ARs, especially for the A2AAR subtype, evaluated in animal models of inflammation has shown anti-inflammatory activity. Investigated compounds were found to display high selectivity and may therefore be of high interest for further development as drugs for treating cancer or neurodegenerative diseases.
Collapse
Affiliation(s)
- Michał Załuski
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30688 Kraków, Poland
| | - Jakub Schabikowski
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30688 Kraków, Poland
| | - Piotr Jaśko
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30688 Kraków, Poland
| | - Adrian Bryła
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30688 Kraków, Poland
| | - Agnieszka Olejarz-Maciej
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30688 Kraków, Poland
| | - Maria Kaleta
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30688 Kraków, Poland
| | - Monika Głuch-Lutwin
- Department of Pharmacobiology, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30688 Kraków, Poland
| | - Andreas Brockmann
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Sonja Hinz
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Małgorzata Zygmunt
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30688 Kraków, Poland
| | - Kamil Kuder
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30688 Kraków, Poland
| | - Gniewomir Latacz
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30688 Kraków, Poland
| | - Christin Vielmuth
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Christa E Müller
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Katarzyna Kieć-Kononowicz
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30688 Kraków, Poland.
| |
Collapse
|
30
|
Jamwal S, Mittal A, Kumar P, Alhayani DM, Al-Aboudi A. Therapeutic Potential of Agonists and Antagonists of A1, A2a, A2b and A3 Adenosine Receptors. Curr Pharm Des 2020; 25:2892-2905. [PMID: 31333104 DOI: 10.2174/1381612825666190716112319] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Accepted: 07/04/2019] [Indexed: 02/04/2023]
Abstract
Adenosine is a naturally occurring nucleoside and an essential component of the energy production and utilization systems of the body. Adenosine is formed by the degradation of adenosine-triphosphate (ATP) during energy-consuming processes. Adenosine regulates numerous physiological processes through activation of four subtypes of G-protein coupled membrane receptors viz. A1, A2A, A2B and A3. Its physiological importance depends on the affinity of these receptors and the extracellular concentrations reached. ATP acts as a neurotransmitter in both peripheral and central nervous systems. In the peripheral nervous system, ATP is involved in chemical transmission in sensory and autonomic ganglia, whereas in central nervous system, ATP, released from synaptic terminals, induces fast excitatory postsynaptic currents. ATP provides the energetics for all muscle movements, heart beats, nerve signals and chemical reactions inside the body. Adenosine has been traditionally considered an inhibitor of neuronal activity and a regulator of cerebral blood flow. Since adenosine is neuroprotective against excitotoxic and metabolic dysfunctions observed in neurological and ocular diseases, the search for adenosinerelated drugs regulating adenosine transporters and receptors can be important for advancement of therapeutic strategies against these diseases. This review will summarize the therapeutic potential and recent SAR and pharmacology of adenosine and its receptor agonists and antagonists.
Collapse
Affiliation(s)
- Sumit Jamwal
- School of Pharmacy and Emerging Sciences, Baddi University of Emerging Sciences and Technologies, Baddi, India
| | - Ashish Mittal
- Department of Pharmaceutical Sciences, M.R.S. Punjab Technical University, Bathinda, Punjab, India
| | - Puneet Kumar
- Department of Pharmaceutical Sciences, M.R.S. Punjab Technical University, Bathinda, Punjab, India
| | - Dana M Alhayani
- Faculty of Pharmacy, Philadelphia University, PO Box - 1, 19392, Amman, Jordan
| | - Amal Al-Aboudi
- Faculty of Science, The University of Jordan, Amman, 11942, Jordan
| |
Collapse
|
31
|
Borah P, Deka S, Mailavaram RP, Deb PK. P1 Receptor Agonists/Antagonists in Clinical Trials - Potential Drug Candidates of the Future. Curr Pharm Des 2020; 25:2792-2807. [PMID: 31333097 DOI: 10.2174/1381612825666190716111245] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 07/03/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND Adenosine mediates various physiological and pathological conditions by acting on its four P1 receptors (A1, A2A, A2B and A3 receptors). Omnipresence of P1 receptors and their activation, exert a wide range of biological activities. Thus, its modulation is implicated in various disorders like Parkinson's disease, asthma, cardiovascular disorders, cancer etc. Hence these receptors have become an interesting target for the researchers to develop potential therapeutic agents. Number of molecules were designed and developed in the past few years and evaluated for their efficacy in various disease conditions. OBJECTIVE The main objective is to provide an overview of new chemical entities which have crossed preclinical studies and reached clinical trials stage following their current status and future prospective. METHODS In this review we discuss current status of the drug candidates which have undergone clinical trials and their prospects. RESULTS Many chemical entities targeting various subtypes of P1 receptors are patented; twenty of them have crossed preclinical studies and reached clinical trials stage. Two of them viz adenosine and regadenoson are approved by the Food and Drug Administration. CONCLUSION This review is an attempt to highlight the current status, progress and probable future of P1 receptor ligands which are under clinical trials as promising novel therapeutic agents and the direction in which research should proceed with a view to come out with novel therapeutic agents.
Collapse
Affiliation(s)
- Pobitra Borah
- Pratiksha Institute of Pharmaceutical Sciences, Panikhaiti, Chandrapur Road, Guwahati, Assam, India
| | - Satyendra Deka
- Pratiksha Institute of Pharmaceutical Sciences, Panikhaiti, Chandrapur Road, Guwahati, Assam, India
| | - Raghu Prasad Mailavaram
- Department of Pharmaceutical Chemistry, Shri Vishnu College of Pharmacy, Vishnupur (Affiliated to Andhra University), Bhimavaram, W.G. Dist., AP, India
| | - Pran Kishore Deb
- Faculty of Pharmacy, Philadelphia University, Amman, PO Box-1, 19392, Jordan
| |
Collapse
|
32
|
Tarnowski M, Tkacz M, Piotrowska K, Zgutka K, Pawlik A. Differential effect of adenosine on rhabdomyosarcoma migration and proliferation. Arch Med Sci 2020; 16:414-427. [PMID: 32190153 PMCID: PMC7069424 DOI: 10.5114/aoms.2018.75506] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 09/03/2017] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION Adenosine and its receptors are involved deeply in the regulation of tumour biology. Purine nucleotides are released from stressed cells in states of hypoxia or radiochemotherapy-induced cell damage. Adenosine exerts its effect through the P1 family of selective receptors. The purpose of the study was to evaluate the exact role of extracellular role on biology of Rhabdomyosarcoma (RMS) cells. MATERIAL AND METHODS Series of in vitro studies accompanied by immunohistochemical, RQ-PCR and shRNA methods have characterised adenosine receptor expression on Rhabdomyosarcoma cell lines, normal skeletal muscle and effect of adenosine on Rhabdomyosarcoma growth and migration. RESULTS Extracellular adenosine (highest at 50 μM, p < 0.05) and AMP (highest at 300 μM, p < 0.05) markedly enhanced chemotaxis in the Boyden chamber assay The reaction is mostly governed by the A1 receptor, which is greatly overexpressed in Rhabdomyosarcoma as compared with normal skeletal muscle. Cell migration induced by adenosine and AMP is blocked by pertussis toxin, phospholipase C and MAP kinase inhibitor, which demonstrates the importance of these signalling pathways. High doses of adenosine have a detrimental effect on cellular proliferation, in a receptor-independent manner (≥ 500 μM; p < 0.05). The blockage of adenosine transporter by dipyridamole abolishes this effect, indicating involvement of an intrinsic pathway. Further increase of adenosine concentration, induced by deaminase inhibitors, augment the effect. CONCLUSIONS Our results suggest that adenosine and AMP trigger cell migration by binding to P1 receptors and directing cancer cells to the sites of hypoxia or cellular damage. Specifically by A1 receptor which is overexpressed in RMS.
Collapse
Affiliation(s)
- Maciej Tarnowski
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | - Marta Tkacz
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | | | - Katarzyna Zgutka
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| |
Collapse
|
33
|
Adenosine Receptor Agonists Exhibit Anti-Platelet Effects and the Potential to Overcome Resistance to P2Y 12 Receptor Antagonists. Molecules 2019; 25:molecules25010130. [PMID: 31905703 PMCID: PMC6982709 DOI: 10.3390/molecules25010130] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 12/16/2019] [Accepted: 12/25/2019] [Indexed: 11/30/2022] Open
Abstract
Large inter-individual variation in platelet response to endogenous agonists and pharmacological agents, including resistance to antiplatelet therapy, prompts a search for novel platelet inhibitors and development new antithrombotic strategies. The present in vitro study evaluates the beneficial effects of three adenosine receptor (AR) agonists (regadenoson, LUF 5835 and NECA), different in terms of their selectivity for platelet adenosine receptors, when used alone and in combination with P2Y12 inhibitors, such as cangrelor or prasugrel metabolite. The anti-platelet effects of AR agonists were evaluated in healthy subjects (in the whole group and after stratification of individuals into high- and low-responders to P2Y12 inhibitors), using whole blood techniques, under flow (thrombus formation) and static conditions (study of platelet activation and aggregation). Compared to P2Y12 antagonists, AR agonists were much less or not effective under static conditions, but demonstrated similar antiplatelet activity in flow. In most cases, AR agonists significantly enhanced the anti-platelet effect of P2Y12 antagonists, despite possessing different selectivity profiles and antiplatelet activities. Importantly, their inhibitory effects in combination with P2Y12 antagonists were similar in high- and low-responders to P2Y12 inhibitors. In conclusion, a combination of anti-platelet agents acting via the P1 and P2 purinergic receptors represents a promising alternative to existing antithrombotic therapy.
Collapse
|
34
|
Wolska N, Rozalski M. Blood Platelet Adenosine Receptors as Potential Targets for Anti-Platelet Therapy. Int J Mol Sci 2019; 20:ijms20215475. [PMID: 31684173 PMCID: PMC6862090 DOI: 10.3390/ijms20215475] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/31/2019] [Accepted: 11/01/2019] [Indexed: 12/21/2022] Open
Abstract
Adenosine receptors are a subfamily of highly-conserved G-protein coupled receptors. They are found in the membranes of various human cells and play many physiological functions. Blood platelets express two (A2A and A2B) of the four known adenosine receptor subtypes (A1, A2A, A2B, and A3). Agonization of these receptors results in an enhanced intracellular cAMP and the inhibition of platelet activation and aggregation. Therefore, adenosine receptors A2A and A2B could be targets for anti-platelet therapy, especially under circumstances when classic therapy based on antagonizing the purinergic receptor P2Y12 is insufficient or problematic. Apart from adenosine, there is a group of synthetic, selective, longer-lasting agonists of A2A and A2B receptors reported in the literature. This group includes agonists with good selectivity for A2A or A2B receptors, as well as non-selective compounds that activate more than one type of adenosine receptor. Chemically, most A2A and A2B adenosine receptor agonists are adenosine analogues, with either adenine or ribose substituted by single or multiple foreign substituents. However, a group of non-adenosine derivative agonists has also been described. This review aims to systematically describe known agonists of A2A and A2B receptors and review the available literature data on their effects on platelet function.
Collapse
Affiliation(s)
- Nina Wolska
- Department of Haemostasis and Haemostatic Disorders, Chair of Biomedical Science, Medical University of Lodz, 92-215 Lodz, Poland.
| | - Marcin Rozalski
- Department of Haemostasis and Haemostatic Disorders, Chair of Biomedical Science, Medical University of Lodz, 92-215 Lodz, Poland.
| |
Collapse
|
35
|
Marwein S, Mishra B, De UC, Acharya PC. Recent Progress of Adenosine Receptor Modulators in the Development of Anticancer Chemotherapeutic Agents. Curr Pharm Des 2019; 25:2842-2858. [DOI: 10.2174/1381612825666190716141851] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 07/01/2019] [Indexed: 01/12/2023]
Abstract
Increased risks of peripheral toxicity and undesired adverse effects associated with chemotherapeutic
agents are the major medical hurdles in cancer treatment that worsen the quality of life of cancer patients. Although
several novel and target-specific anticancer agents have been discovered in the recent past, none of them
have proved to be effective in the management of metastatic tumor. Therefore, there is a continuous effort for the
discovery of safer and effective cancer chemotherapeutic agent. Adenosine receptors have been identified as an
important target to combat cancer because of their inherent role in the antitumor process. The antitumor property
of the adenosine receptor is primarily attributed to their inherited immune response against the tumors. These
findings have opened a new chapter in the anticancer drug discovery through adenosine receptor-mediated immunomodulation.
This review broadly outlines the biological mechanism of adenosine receptors in mediating the
selective cytotoxicity as well as the discovery of various classes of adenosine receptor modulators in the effective
management of solid tumors.
Collapse
Affiliation(s)
- Sarapynbiang Marwein
- Department of Pharmacy, Tripura University (A Central University), Suryamaninagar-799022, Tripura (W), India
| | - Bijayashree Mishra
- Department of Chemistry, Tripura University (A Central University), Suryamaninagar-799022, Tripura (W), India
| | - Utpal C. De
- Department of Chemistry, Tripura University (A Central University), Suryamaninagar-799022, Tripura (W), India
| | - Pratap C. Acharya
- Department of Pharmacy, Tripura University (A Central University), Suryamaninagar-799022, Tripura (W), India
| |
Collapse
|
36
|
Gorain B, Choudhury H, Yee GS, Bhattamisra SK. Adenosine Receptors as Novel Targets for the Treatment of Various Cancers. Curr Pharm Des 2019; 25:2828-2841. [DOI: 10.2174/1381612825666190716102037] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 07/01/2019] [Indexed: 01/28/2023]
Abstract
Adenosine is a ubiquitous signaling nucleoside molecule, released from different cells within the body
to act on vasculature and immunoescape. The physiological action on the proliferation of tumour cell has been
reported by the presence of high concentration of adenosine within the tumour microenvironment, which results
in the progression of the tumour, even leading to metastases. The activity of adenosine exclusively depends upon
the interaction with four subtypes of heterodimeric G-protein-coupled adenosine receptors (AR), A1, A2A, A2B,
and A3-ARs on the cell surface. Research evidence supports that the activation of those receptors via specific
agonist or antagonist can modulate the proliferation of tumour cells. The first category of AR, A1 is known to play
an antitumour activity via tumour-associated microglial cells to prevent the development of glioblastomas.
A2AAR are found in melanoma, lung, and breast cancer cells, where tumour proliferation is stimulated due to
inhibition of the immune response via inhibition of natural killer cells cytotoxicity, T cell activity, and tumourspecific
CD4+/CD8+ activity. Alternatively, A2BAR helps in the development of tumour upon activation via
upregulation of angiogenin factor in the microvascular endothelial cells, inhibition of MAPK and ERK 1/2 phosphorylation
activity. Lastly, A3AR is expressed in low levels in normal cells whereas the expression is upregulated
in tumour cells, however, agonists to this receptor inhibit tumour proliferation through modulation of Wnt
and NF-κB signaling pathways. Several researchers are in search for potential agents to modulate the overexpressed
ARs to control cancer. Active components of A2AAR antagonists and A3AR agonists have already entered
in Phase-I clinical research to prove their safety in human. This review focused on novel research targets towards
the prevention of cancer progression through stimulation of the overexpressed ARs with the hope to protect lives
and advance human health.
Collapse
Affiliation(s)
- Bapi Gorain
- School of Pharmacy, Faculty of Health and Medical Science, Taylor’s University, Subang Jaya, Selangor, Malaysia
| | - Hira Choudhury
- Department of Pharmaceutical Technology, School of Pharmacy, International Medical University, Bukit Jalil 57000, Kuala Lumpur, Malaysia
| | - Gan Sook Yee
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil 57000, Kuala Lumpur, Malaysia
| | - Subrat Kumar Bhattamisra
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil 57000, Kuala Lumpur, Malaysia
| |
Collapse
|
37
|
Cruz FF, Pereira TCB, Altenhofen S, da Costa KM, Bogo MR, Bonan CD, Morrone FB. Characterization of the adenosinergic system in a zebrafish embryo radiotherapy model. Comp Biochem Physiol C Toxicol Pharmacol 2019; 224:108572. [PMID: 31306805 DOI: 10.1016/j.cbpc.2019.108572] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 06/27/2019] [Accepted: 07/09/2019] [Indexed: 12/15/2022]
Abstract
Adenosine is a nucleoside that acts as a signaling molecule by activating P1 purinergic receptors (A1, A2A, A2B and A3). This activation is involved in immune responses, inflammation, and tissue remodeling and tumor progression. Gamma rays are a type of ionizing radiation widely adopted in radiotherapy of tumors. Although it brings benefits to the success of the therapeutic scheme, it can trigger cellular damages, inducing a perpetual inflammatory response that culminates in adverse effects and severe toxicity. Our study aims to characterize the adenosinergic system in a zebrafish embryo radiotherapy model, relating the adenosine signaling to the changes elicited by radiation exposure. To standardize the radiotherapy procedure, we established a toxicological profile after exposure. Zebrafish were irradiated with different doses of gamma rays (2, 5, 10, 15 and 20 Gy) at 24 hpf. Survival, hatching rate, heartbeats, locomotor activity and morphological changes were determined during embryos development. Although without significant difference in survival, gamma-irradiated embryos had their heartbeats increased and presented decreased hatching time, changes in locomotor activity and important morphological alterations. The exposure to 10 Gy disrupted the ecto-5'-nucleotidase/CD73 and adenosine deaminase/ADA enzymatic activity, impairing adenosine metabolism. We also demonstrated that radiation decreased A2B receptor gene expression, suggesting the involvement of extracellular adenosine in the changes prompted by radiotherapy. Our results indicate that the components of the adenosinergic system may be potential targets to improve radiotherapy and manage the tissue damage and toxicity of ionizing radiation.
Collapse
Affiliation(s)
- Fernanda Fernandes Cruz
- Programa de Pós-Graduação em Biologia Celular e Molecular, Escola de Ciências, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil; Laboratório de Farmacologia Aplicada, Escola de Ciências da Saúde, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Talita Carneiro Brandão Pereira
- Programa de Pós-Graduação em Biologia Celular e Molecular, Escola de Ciências, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil; Laboratório de Biologia Genômica e Molecular, Escola de Ciências, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Stefani Altenhofen
- Programa de Pós-Graduação em Medicina e Ciências da Saúde, Escola de Medicina, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil; Laboratório de Neuroquímica e Psicofarmacologia, Escola de Ciências, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Kesiane Mayra da Costa
- Programa de Pós-Graduação em Medicina e Ciências da Saúde, Escola de Medicina, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil; Laboratório de Biologia Genômica e Molecular, Escola de Ciências, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Maurício Reis Bogo
- Programa de Pós-Graduação em Biologia Celular e Molecular, Escola de Ciências, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil; Programa de Pós-Graduação em Medicina e Ciências da Saúde, Escola de Medicina, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil; Laboratório de Biologia Genômica e Molecular, Escola de Ciências, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Carla Denise Bonan
- Programa de Pós-Graduação em Biologia Celular e Molecular, Escola de Ciências, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil; Programa de Pós-Graduação em Medicina e Ciências da Saúde, Escola de Medicina, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil; Laboratório de Neuroquímica e Psicofarmacologia, Escola de Ciências, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Fernanda Bueno Morrone
- Programa de Pós-Graduação em Biologia Celular e Molecular, Escola de Ciências, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil; Laboratório de Farmacologia Aplicada, Escola de Ciências da Saúde, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil; Programa de Pós-Graduação em Medicina e Ciências da Saúde, Escola de Medicina, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| |
Collapse
|
38
|
Borges-Martins VPP, Ferreira DDP, Souto AC, Oliveira Neto JG, Pereira-Figueiredo D, da Costa Calaza K, de Jesus Oliveira K, Manhães AC, de Melo Reis RA, Kubrusly RCC. Caffeine regulates GABA transport via A 1R blockade and cAMP signaling. Neurochem Int 2019; 131:104550. [PMID: 31563462 DOI: 10.1016/j.neuint.2019.104550] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 09/18/2019] [Accepted: 09/19/2019] [Indexed: 01/06/2023]
Abstract
Caffeine is the most consumed psychostimulant drug in the world, acting as a non-selective antagonist of adenosine receptors A1R and A2AR, which are widely expressed in retinal layers. We have previously shown that caffeine, when administered acutely, acts on A1R to potentiate the NMDA receptor-induced GABA release. Now we asked if long-term caffeine exposure also modifies GABA uptake in the avian retina and which mechanisms are involved in this process. Chicken embryos aged E11 were injected with a single dose of caffeine (30 mg/kg) in the air chamber. Retinas were dissected on E15 for ex vivo neurochemical assays. Our results showed that [3H]-GABA uptake was dependent on Na+ and blocked at 4 °C or by NO-711 and caffeine. This decrease was observed after 60 min of [3H]-GABA uptake assay at E15, which is accompanied by an increase in [3H]-GABA release. Caffeine increased the protein levels of A1R without altering ADORA1 mRNA and was devoid of effects on A2AR density or ADORA2A mRNA levels. The decrease of GABA uptake promoted by caffeine was reverted by A1R activation with N6-cyclohexyl adenosine (CHA) but not by A2AR activation with CGS 21680. Caffeine exposure increased cAMP levels and GAT-1 protein levels, which was evenly expressed between E11-E15. As expected, we observed an increase of GABA containing amacrine cells and processes in the IPL, also, cAMP pathway blockage by H-89 decreased caffeine mediated [3H]-GABA uptake. Our data support the idea that chronic injection of caffeine alters GABA transport via A1R during retinal development and that the cAMP/PKA pathway plays an important role in the regulation of GAT-1 function.
Collapse
Affiliation(s)
| | - Danielle Dias Pinto Ferreira
- Laboratório de Doenças Neurodegenerativas, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Arthur Cardoso Souto
- Laboratório de Neurofarmacologia, Departamento de Fisiologia e Farmacologia, Universidade Federal Fluminense, Niterói, Brazil.
| | - Jessika Geisebel Oliveira Neto
- Laboratório de Fisiologia Endócrina e Metabologia, Departamento de Fisiologia e Farmacologia, Universidade Federal Fluminense, Niterói, Brazil.
| | - Danniel Pereira-Figueiredo
- Laboratório de Neurobiologia da Retina, Departmento de Neurobiologia, Instituto de Biologia, Universidade Federal Fluminense, Niterói, Brazil.
| | - Karin da Costa Calaza
- Laboratório de Neurobiologia da Retina, Departmento de Neurobiologia, Instituto de Biologia, Universidade Federal Fluminense, Niterói, Brazil.
| | - Karen de Jesus Oliveira
- Laboratório de Fisiologia Endócrina e Metabologia, Departamento de Fisiologia e Farmacologia, Universidade Federal Fluminense, Niterói, Brazil.
| | - Alex Christian Manhães
- Laboratório de Neurofisiologia, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Ricardo Augusto de Melo Reis
- Laboratório de Neuroquímica, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Regina Célia Cussa Kubrusly
- Laboratório de Neurofarmacologia, Departamento de Fisiologia e Farmacologia, Universidade Federal Fluminense, Niterói, Brazil.
| |
Collapse
|
39
|
Bonyanian Z, Walker M, Du Toit E, Rose'Meyer RB. Multiple adenosine receptor subtypes stimulate wound healing in human EA.hy926 endothelial cells. Purinergic Signal 2019; 15:357-366. [PMID: 31254200 DOI: 10.1007/s11302-019-09668-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 06/20/2019] [Indexed: 10/26/2022] Open
Abstract
Wound healing is an important outcome of tissue damage and can be stimulated by adenosine released from cells during events such as tissue injury, ischaemia or tumour growth. The aim of this research was to determine the potency and efficacy of adenosine A1, A2A and A2B receptor agonists on the rate of wound healing and cell proliferation in human EA.hy926 endothelial cells. Real-time PCR data showed that only adenosine A1, A2A and A2B receptor mRNA were expressed in this cell line. All three adenosine receptor agonists, CPA, CGS21680 and NECA, significantly increased the rate of wound healing in human EAhy926 endothelial cells with the following order of potency CGS21680>CPA>NECA and efficacy CPA>NECA>CGS21680. The selective adenosine A1, A2A and A2B receptor antagonists, DPCPX, ZM241385 and MRS1754 (all at 10 nM), reversed the effects of their respective agonists. EAhy926 endothelial cell proliferation was also significantly increased with the adenosine A1 and A2B receptor agonists, CPA and NECA. Western blot analysis demonstrated that adenosine A2A and A1 receptor protein levels were highly expressed compared with the adenosine A2B receptors in the EAhy926 endothelial cell lines. While all three adenosine A1, A2A and A2B receptor subtypes contribute to cell proliferation and wound healing in human EAhy926 endothelial cells, treatments selectively targeting receptor subtypes may further enhance wound healing.
Collapse
Affiliation(s)
- Zeinab Bonyanian
- School of Medical Sciences, Griffith University, Gold Coast Campus Southport, Queensland, 4122, Australia
| | - Matthew Walker
- School of Medical Sciences, Griffith University, Gold Coast Campus Southport, Queensland, 4122, Australia
| | - Eugene Du Toit
- School of Medical Sciences, Griffith University, Gold Coast Campus Southport, Queensland, 4122, Australia
| | - Roselyn B Rose'Meyer
- School of Medical Sciences, Griffith University, Gold Coast Campus Southport, Queensland, 4122, Australia.
| |
Collapse
|
40
|
Design, synthesis and biological evaluation of 2-hydrazinyladenosine derivatives as A 2A adenosine receptor ligands. Eur J Med Chem 2019; 179:310-324. [PMID: 31255928 DOI: 10.1016/j.ejmech.2019.06.050] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 06/10/2019] [Accepted: 06/17/2019] [Indexed: 11/22/2022]
Abstract
To obtain potential A2A adenosine receptor agonists, a series of 2-hydrazinyladenosine derivatives were synthesized and assayed for adenosine receptors activity using radioligand binding activity assays. The binding activity of the subtypes was examined, and the structure-activity relationship of this class of compounds at the A2A receptor was investigated. A fragment-based computer-aided design method was used to modify the 2-position side chain structures with different structural fragments, and the newly generated molecules were docked to the A2A receptor to assess scoring and screening activity. To synthesize compounds with better scoring activity, the newly synthesized compounds were tested for in vitro receptor binding activity. 2-Hydrazinyladenosine derivatives of 32 new structural types were designed and synthesized, with the most potent adenosine derivative 23 exhibiting a Ki value of 1.8 nM for A2AAR and significant selectivity for the A2A receptor compared to the A1 receptor. In addition to, compound 23, 24, 30, 31, and 42 also exhibited potent A2A receptor selectivity, with Ki values for the A2A receptor of 6.4, 20, 67 and 6.3 nM, respectively. We also found that compound 35 has a high A1 receptor selectivity, with a Ki value for the A1 receptor of 4.5 nM. Further functional assays also demonstrated that these compounds have potent A2A receptor agonist activity. The study shows the applicability of an in silico fragment-based molecular design for rational lead optimization in A2AAR.
Collapse
|
41
|
Singh BL, Chen L, Cai H, Shi H, Wang Y, Yu C, Chen X, Han X, Cai X. Activation of adenosine A2a receptor accelerates and A2a receptor antagonist reduces intermittent hypoxia induced PC12 cell injury via PKC-KATP pathway. Brain Res Bull 2019; 150:118-126. [PMID: 31129168 DOI: 10.1016/j.brainresbull.2019.05.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 05/19/2019] [Accepted: 05/21/2019] [Indexed: 02/07/2023]
Abstract
Obstructive sleep apnea hypopnea syndrome (OSAHS) is associated with multiple system diseases. Neurocognitive dysfunction resulting from central nervous system complications has been reported, especially in children with OSAHS. Chronic intermittent hypoxia is accepted to be the major pathophysiological mechanism of OSAHS. Adenosine plays an important role in cellular function via interactions with its receptors. A2a receptor has been recognized as a factor involved in neuroprotection. However, the role of adenosine A2a receptor in intermittent hypoxia induced cellular injury is not completely understood. In this study, we aim to investigate the underlying mechanisms of A2a receptor mediated cellular damage caused by intermittent hypoxia in PC12 cells. We found that activated A2a receptor by CGS21680 decreased cellular viability, increased PKC as well as ATP-sensitive potassium channel (KATP) subunits expression Kir6.2 and SUR1. Inhibition of A2a receptor by SCH58261 increased cellular viability, suppressed PKC and SUR1 expression level, ultimately showing a protective role in PC12 cells. Moreover, we observed that CHE, which is an antagonist of PKC, downregulated Kir6.2 and SUR1 expression and increased cellular viability. Additionally, we found that A2a receptor activation induced cell injury was associated with increased Cleaved-Caspase 3 expression, which can be decreased by inhibition of A2a receptor or PKC. In conclusion, our findings indicate that A2a receptor induced KATP expression by PKC activation and plays a role in accelerating PC12 cells injury induced by intermittent hypoxia exposure via A2a-PKC-KATP signal pathway mediated apoptosis.
Collapse
Affiliation(s)
- Brett Lyndall Singh
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 109 Xueyuan Western Road, Wenzhou, Zhejiang, 325027, PR China; The Second School Of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Liya Chen
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 109 Xueyuan Western Road, Wenzhou, Zhejiang, 325027, PR China; The Second School Of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Huilin Cai
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 109 Xueyuan Western Road, Wenzhou, Zhejiang, 325027, PR China; The Second School Of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Hua Shi
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 109 Xueyuan Western Road, Wenzhou, Zhejiang, 325027, PR China; The Second School Of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Yueyuan Wang
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 109 Xueyuan Western Road, Wenzhou, Zhejiang, 325027, PR China; The Second School Of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Chenyi Yu
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 109 Xueyuan Western Road, Wenzhou, Zhejiang, 325027, PR China
| | - Xu Chen
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 109 Xueyuan Western Road, Wenzhou, Zhejiang, 325027, PR China
| | - Xinru Han
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 109 Xueyuan Western Road, Wenzhou, Zhejiang, 325027, PR China; The Second School Of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Xiaohong Cai
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 109 Xueyuan Western Road, Wenzhou, Zhejiang, 325027, PR China; The Second School Of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, PR China.
| |
Collapse
|
42
|
Zhang D, Ma J, Meng X, Xu Z, Zhang J, Fang Y, Guo Y. Electrochemical aptamer-based microsensor for real-time monitoring of adenosine in vivo. Anal Chim Acta 2019; 1076:55-63. [PMID: 31203964 DOI: 10.1016/j.aca.2019.05.035] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 05/07/2019] [Accepted: 05/14/2019] [Indexed: 11/24/2022]
Abstract
In this work, an implantable and minimally invasive micro-aptasensor for adenosine monitoring in vivo, based on flexible integrated electrodes, was developed. Firstly the sensor was made by the modification of a needle-type electrode with reduced graphene oxide and gold nanoclusters (rGO-AuNCs) using two-step electrodeposition. Secondly Sulfhydryl-terminated capture probe (ssDNA1) was immobilized on rGO-AuNCs modified electrode surface by self-assembly, and then it was hybridized with adenosine aptamer (ssDNA2). Lastly methylene blue (MB) as an electrochemical indicator was adsorbed on the aptamer through specific interaction of MB with guanine base. The peak current of MB decreased linearly with increasing adenosine concentration due to the formation of aptamer-adenosine complex and displacement of the aptamer from the modified electrode surface. The sensor showed a low detection limit of 0.1 nM with signal-to-noise ratio equal to 3 as well as a wide linear response range (0.1 nM-1 mM) in vitro. Also, a high selectivity was demonstrated for adenosine in relation to uridine, guanosine, and cytidine. Experiments in vivo demonstrated fast responses for a range of adenosine concentrations. This work demonstrates a promising path for implantable devices for the determination of biomolecules in vivo, thus allowing for health tests, detection of infectious diseases, and other medical conditions.
Collapse
Affiliation(s)
- Di Zhang
- Binhai Industrial Technology Research Institute of Zhejiang University, Tianjin, 300301, PR China
| | - Jiajia Ma
- Research Center of Experimental Acupuncture Science, College of Acumox and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China
| | - Xiangwen Meng
- Research Center of Experimental Acupuncture Science, College of Acumox and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China
| | - Zhifang Xu
- Research Center of Experimental Acupuncture Science, College of Acumox and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China
| | - Jian Zhang
- Research Center of Experimental Acupuncture Science, College of Acumox and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China
| | - Yuxin Fang
- Research Center of Experimental Acupuncture Science, College of Acumox and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China.
| | - Yi Guo
- Research Center of Experimental Acupuncture Science, College of Acumox and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China; College of Chinese Medical, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China.
| |
Collapse
|
43
|
Li X, Liang D, Shao H, Born WK, Kaplan HJ, Sun D. Adenosine receptor activation in the Th17 autoimmune responses of experimental autoimmune uveitis. Cell Immunol 2019; 339:24-28. [DOI: 10.1016/j.cellimm.2018.09.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 09/07/2018] [Accepted: 09/18/2018] [Indexed: 01/16/2023]
|
44
|
Involvement of A2B adenosine receptors as anti-inflammatory in gestational diabesity. Mol Aspects Med 2019; 66:31-39. [DOI: 10.1016/j.mam.2019.01.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Revised: 11/23/2018] [Accepted: 01/17/2019] [Indexed: 02/07/2023]
|
45
|
Babich V, Vadnagara K, Di Sole F. Adenosine A 2A receptor blocks the A 1 receptor inhibition of renal Na + transport and oxygen consumption. J Cell Physiol 2019; 234:13917-13930. [PMID: 30633335 DOI: 10.1002/jcp.28074] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Accepted: 12/18/2018] [Indexed: 11/11/2022]
Abstract
A high renal oxygen (O2 ) need is primarily associated with the renal tubular O2 consumption (VO2 ) necessary for a high rate of sodium (Na+ ) transport. Limited O2 availability leads to increased levels of adenosine, which regulates the kidney via activation of both A1 and A2A adenosine receptors (A1R and A2AR, respectively). The relative contributions of A1R and A2AR to the regulation of renal Na+ transport and VO2 have not been determined. We demonstrated that A1R activation has a dose-dependent biphasic effect on both renal Na+ /H+ exchanger-3 (NHE3), a major player in Na+ transport, and VO2 . Here, we report concentration-dependent effects of adenosine: less than 5 × 10-7 M adenosine-stimulated NHE3 activity; between 5 × 10-7 M and 10-5 M adenosine-inhibited NHE3 activity; and greater than 10-5 M adenosine reversed the change in NHE3 activity (returned to baseline). A1R activation mediated the activation and inhibition of NHE3 activity, whereas 10-4 M adenosine had no effect on the NHE3 activity due to A2AR activation. The following occurred when A1R and A2AR were activated: (a) Blockade of the A2AR receptor restored the NHE3 inhibition mediated by A1R activation, (b) the NHE-dependent effect on VO2 mediated by A1R activation became NHE independent, and (c) A2AR bound to A1R. In summary, A1R affects VO2 via NHE-dependent mechanisms, whereas A2AR acts via NHE-independent mechanisms. When both A1R and A2AR are activated, the A2AR effect on NHE3 and VO2 predominates, possibly via an A1R-A2AR protein interaction. A2AR-A1R heterodimerization is proposed as the molecular mechanism enabling the NHE-independent control of renal VO2 .
Collapse
Affiliation(s)
- Victor Babich
- Department of Physiology and Pharmacology, Des Moines University, Des Moines, Iowa.,School of Liberal Arts and Sciences, Mercy College of Health Sciences, Des Moines, Iowa.,Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas.,Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Komal Vadnagara
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Francesca Di Sole
- Department of Physiology and Pharmacology, Des Moines University, Des Moines, Iowa.,Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas.,Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
46
|
Adenosine receptor agonists deepen the inhibition of platelet aggregation by P2Y 12 antagonists. Vascul Pharmacol 2018; 113:47-56. [PMID: 30471364 DOI: 10.1016/j.vph.2018.11.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 10/01/2018] [Accepted: 11/18/2018] [Indexed: 11/24/2022]
Abstract
Several adenosine receptor (AR) agonists have been shown in the past to possess anti-platelet potential; however, the adjunctive role of AR agonists in anti-platelet therapy with the use of P2Y12 receptor inhibitors has not been elucidated so far. This in vitro aggregation-based study investigates whether the inhibition of platelet function mediated by cangrelor or prasugrel metabolite can be potentiated by AR agonists. It evaluates the effect of non-selective (2-chloroadenosine), A2A-selective (UK 432097, MRE 0094, PSB 0777) and A2B-selective AR agonists (BAY 60-6583) on platelet function in relation to their toxicity, specificity towards adenosine receptor subtypes, structure and solubility. UK 432097, 2-chloroadenosine, MRE 0094 and PSB 0777 were found to be more or less potent inhibitors of ADP-induced platelet aggregation when acting alone, and that they remained non-cytotoxic to the cells. These AR agonists were also effective in the potentiation of the effects exerted by P2Y12 antagonists. Considering the estimated IC50 value, UK 432097, showing a relatively high binding affinity to the A2A adenosine receptor, has been identified as the most potent anti-aggregatory agent. This compound diminished platelet aggregation at nanomolar concentrations and further augmented platelet inhibition by P2Y12 antagonists by approx. 60% (P < .01). Our results indicate the importance of adenosine receptors as therapeutic targets and point out challenges and potential benefits of therapeutic use of a combined therapy of P2Y12 antagonist and AR agonist in cardioprotection. Our comparative analysis of the effects of AR agonists on platelet response in plasma and whole blood may indirectly suggest that other blood morphology elements contribute little to the inhibition of platelet function by AR agonists.
Collapse
|
47
|
Poleszak E, Szopa A, Bogatko K, Wyska E, Wośko S, Świąder K, Doboszewska U, Wlaź A, Wróbel A, Wlaź P, Serefko A. Antidepressant-Like Activity of Typical Antidepressant Drugs in the Forced Swim Test and Tail Suspension Test in Mice Is Augmented by DMPX, an Adenosine A 2A Receptor Antagonist. Neurotox Res 2018; 35:344-352. [PMID: 30267268 PMCID: PMC6331646 DOI: 10.1007/s12640-018-9959-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 08/31/2018] [Accepted: 09/11/2018] [Indexed: 01/06/2023]
Abstract
Unsatisfactory therapeutic effects of currently used antidepressants force to search for new pharmacological treatment strategies. Recent research points to the relationship between depressive disorders and the adenosinergic system. Therefore, the main goal of our studies was to evaluate the effects of DMPX (3 mg/kg, i.p.), which possesses selectivity for adenosine A2A receptors versus A1 receptors, on the activity of imipramine (15 mg/kg, i.p.), escitalopram (2.5 mg/kg, i.p.), and reboxetine (2 mg/kg, i.p.) given in subtherapeutic doses. The studies carried out using the forced swim and tail suspension tests in mice showed that DMPX at a dose of 6 and 12 mg/kg exerts antidepressant-like effect and does not affect the locomotor activity. Co-administration of DMPX at a dose of 3 mg/kg with the studied antidepressant drugs caused the reduction of immobility time in both behavioral tests. The observed effect was not associated with an increase in the locomotor activity. To evaluate whether the observed effects were due to a pharmacokinetic/pharmacodynamic interaction, the levels of the antidepressants in blood and brain were measured using high-performance liquid chromatography. It can be assumed that the interaction between DMPX and imipramine was exclusively pharmacodynamic in nature, whereas an increased antidepressant activity of escitalopram and reboxetine was at least partly related to its pharmacokinetic interaction with DMPX.
Collapse
Affiliation(s)
- Ewa Poleszak
- Department of Applied Pharmacy, Medical University of Lublin, Chodźki 1, PL 20-093, Lublin, Poland.
| | - Aleksandra Szopa
- Department of Applied Pharmacy, Medical University of Lublin, Chodźki 1, PL 20-093, Lublin, Poland.
| | - Karolina Bogatko
- Department of Applied Pharmacy, Medical University of Lublin, Chodźki 1, PL 20-093, Lublin, Poland
| | - Elżbieta Wyska
- Department of Pharmacokinetics and Physical Pharmacy, Collegium Medicum, Jagiellonian University, Medyczna 9, PL 30-688, Kraków, Poland
| | - Sylwia Wośko
- Department of Applied Pharmacy, Medical University of Lublin, Chodźki 1, PL 20-093, Lublin, Poland
| | - Katarzyna Świąder
- Department of Applied Pharmacy, Medical University of Lublin, Chodźki 1, PL 20-093, Lublin, Poland
| | - Urszula Doboszewska
- Department of Animal Physiology, Institute of Biology and Biochemistry, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Akademicka 19, PL 20-033, Lublin, Poland
| | - Aleksandra Wlaź
- Department of Pathophysiology, Medical University of Lublin, Jaczewskiego 8, PL 20-090, Lublin, Poland
| | - Andrzej Wróbel
- Second Department of Gynecology, Medical University of Lublin, Jaczewskiego 8, PL 20-090, Lublin, Poland
| | - Piotr Wlaź
- Department of Animal Physiology, Institute of Biology and Biochemistry, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Akademicka 19, PL 20-033, Lublin, Poland
| | - Anna Serefko
- Department of Applied Pharmacy, Medical University of Lublin, Chodźki 1, PL 20-093, Lublin, Poland
| |
Collapse
|
48
|
Soleimani A, Bahreyni A, Roshan MK, Soltani A, Ryzhikov M, Shafiee M, Soukhtanloo M, Jaafari MR, Mashkani B, Hassanian SM. Therapeutic potency of pharmacological adenosine receptors agonist/antagonist on cancer cell apoptosis in tumor microenvironment, current status, and perspectives. J Cell Physiol 2018; 234:2329-2336. [PMID: 30191994 DOI: 10.1002/jcp.27249] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Accepted: 07/24/2018] [Indexed: 12/14/2022]
Abstract
The hypoxic niche of tumor leads to a tremendous increase in the extracellular adenosine concentration through alteration of adenosine metabolism in the tumor microenvironment (TME). This consequently affects cancer progression, local immune responses, and apoptosis of tumor cells. Regulatory effect of adenosine on apoptosis in TME depends on the cancer cell type, pharmacological characteristics of adenosine receptor subtypes, and the adenosine concentration in the tumor niche. Exploiting specific pharmacological adenosine receptor agonist and antagonist inducing apoptosis in cancer cells can be considered as a proper procedure to control cancer progression. This review summarizes the regulatory role of adenosine in cancer cell apoptosis for a better understanding, and hence better management of the disease.
Collapse
Affiliation(s)
- Anvar Soleimani
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Bahreyni
- Department of Clinical Biochemistry and Immunogenetic Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Mazandaran, Iran
| | - Mostafa K Roshan
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Arash Soltani
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mikhail Ryzhikov
- Division of Pulmonary and Critical Care Medicine, Washington University, School of Medicine, Saint Louis, Missouri
| | - Mojtaba Shafiee
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Soukhtanloo
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud R Jaafari
- Nanotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Baratali Mashkani
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed M Hassanian
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Microanatomy Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
49
|
Koussémou M, Lorenz K, Klotz KN. The A2B adenosine receptor in MDA-MB-231 breast cancer cells diminishes ERK1/2 phosphorylation by activation of MAPK-phosphatase-1. PLoS One 2018; 13:e0202914. [PMID: 30157211 PMCID: PMC6114864 DOI: 10.1371/journal.pone.0202914] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 08/11/2018] [Indexed: 02/04/2023] Open
Abstract
It was previously shown that the estrogen-receptor negative breast cancer cell line MBA-MD-231 expresses high levels of A2B adenosine receptors as the sole adenosine receptor subtype. These receptors couple to both, stimulation of adenylyl cyclase and a Ca2+ signal. In order to establish a potential role of A2B adenosine receptors in tumor growth and development MAPK signaling was investigated in these breast cancer cells. Although it is known that A2B adenosine receptors may stimulate MAPK it was found that in MBA-MD-231 cells ERK1/2 phosphorylation is reduced upon agonist-stimulation of A2B adenosine receptors. This reduction is also triggered by forskolin, but abolished by the PKA inhibitor H89, suggesting an important role for the cAMP-PKA pathway. Likewise, a role for intracellular Ca2+ was established as the Ca2+ chelator 1,2-bis-(o-aminophenoxy)-ethane-N,N,N’,N’-tetraacetic acid, tetraacetoxymethyl ester (BAPTA-AM) abolished the reduction of ERK1/2 phosphorylation triggered by A2B stimulation. It was shown that various pathways downstream from A2B adenosine receptors resulted in a stimulation of MAPK phosphatase-1 (MKP-1) which dephosphorylates phospho ERK1/2, and thus plays a critical role in the regulation of the phosphorylation state of ERK1/2. The reduction of ERK1/2 phosphorylation mediated by A2B adenosine receptors might provide an interesting approach for adjuvant treatment leading to reduced growth of certain tumors expressing the A2B subtype.
Collapse
Affiliation(s)
- Marthe Koussémou
- Institut für Pharmakologie und Toxikologie, Universität Würzburg, Würzburg, Germany
| | - Kristina Lorenz
- Institut für Pharmakologie und Toxikologie, Universität Würzburg, Würzburg, Germany
- Leibniz-Institut für Analytische Wissenschaften–ISAS–e.V., Bunsen-Dortmund, Germany, and West German Heart and Vascular Center Essen, Essen, Germany
| | - Karl-Norbert Klotz
- Institut für Pharmakologie und Toxikologie, Universität Würzburg, Würzburg, Germany
- * E-mail:
| |
Collapse
|
50
|
Osborne DM, Sandau US, Jones AT, Vander Velden JW, Weingarten AM, Etesami N, Huo Y, Shen HY, Boison D. Developmental role of adenosine kinase for the expression of sex-dependent neuropsychiatric behavior. Neuropharmacology 2018; 141:89-97. [PMID: 30145320 DOI: 10.1016/j.neuropharm.2018.08.025] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 08/20/2018] [Accepted: 08/22/2018] [Indexed: 12/28/2022]
Abstract
Deficits in social memory, cognition, and aberrant responses to stimulants are common among persons affected by schizophrenia and other conditions with a presumed developmental etiology. We previously found that expression changes in the adenosine metabolizing enzyme adenosine kinase (ADK) in the adult brain are associated with deficits in various cognitive domains. To distinguish between developmental and adult functions of ADK, we used two transgenic mouse lines with widespread disruption of ADK expression in the adult brain, but differences in the onset of ADK deletion. Specifically, we compared Nestin-Cre+/-:ADK-floxfl/fl (ADKΔBrain) mice with global loss of ADK in the whole brain, beginning in mid-gestation and persisting for life, with Gfa2-Cre+/-:ADK-floxfl/fl (ADKΔAstro) mice that have normal ADK expression throughout development, but lose astrocyte-specific ADK-expression in young adulthood. Because ADK-expression in adulthood is generally confined to astrocytes, adult ADKΔAstro mice show a similar expression profile of ADK in key areas of the brain related to neuropsychiatric behavior, compared to adult ADKΔBrain mice. We sought to determine a neurodevelopmental role of ADK on the expression of psychiatric behaviors in adult male and female mice. Adult ADKΔBrain mice showed significant deficits in social memory in males, significant contextual learning impairments in both sexes, and a hyper-responsiveness to amphetamine in males. In contrast, ADKΔAstro mice showed normal social memory and contextual learning but hypo-responsiveness to amphetamine in males. Our results demonstrate a key developmental role of ADK in mediating behaviors in adulthood related to neuropsychiatric disease and support the greater prevalence of these disorders among males.
Collapse
Affiliation(s)
- D M Osborne
- Robert Stone Dow Neurobiology Laboratories, Legacy Research Institute, Portland, OR, USA.
| | - U S Sandau
- Robert Stone Dow Neurobiology Laboratories, Legacy Research Institute, Portland, OR, USA
| | - A T Jones
- Robert Stone Dow Neurobiology Laboratories, Legacy Research Institute, Portland, OR, USA
| | - J W Vander Velden
- Robert Stone Dow Neurobiology Laboratories, Legacy Research Institute, Portland, OR, USA
| | - A M Weingarten
- Robert Stone Dow Neurobiology Laboratories, Legacy Research Institute, Portland, OR, USA
| | - N Etesami
- Robert Stone Dow Neurobiology Laboratories, Legacy Research Institute, Portland, OR, USA
| | - Y Huo
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - H Y Shen
- Robert Stone Dow Neurobiology Laboratories, Legacy Research Institute, Portland, OR, USA
| | - D Boison
- Robert Stone Dow Neurobiology Laboratories, Legacy Research Institute, Portland, OR, USA
| |
Collapse
|