1
|
Barik S, Riddell T. The Brain-Heart Network of Syncope. Int J Mol Sci 2024; 25:6959. [PMID: 39000068 PMCID: PMC11241714 DOI: 10.3390/ijms25136959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 06/23/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024] Open
Abstract
Observed and recorded in various forms since ancient times, 'syncope' is often popularly called 'fainting', such that the two terms are used synonymously. Syncope/fainting can be caused by a variety of conditions, including but not limited to head injuries, vertigo, and oxygen deficiency. Here, we draw on a large body of literature on syncope, including the role of a recently discovered set of specialized mammalian neurons. Although the etiology of syncope still remains a mystery, we have attempted to provide a comprehensive account of what is known and what still needs to be performed. Much of our understanding of syncope is owing to studies in the laboratory mouse, whereas evidence from human patients remains scarce. Interestingly, the cardioinhibitory Bezold-Jarisch reflex, recognized in the early 1900s, has an intriguing similarity to-and forms the basis of-syncope. In this review, we have integrated this minimal model into the modern view of the brain-neuron-heart signaling loop of syncope, to which several signaling events contribute. Molecular signaling is our major focus here, presented in terms of a normal heart, and thus, syncope due to abnormal or weak heart activity is not discussed in detail. In addition, we have offered possible directions for clinical intervention based on this model. Overall, this article is expected to generate interest in chronic vertigo and syncope/fainting, an enigmatic condition that affects most humans at some point in life; it is also hoped that this may lead to a mechanism-based clinical intervention in the future.
Collapse
Affiliation(s)
- Sailen Barik
- Independent Researcher, EonBio, 3780 Pelham Drive, Mobile, AL 36619, USA
| | | |
Collapse
|
2
|
Oliva P, Suresh RR, Pasquini S, Salmaso V, Will EJ, Tosh DK, Gao ZG, Liu N, Gavrilova O, Vincenzi F, Varani K, Jacobson KA. 2-Amino-5-arylethynyl-thiophen-3-yl-(phenyl)methanones as A 1 Adenosine Receptor Positive Allosteric Modulators. ACS Med Chem Lett 2023; 14:1640-1646. [PMID: 38116442 PMCID: PMC10726435 DOI: 10.1021/acsmedchemlett.3c00315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 10/06/2023] [Indexed: 12/21/2023] Open
Abstract
A1 adenosine receptor (A1AR) agonists have cerebroprotective, cardioprotective, antinociceptive, and other pharmaceutical applications. We explored the structure-activity relationship of 5-arylethynyl aminothiophenes as A1AR positive allosteric modulators (PAMs). The derivatives were compared in binding and functional assays at the human A1AR, indicating that some fluoro-substituted analogues have enhanced PAM activity. We identified substitution of the terminal phenyl ring in 12 (2-F-Ph), 15 (3,4-F2-Ph, MRS7935), and 21 (2-CF3-Ph) as particularly enhancing the PAM activity. 15 was also shown to act as an A1 ago-PAM with EC50 ≈ 2 μM, without activity (30 μM) at other ARs. Molecular modeling indicated that both the 5-arylethynyl and the 4-neopentyl groups are located in a region outside the receptor transmembrane helix bundle that is in contact with the phospholipid bilayer, consistent with the preference for nonpolar substitution of the aryl moiety. Although they are hydrophobic, these PAMs could provide potential drug candidate molecules for engaging protective A1ARs.
Collapse
Affiliation(s)
- Paola Oliva
- Laboratory
of Bioorganic Chemistry and Mouse Metabolism Core, National
Institute of Diabetes and Digestive and Kidney
Disease, National Institutes of Health, 9000 Rockville Pike, Bethesda, Maryland 20892, United States
| | - R. Rama Suresh
- Laboratory
of Bioorganic Chemistry and Mouse Metabolism Core, National
Institute of Diabetes and Digestive and Kidney
Disease, National Institutes of Health, 9000 Rockville Pike, Bethesda, Maryland 20892, United States
| | - Silvia Pasquini
- Department
of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Via Fossato di Mortara 17-19, 44121 Ferrara, Italy
| | - Veronica Salmaso
- Laboratory
of Bioorganic Chemistry and Mouse Metabolism Core, National
Institute of Diabetes and Digestive and Kidney
Disease, National Institutes of Health, 9000 Rockville Pike, Bethesda, Maryland 20892, United States
| | - Edward J. Will
- Laboratory
of Bioorganic Chemistry and Mouse Metabolism Core, National
Institute of Diabetes and Digestive and Kidney
Disease, National Institutes of Health, 9000 Rockville Pike, Bethesda, Maryland 20892, United States
| | - Dilip K. Tosh
- Laboratory
of Bioorganic Chemistry and Mouse Metabolism Core, National
Institute of Diabetes and Digestive and Kidney
Disease, National Institutes of Health, 9000 Rockville Pike, Bethesda, Maryland 20892, United States
| | - Zhan-Guo Gao
- Laboratory
of Bioorganic Chemistry and Mouse Metabolism Core, National
Institute of Diabetes and Digestive and Kidney
Disease, National Institutes of Health, 9000 Rockville Pike, Bethesda, Maryland 20892, United States
| | - Naili Liu
- Laboratory
of Bioorganic Chemistry and Mouse Metabolism Core, National
Institute of Diabetes and Digestive and Kidney
Disease, National Institutes of Health, 9000 Rockville Pike, Bethesda, Maryland 20892, United States
| | - Oksana Gavrilova
- Laboratory
of Bioorganic Chemistry and Mouse Metabolism Core, National
Institute of Diabetes and Digestive and Kidney
Disease, National Institutes of Health, 9000 Rockville Pike, Bethesda, Maryland 20892, United States
| | - Fabrizio Vincenzi
- Department
of Translational Medicine, University of
Ferrara, Via Fossato
di Mortara 17-19, 44121 Ferrara, Italy
| | - Katia Varani
- Department
of Translational Medicine, University of
Ferrara, Via Fossato
di Mortara 17-19, 44121 Ferrara, Italy
| | - Kenneth A. Jacobson
- Laboratory
of Bioorganic Chemistry and Mouse Metabolism Core, National
Institute of Diabetes and Digestive and Kidney
Disease, National Institutes of Health, 9000 Rockville Pike, Bethesda, Maryland 20892, United States
| |
Collapse
|
3
|
Haddad M, Cherchi F, Alsalem M, Al-saraireh YM, Madae’en S. Adenosine Receptors as Potential Therapeutic Analgesic Targets. Int J Mol Sci 2023; 24:13160. [PMID: 37685963 PMCID: PMC10487796 DOI: 10.3390/ijms241713160] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 08/18/2023] [Accepted: 08/23/2023] [Indexed: 09/10/2023] Open
Abstract
Pain represents an international burden and a major socio-economic public health problem. New findings, detailed in this review, suggest that adenosine plays a significant role in neuropathic and inflammatory pain, by acting on its metabotropic adenosine receptors (A1AR, A2AAR, A2BAR, A3AR). Adenosine receptor ligands have a practical translational potential based on the favorable efficacy and safety profiles that emerged from clinical research on various agonists and antagonists for different pathologies. The present review collects the latest studies on selected adenosine receptor ligands in different pain models. Here, we also covered the many hypothesized pathways and the role of newly synthesized allosteric adenosine receptor modulators. This review aims to present a summary of recent research on adenosine receptors as prospective therapeutic targets for a range of pain-related disorders.
Collapse
Affiliation(s)
- Mansour Haddad
- Faculty of Pharmacy, Yarmouk University, Irbid 21163, Jordan
| | - Federica Cherchi
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology and Toxicology, University of Florence, 50139 Florence, Italy;
| | - Mohammad Alsalem
- School of Medicine, The University of Jordan, Amman 11942, Jordan;
| | - Yousef M. Al-saraireh
- Department of Pharmacology, Faculty of Medicine, Mutah University, P.O. Box 7, Al-Karak 61710, Jordan;
| | - Saba Madae’en
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmaceutical Sciences, The Hashemite University, Zarqa 13133, Jordan;
| |
Collapse
|
4
|
IJzerman AP, Jacobson KA, Müller CE, Cronstein BN, Cunha RA. International Union of Basic and Clinical Pharmacology. CXII: Adenosine Receptors: A Further Update. Pharmacol Rev 2022; 74:340-372. [PMID: 35302044 PMCID: PMC8973513 DOI: 10.1124/pharmrev.121.000445] [Citation(s) in RCA: 74] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Our previous International Union of Basic and Clinical Pharmacology report on the nomenclature and classification of adenosine receptors (2011) contained a number of emerging developments with respect to this G protein-coupled receptor subfamily, including protein structure, protein oligomerization, protein diversity, and allosteric modulation by small molecules. Since then, a wealth of new data and results has been added, allowing us to explore novel concepts such as target binding kinetics and biased signaling of adenosine receptors, to examine a multitude of receptor structures and novel ligands, to gauge new pharmacology, and to evaluate clinical trials with adenosine receptor ligands. This review should therefore be considered a further update of our previous reports from 2001 and 2011. SIGNIFICANCE STATEMENT: Adenosine receptors (ARs) are of continuing interest for future treatment of chronic and acute disease conditions, including inflammatory diseases, neurodegenerative afflictions, and cancer. The design of AR agonists ("biased" or not) and antagonists is largely structure based now, thanks to the tremendous progress in AR structural biology. The A2A- and A2BAR appear to modulate the immune response in tumor biology. Many clinical trials for this indication are ongoing, whereas an A2AAR antagonist (istradefylline) has been approved as an anti-Parkinson agent.
Collapse
Affiliation(s)
- Adriaan P IJzerman
- Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands (A.P.IJ.); National Institutes of Health, National Institute of Diabetes and Digestive and Kidney Diseases, Molecular Recognition Section, Bethesda, Maryland (K.A.J.); Universität Bonn, Bonn, Germany (C.E.M.); New York University School of Medicine, New York, New York (B.N.C.); and Center for Neurosciences and Cell Biology and Faculty of Medicine, University of Coimbra, Coimbra, Portugal (R.A.C.)
| | - Kenneth A Jacobson
- Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands (A.P.IJ.); National Institutes of Health, National Institute of Diabetes and Digestive and Kidney Diseases, Molecular Recognition Section, Bethesda, Maryland (K.A.J.); Universität Bonn, Bonn, Germany (C.E.M.); New York University School of Medicine, New York, New York (B.N.C.); and Center for Neurosciences and Cell Biology and Faculty of Medicine, University of Coimbra, Coimbra, Portugal (R.A.C.)
| | - Christa E Müller
- Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands (A.P.IJ.); National Institutes of Health, National Institute of Diabetes and Digestive and Kidney Diseases, Molecular Recognition Section, Bethesda, Maryland (K.A.J.); Universität Bonn, Bonn, Germany (C.E.M.); New York University School of Medicine, New York, New York (B.N.C.); and Center for Neurosciences and Cell Biology and Faculty of Medicine, University of Coimbra, Coimbra, Portugal (R.A.C.)
| | - Bruce N Cronstein
- Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands (A.P.IJ.); National Institutes of Health, National Institute of Diabetes and Digestive and Kidney Diseases, Molecular Recognition Section, Bethesda, Maryland (K.A.J.); Universität Bonn, Bonn, Germany (C.E.M.); New York University School of Medicine, New York, New York (B.N.C.); and Center for Neurosciences and Cell Biology and Faculty of Medicine, University of Coimbra, Coimbra, Portugal (R.A.C.)
| | - Rodrigo A Cunha
- Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands (A.P.IJ.); National Institutes of Health, National Institute of Diabetes and Digestive and Kidney Diseases, Molecular Recognition Section, Bethesda, Maryland (K.A.J.); Universität Bonn, Bonn, Germany (C.E.M.); New York University School of Medicine, New York, New York (B.N.C.); and Center for Neurosciences and Cell Biology and Faculty of Medicine, University of Coimbra, Coimbra, Portugal (R.A.C.)
| |
Collapse
|
5
|
Kim TH, Bormate KJ, Custodio RJP, Cheong JH, Lee BK, Kim HJ, Jung YS. Involvement of the adenosine A 1 receptor in the hypnotic effect of rosmarinic acid. Biomed Pharmacother 2022; 146:112483. [PMID: 34891112 DOI: 10.1016/j.biopha.2021.112483] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 11/23/2021] [Accepted: 11/30/2021] [Indexed: 11/18/2022] Open
Abstract
Insomnia, the most common sleep disorder, is characterized by a longer sleep latency, greater sleep fragmentation, and consequent excessive daytime fatigue. Due to the various side effects of prescribed hypnotics, demand for new drugs is still high. Recent studies have suggested the adenosine receptor (AR) as a potential therapeutic target for insomnia, however, clinically useful hypnotics targeting AR are not yet available. In the present study, we evaluated the hypnotic effect of rosmarinic acid, a phenolic compound widely found in medicinal plants, through pentobarbital-induced sleep test, electroencephalography/electromyography (EEG/EMG), and immunohistochemistry in mice. The underlying mechanisms were assessed by pharmacological approach using 8-cyclopentyl-1,3-dipropylxanthine (DPCPX) and SCH5826, antagonists for A1R and A2AR, respectively. Receptor-binding assay and functional agonism were also performed. Our study provides a new evidence that rosmarinic acid has a direct binding activity (Ki = 14.21 ± 0.3 μM) and agonistic activity for A1R. We also found that rosmarinic acid significantly decreased sleep fragmentation and onset latency to NREM sleep, and these effects were abolished by DPCPX. The results from c-Fos immunostaining showed that rosmarinic acid decreased the neuronal activity in wake-promoting brain regions, such as the basal forebrain and the lateral hypothalamus, while increasing the neuronal activity in the ventrolateral preoptic nucleus, a sleep-promoting region; all these effects were significantly inhibited by DPCPX. Taken together, this study suggests that rosmarinic acid possesses novel activity as an A1R agonist and thereby exerts a hypnotic effect, and thus it may serve as a potential therapeutic agent for insomnia through targeting A1R.
Collapse
Affiliation(s)
- Tae-Ho Kim
- College of Pharmacy, Research Institute of Pharmaceutical Sciences and Technology, Ajou University, Suwon 16499, Republic of Korea
| | - Katrina Joy Bormate
- College of Pharmacy, Research Institute of Pharmaceutical Sciences and Technology, Ajou University, Suwon 16499, Republic of Korea
| | | | - Jae Hoon Cheong
- School of Pharmacy, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Bo Kyung Lee
- College of Pharmacy, Research Institute of Pharmaceutical Sciences and Technology, Ajou University, Suwon 16499, Republic of Korea
| | - Hee Jin Kim
- Uimyung Research Institute in Neuroscience, Sahmyook University, Seoul 01795, Republic of Korea.
| | - Yi-Sook Jung
- College of Pharmacy, Research Institute of Pharmaceutical Sciences and Technology, Ajou University, Suwon 16499, Republic of Korea.
| |
Collapse
|
6
|
Adenosine Receptors in Neuropsychiatric Disorders: Fine Regulators of Neurotransmission and Potential Therapeutic Targets. Int J Mol Sci 2022; 23:ijms23031219. [PMID: 35163142 PMCID: PMC8835915 DOI: 10.3390/ijms23031219] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/20/2022] [Accepted: 01/21/2022] [Indexed: 12/16/2022] Open
Abstract
Adenosine exerts an important role in the modulation of central nervous system (CNS) activity. Through the interaction with four G-protein coupled receptor (GPCR) subtypes, adenosine subtly regulates neurotransmission, interfering with the dopaminergic, glutamatergic, noradrenergic, serotoninergic, and endocannabinoid systems. The inhibitory and facilitating actions of adenosine on neurotransmission are mainly mediated by A1 and A2A adenosine receptors (ARs), respectively. Given their role in the CNS, ARs are promising therapeutic targets for neuropsychiatric disorders where altered neurotransmission represents the most likely etiological hypothesis. Activating or blocking ARs with specific pharmacological agents could therefore restore the balance of altered neurotransmitter systems, providing the rationale for the potential treatment of these highly debilitating conditions. In this review, we summarize and discuss the most relevant studies concerning AR modulation in psychotic and mood disorders such as schizophrenia, bipolar disorders, depression, and anxiety, as well as neurodevelopment disorders such as autism spectrum disorder (ASD), fragile X syndrome (FXS), attention-deficit hyperactivity disorder (ADHD), and neuropsychiatric aspects of neurodegenerative disorders.
Collapse
|
7
|
Li W, Hu B, Liu H, Luan J, Chen L, Wang S, Fan L, Wang J. In silico investigation of the selectivity mechanism of A 1AR and A 2AAR antagonism. NEW J CHEM 2022. [DOI: 10.1039/d2nj03536g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Adenosine A1 receptor (A1AR) and adenosine A2A receptor (A2AAR) are AR isoforms that share high homology but play many different roles in terms of regulating arteriolar pressure and urine flow as well as relieving neurodegenerative disorders.
Collapse
Affiliation(s)
- Weixia Li
- Key Laboratory of Structure-Based Drug Design &Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
- Key Laboratory of Intelligent Drug Design and New Drug Discovery of Liaoning Province, Shenyang Pharmaceutical University, Shenyang 110016, China
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Baichun Hu
- Key Laboratory of Structure-Based Drug Design &Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
- Key Laboratory of Intelligent Drug Design and New Drug Discovery of Liaoning Province, Shenyang Pharmaceutical University, Shenyang 110016, China
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Haihan Liu
- Key Laboratory of Structure-Based Drug Design &Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
- Key Laboratory of Intelligent Drug Design and New Drug Discovery of Liaoning Province, Shenyang Pharmaceutical University, Shenyang 110016, China
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Jiasi Luan
- Key Laboratory of Structure-Based Drug Design &Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
- Key Laboratory of Intelligent Drug Design and New Drug Discovery of Liaoning Province, Shenyang Pharmaceutical University, Shenyang 110016, China
- School of Medical Devices, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Lu Chen
- Key Laboratory of Structure-Based Drug Design &Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
- Key Laboratory of Intelligent Drug Design and New Drug Discovery of Liaoning Province, Shenyang Pharmaceutical University, Shenyang 110016, China
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Shizun Wang
- Key Laboratory of Structure-Based Drug Design &Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
- Key Laboratory of Intelligent Drug Design and New Drug Discovery of Liaoning Province, Shenyang Pharmaceutical University, Shenyang 110016, China
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Liye Fan
- Key Laboratory of Structure-Based Drug Design &Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
- Key Laboratory of Intelligent Drug Design and New Drug Discovery of Liaoning Province, Shenyang Pharmaceutical University, Shenyang 110016, China
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Jian Wang
- Key Laboratory of Structure-Based Drug Design &Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
- Key Laboratory of Intelligent Drug Design and New Drug Discovery of Liaoning Province, Shenyang Pharmaceutical University, Shenyang 110016, China
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| |
Collapse
|
8
|
Zuccarini M, Lambertucci C, Carluccio M, Giuliani P, Ronci M, Spinaci A, Volpini R, Ciccarelli R, Di Iorio P. Multipotent Stromal Cells from Subcutaneous Adipose Tissue of Normal Weight and Obese Subjects: Modulation of Their Adipogenic Differentiation by Adenosine A 1 Receptor Ligands. Cells 2021; 10:cells10123560. [PMID: 34944069 PMCID: PMC8700077 DOI: 10.3390/cells10123560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/09/2021] [Accepted: 12/15/2021] [Indexed: 11/18/2022] Open
Abstract
Adenosine A1 receptor (A1R) activation, stimulating lipogenesis and decreasing insulin resistance, could be useful for metabolic syndrome management in obese subjects. Since full A1R agonists induce harmful side-effects, while partial agonists show a better pharmacological profile, we investigated the influence of two derivatives of the full A1R agonist 2-chloro-N6-cyclopentyladenosine (CCPA), C1 and C2 behaving as A1R partial agonists in animal models, on the adipogenic differentiation of stromal/stem cells (ASCs) from human subcutaneous adipose tissue, which mainly contribute to increase fat mass in obesity. The ASCs from normal-weight subjects showed increased proliferation and A1R expression but reduced adipogenic differentiation compared to obese individual-derived ASCs. Cell exposure to CCPA, C1, C2 or DPCPX, an A1R antagonist, did not affect ASC proliferation, while mainly C2 and DPCPX significantly decreased adipogenic differentiation of both ASC types, reducing the activity of glycerol-3-phosphate dehydrogenase and the expression of PPARγ and FABP-4, all adipogenic markers, and phosphorylation of Akt in the phosphatidylinositol-3-kinase pathway, which plays a key-role in adipogenesis. While requiring confirmation in in vivo models, our results suggest that A1R partial agonists or antagonists, by limiting ASC differentiation into adipocytes and, thereby, fat mass expansion, could favor development/worsening of metabolic syndrome in obese subjects without a dietary control.
Collapse
Affiliation(s)
- Mariachiara Zuccarini
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Via dei Vestini 29, 66100 Chieti, Italy; (M.Z.); (M.C.); (P.G.); (P.D.I.)
- Center for Advanced Study and Technologies (CAST), University of Chieti-Pescara, Via L. Polacchi 11, 66100 Chieti, Italy
| | - Catia Lambertucci
- Unit of Medicinal Chemistry, School of Pharmacy, University of Camerino, Via S. Agostino 1, 62032 Camerino, Italy; (C.L.); (A.S.); (R.V.)
| | - Marzia Carluccio
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Via dei Vestini 29, 66100 Chieti, Italy; (M.Z.); (M.C.); (P.G.); (P.D.I.)
- Center for Advanced Study and Technologies (CAST), University of Chieti-Pescara, Via L. Polacchi 11, 66100 Chieti, Italy
- Stem TeCh Group, Via L. Polacchi 11, 66100 Chieti, Italy
| | - Patricia Giuliani
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Via dei Vestini 29, 66100 Chieti, Italy; (M.Z.); (M.C.); (P.G.); (P.D.I.)
- Center for Advanced Study and Technologies (CAST), University of Chieti-Pescara, Via L. Polacchi 11, 66100 Chieti, Italy
| | - Maurizio Ronci
- Department of Pharmacy, University of Chieti-Pescara, Via dei Vestini 29, 66100 Chieti, Italy;
| | - Andrea Spinaci
- Unit of Medicinal Chemistry, School of Pharmacy, University of Camerino, Via S. Agostino 1, 62032 Camerino, Italy; (C.L.); (A.S.); (R.V.)
| | - Rosaria Volpini
- Unit of Medicinal Chemistry, School of Pharmacy, University of Camerino, Via S. Agostino 1, 62032 Camerino, Italy; (C.L.); (A.S.); (R.V.)
| | - Renata Ciccarelli
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Via dei Vestini 29, 66100 Chieti, Italy; (M.Z.); (M.C.); (P.G.); (P.D.I.)
- Center for Advanced Study and Technologies (CAST), University of Chieti-Pescara, Via L. Polacchi 11, 66100 Chieti, Italy
- Unit of Medicinal Chemistry, School of Pharmacy, University of Camerino, Via S. Agostino 1, 62032 Camerino, Italy; (C.L.); (A.S.); (R.V.)
- Correspondence:
| | - Patrizia Di Iorio
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Via dei Vestini 29, 66100 Chieti, Italy; (M.Z.); (M.C.); (P.G.); (P.D.I.)
- Center for Advanced Study and Technologies (CAST), University of Chieti-Pescara, Via L. Polacchi 11, 66100 Chieti, Italy
| |
Collapse
|
9
|
Wang J, Arantes PR, Bhattarai A, Hsu RV, Pawnikar S, Huang YMM, Palermo G, Miao Y. Gaussian accelerated molecular dynamics (GaMD): principles and applications. WILEY INTERDISCIPLINARY REVIEWS. COMPUTATIONAL MOLECULAR SCIENCE 2021; 11:e1521. [PMID: 34899998 PMCID: PMC8658739 DOI: 10.1002/wcms.1521] [Citation(s) in RCA: 95] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 01/28/2021] [Indexed: 12/20/2022]
Abstract
Gaussian accelerated molecular dynamics (GaMD) is a robust computational method for simultaneous unconstrained enhanced sampling and free energy calculations of biomolecules. It works by adding a harmonic boost potential to smooth biomolecular potential energy surface and reduce energy barriers. GaMD greatly accelerates biomolecular simulations by orders of magnitude. Without the need to set predefined reaction coordinates or collective variables, GaMD provides unconstrained enhanced sampling and is advantageous for simulating complex biological processes. The GaMD boost potential exhibits a Gaussian distribution, thereby allowing for energetic reweighting via cumulant expansion to the second order (i.e., "Gaussian approximation"). This leads to accurate reconstruction of free energy landscapes of biomolecules. Hybrid schemes with other enhanced sampling methods, such as the replica exchange GaMD (rex-GaMD) and replica exchange umbrella sampling GaMD (GaREUS), have also been introduced, further improving sampling and free energy calculations. Recently, new "selective GaMD" algorithms including the ligand GaMD (LiGaMD) and peptide GaMD (Pep-GaMD) enabled microsecond simulations to capture repetitive dissociation and binding of small-molecule ligands and highly flexible peptides. The simulations then allowed highly efficient quantitative characterization of the ligand/peptide binding thermodynamics and kinetics. Taken together, GaMD and its innovative variants are applicable to simulate a wide variety of biomolecular dynamics, including protein folding, conformational changes and allostery, ligand binding, peptide binding, protein-protein/nucleic acid/carbohydrate interactions, and carbohydrate/nucleic acid interactions. In this review, we present principles of the GaMD algorithms and recent applications in biomolecular simulations and drug design.
Collapse
Affiliation(s)
- Jinan Wang
- Center for Computational Biology and Department of Molecular Biosciences, University of Kansas, 2030 Becker Dr., Lawrence, KS, 66047, United States
| | - Pablo R Arantes
- Department of Bioengineering, University of California Riverside, 900 University Avenue, Riverside, CA 92512, United States
| | - Apurba Bhattarai
- Center for Computational Biology and Department of Molecular Biosciences, University of Kansas, 2030 Becker Dr, Lawrence, KS, 66047, United States
| | - Rohaine V Hsu
- Department of Bioengineering, University of California Riverside, 900 University Avenue, Riverside, CA 92512, United States
| | - Shristi Pawnikar
- Center for Computational Biology and Department of Molecular Biosciences, University of Kansas, 2030 Becker Dr., Lawrence, KS, 66047, United States
| | - Yu-Ming M Huang
- Department of Physics & Astronomy, Wayne State University, 666 W Hancock St, Detroit, MI 48207, USA
| | - Giulia Palermo
- Department of Bioengineering and Department of Chemistry, University of California Riverside, 900 University Avenue, Riverside, CA 92512, United States
| | - Yinglong Miao
- Center for Computational Biology and Department of Molecular Biosciences, University of Kansas, 2030 Becker Dr., Lawrence, Kansas 66047, United States
| |
Collapse
|
10
|
Kim YB, Choi J, Park C, Choi H, In J, Yang H. Effects of sevoflurane and adenosine receptor antagonist on the sugammadex-induced recovery from rocuronium-induced neuromuscular blockade in rodent phrenic nerve-hemidiaphragm tissue specimens. Pharmacol Res Perspect 2021; 9:e00827. [PMID: 34337892 PMCID: PMC8326504 DOI: 10.1002/prp2.827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/03/2021] [Accepted: 06/03/2021] [Indexed: 11/17/2022] Open
Abstract
Sevoflurane affects on the A1 receptor in the central nervous system and potentiates the action of neuromuscular blocking agents. In the present study, we investigated whether sevoflurane (SEVO) has the ability to potentiate the neuromuscular blocking effect of rocuronium and if the specific antagonist of adenosine receptor (SLV320) can reverse this effect. In this study, phrenic nerve-hemidiaphragm tissue specimens were obtained from 40 Sprague-Dawley (SD) rats. The specimens were immersed in an organ bath filled with Krebs buffer and stimulated by a train-of-four (TOF) pattern using indirect supramaximal stimulation at 20 s intervals. The specimens were randomly allocated to control, 2-chloroadenosine (CADO), SEVO, or SLV320 + SEVO groups. In the CADO and SLV320 + SEVO groups, CADO and SLV320 were added to the organ bath from the start to a concentration of 10 μM and 10 nM, respectively. We then proceeded with rocuronium-induced blockade of >95% depression of the first twitch tension of TOF (T1) and TOF ratio (TOFR). In the SEVO and SLV320 + SEVO groups, SEVO was added to the Krebs buffer solution to concentration of 400-500 μM for 10 min. Sugammadex-induced T1 and TOFR recovery was monitored for 30 min until >95% of T1 and >0.9 of TOFR were confirmed, and the recovery pattern was compared by plotting these data. T1 recovery in the SEVO and CADO groups was significantly delayed compared with the control and SLV320 + SEVO groups (p < .05). In conclusion, sevoflurane affects on the A1 receptor at the neuromuscular junction and delays sugammadex-induced recovery from neuromuscular blockade.
Collapse
Affiliation(s)
- Yong Beom Kim
- Department of Anesthesiology and Pain MedicineCollege of MedicineGil Medical CenterGachon UniversityIncheonRepublic of Korea
| | - Jae‐Moon Choi
- Department of Anesthesiology and Pain MedicineAsan Medical CenterUlsan University, College of MedicineSeoulRepublic of Korea
| | - Chungon Park
- Department of Anesthesiology and Pain MedicineCollege of MedicineGil Medical CenterGachon UniversityIncheonRepublic of Korea
| | - Hey‐Ran Choi
- Department of Anaesthesiology and Pain MedicineSeoul Paik HospitalInje University, College of MedicineSeoulRepublic of Korea
| | - Junyong In
- Department of Anesthesiology and Pain MedicineCollege of MedicineIlsan HospitalDonggook UniversityGoyangGyeonggi DoRepublic of Korea
| | - Hong‐Seuk Yang
- Department of Anaesthesiology and Pain MedicineDaejeon Eulji HospitalEulji UniversityDaejeonRepublic of Korea
| |
Collapse
|
11
|
Leduc-Pessah H, Xu C, Fan CY, Dalgarno R, Kohro Y, Sparanese S, Burke NN, Jacobson KA, Altier C, Salvemini D, Trang T. Spinal A 3 adenosine receptor activation acutely restores morphine antinociception in opioid tolerant male rats. J Neurosci Res 2021; 100:251-264. [PMID: 34075613 DOI: 10.1002/jnr.24869] [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: 01/09/2021] [Revised: 04/20/2021] [Accepted: 05/12/2021] [Indexed: 01/01/2023]
Abstract
Opioids are potent analgesics, but their pain-relieving effects diminish with repeated use. The reduction in analgesic potency is a hallmark of opioid analgesic tolerance, which hampers opioid pain therapy. In the central nervous system, opioid analgesia is critically modulated by adenosine, a purine nucleoside implicated in the beneficial and detrimental actions of opioid medications. Here, we focus on the A3 adenosine receptor (A3 AR) in opioid analgesic tolerance. Intrathecal administration of the A3 AR agonist MRS5698 with daily systemic morphine in male rats attenuated the reduction in morphine antinociception over 7 days. In rats with established morphine tolerance, intrathecal MRS5698 partially restored the antinociceptive effects of morphine. However, when MRS5698 was discontinued, these animals displayed a reduced antinociceptive response to morphine. Our results suggest that MRS5698 acutely and transiently potentiates morphine antinociception in tolerant rats. By contrast, in morphine-naïve rats MRS5698 treatment did not impact thermal nociceptive threshold or affect antinociceptive response to a single injection of morphine. Furthermore, we found that morphine-induced adenosine release in cerebrospinal fluid was blunted in tolerant animals, but total spinal A3 AR expression was not affected. Collectively, our findings indicate that spinal A3 AR activation acutely potentiates morphine antinociception in the opioid tolerant state.
Collapse
Affiliation(s)
- Heather Leduc-Pessah
- Department of Comparative Biology and Experimental Medicine, University of Calgary, Calgary, AB, Canada.,Department of Physiology & Pharmacology, University of Calgary, Calgary, AB, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Cynthia Xu
- Department of Comparative Biology and Experimental Medicine, University of Calgary, Calgary, AB, Canada.,Department of Physiology & Pharmacology, University of Calgary, Calgary, AB, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Churmy Y Fan
- Department of Comparative Biology and Experimental Medicine, University of Calgary, Calgary, AB, Canada.,Department of Physiology & Pharmacology, University of Calgary, Calgary, AB, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Rebecca Dalgarno
- Department of Comparative Biology and Experimental Medicine, University of Calgary, Calgary, AB, Canada.,Department of Physiology & Pharmacology, University of Calgary, Calgary, AB, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Yuta Kohro
- Department of Comparative Biology and Experimental Medicine, University of Calgary, Calgary, AB, Canada.,Department of Physiology & Pharmacology, University of Calgary, Calgary, AB, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Sydney Sparanese
- Department of Comparative Biology and Experimental Medicine, University of Calgary, Calgary, AB, Canada.,Department of Physiology & Pharmacology, University of Calgary, Calgary, AB, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Nikita N Burke
- Department of Comparative Biology and Experimental Medicine, University of Calgary, Calgary, AB, Canada.,Department of Physiology & Pharmacology, University of Calgary, Calgary, AB, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Kenneth A Jacobson
- National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, USA
| | - Christophe Altier
- Department of Comparative Biology and Experimental Medicine, University of Calgary, Calgary, AB, Canada.,Department of Physiology & Pharmacology, University of Calgary, Calgary, AB, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada.,Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Daniela Salvemini
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO, USA.,Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - Tuan Trang
- Department of Comparative Biology and Experimental Medicine, University of Calgary, Calgary, AB, Canada.,Department of Physiology & Pharmacology, University of Calgary, Calgary, AB, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
12
|
Procopio MC, Lauro R, Nasso C, Carerj S, Squadrito F, Bitto A, Di Bella G, Micari A, Irrera N, Costa F. Role of Adenosine and Purinergic Receptors in Myocardial Infarction: Focus on Different Signal Transduction Pathways. Biomedicines 2021; 9:biomedicines9020204. [PMID: 33670488 PMCID: PMC7922652 DOI: 10.3390/biomedicines9020204] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 02/15/2021] [Indexed: 12/24/2022] Open
Abstract
Myocardial infarction (MI) is a dramatic event often caused by atherosclerotic plaque erosion or rupture and subsequent thrombotic occlusion of a coronary vessel. The low supply of oxygen and nutrients in the infarcted area may result in cardiomyocytes necrosis, replacement of intact myocardium with non-contractile fibrous tissue and left ventricular (LV) function impairment if blood flow is not quickly restored. In this review, we summarized the possible correlation between adenosine system, purinergic system and Wnt/β-catenin pathway and their role in the pathogenesis of cardiac damage following MI. In this context, several pathways are involved and, in particular, the adenosine receptors system shows different interactions between its members and purinergic receptors: their modulation might be effective not only for a normal functional recovery but also for the treatment of heart diseases, thus avoiding fibrosis, reducing infarcted area and limiting scaring. Similarly, it has been shown that Wnt/β catenin pathway is activated following myocardial injury and its unbalanced activation might promote cardiac fibrosis and, consequently, LV systolic function impairment. In this regard, the therapeutic benefits of Wnt inhibitors use were highlighted, thus demonstrating that Wnt/β-catenin pathway might be considered as a therapeutic target to prevent adverse LV remodeling and heart failure following MI.
Collapse
Affiliation(s)
- Maria Cristina Procopio
- Department of Clinical and Experimental Medicine, University of Messina, 98165 Messina, Italy; (M.C.P.); (R.L.); (C.N.); (S.C.); (F.S.); (A.B.); (G.D.B.); (F.C.)
| | - Rita Lauro
- Department of Clinical and Experimental Medicine, University of Messina, 98165 Messina, Italy; (M.C.P.); (R.L.); (C.N.); (S.C.); (F.S.); (A.B.); (G.D.B.); (F.C.)
| | - Chiara Nasso
- Department of Clinical and Experimental Medicine, University of Messina, 98165 Messina, Italy; (M.C.P.); (R.L.); (C.N.); (S.C.); (F.S.); (A.B.); (G.D.B.); (F.C.)
| | - Scipione Carerj
- Department of Clinical and Experimental Medicine, University of Messina, 98165 Messina, Italy; (M.C.P.); (R.L.); (C.N.); (S.C.); (F.S.); (A.B.); (G.D.B.); (F.C.)
| | - Francesco Squadrito
- Department of Clinical and Experimental Medicine, University of Messina, 98165 Messina, Italy; (M.C.P.); (R.L.); (C.N.); (S.C.); (F.S.); (A.B.); (G.D.B.); (F.C.)
| | - Alessandra Bitto
- Department of Clinical and Experimental Medicine, University of Messina, 98165 Messina, Italy; (M.C.P.); (R.L.); (C.N.); (S.C.); (F.S.); (A.B.); (G.D.B.); (F.C.)
| | - Gianluca Di Bella
- Department of Clinical and Experimental Medicine, University of Messina, 98165 Messina, Italy; (M.C.P.); (R.L.); (C.N.); (S.C.); (F.S.); (A.B.); (G.D.B.); (F.C.)
| | - Antonio Micari
- Department of Biomedical and Dental Sciences and Morphological and Functional Imaging, University of Messina, A.O.U. Policlinic “G. Martino”, 98165 Messina, Italy;
| | - Natasha Irrera
- Department of Clinical and Experimental Medicine, University of Messina, 98165 Messina, Italy; (M.C.P.); (R.L.); (C.N.); (S.C.); (F.S.); (A.B.); (G.D.B.); (F.C.)
- Correspondence: ; Tel.: +39-090-221-3093; Fax: +39-090-221-23-81
| | - Francesco Costa
- Department of Clinical and Experimental Medicine, University of Messina, 98165 Messina, Italy; (M.C.P.); (R.L.); (C.N.); (S.C.); (F.S.); (A.B.); (G.D.B.); (F.C.)
| |
Collapse
|
13
|
Spinozzi E, Baldassarri C, Acquaticci L, Del Bello F, Grifantini M, Cappellacci L, Riccardo P. Adenosine receptors as promising targets for the management of ocular diseases. Med Chem Res 2021; 30:353-370. [PMID: 33519168 PMCID: PMC7829661 DOI: 10.1007/s00044-021-02704-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 12/01/2020] [Indexed: 12/15/2022]
Abstract
The ocular drug discovery arena has undergone a significant improvement in the last few years culminating in the FDA approvals of 8 new drugs. However, despite a large number of drugs, generics, and combination products available, it remains an urgent need to find breakthrough strategies and therapies for tackling ocular diseases. Targeting the adenosinergic system may represent an innovative strategy for discovering new ocular therapeutics. This review focused on the recent advance in the field and described the numerous nucleoside and non-nucleoside modulators of the four adenosine receptors (ARs) used as potential tools or clinical drug candidates.
Collapse
Affiliation(s)
- Eleonora Spinozzi
- School of Pharmacy Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, 62032 Camerino, Italy
| | - Cecilia Baldassarri
- School of Pharmacy Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, 62032 Camerino, Italy
| | - Laura Acquaticci
- School of Pharmacy Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, 62032 Camerino, Italy
| | - Fabio Del Bello
- School of Pharmacy Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, 62032 Camerino, Italy
| | - Mario Grifantini
- School of Pharmacy Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, 62032 Camerino, Italy
| | - Loredana Cappellacci
- School of Pharmacy Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, 62032 Camerino, Italy
| | - Petrelli Riccardo
- School of Pharmacy Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, 62032 Camerino, Italy
| |
Collapse
|
14
|
Farr SA, Cuzzocrea S, Esposito E, Campolo M, Niehoff ML, Doyle TM, Salvemini D. Adenosine A 3 receptor as a novel therapeutic target to reduce secondary events and improve neurocognitive functions following traumatic brain injury. J Neuroinflammation 2020; 17:339. [PMID: 33183330 PMCID: PMC7659122 DOI: 10.1186/s12974-020-02009-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 10/22/2020] [Indexed: 12/17/2022] Open
Abstract
Background Traumatic brain injury (TBI) is a common pathological condition that presently lacks a specific pharmacological treatment. Adenosine levels rise following TBI, which is thought to be neuroprotective against secondary brain injury. Evidence from stroke and inflammatory disease models suggests that adenosine signaling through the G protein-coupled A3 adenosine receptor (A3AR) can provide antiinflammatory and neuroprotective effects. However, the role of A3AR in TBI has not been investigated. Methods Using the selective A3AR agonist, MRS5980, we evaluated the effects of A3AR activation on the pathological outcomes and cognitive function in CD1 male mouse models of TBI. Results When measured 24 h after controlled cortical impact (CCI) TBI, male mice treated with intraperitoneal injections of MRS5980 (1 mg/kg) had reduced secondary tissue injury and brain infarction than vehicle-treated mice with TBI. These effects were associated with attenuated neuroinflammation marked by reduced activation of nuclear factor of kappa light polypeptide gene enhancer in B cells (NFκB) and MAPK (p38 and extracellular signal-regulated kinase (ERK)) pathways and downstream NOD-like receptor pyrin domain-containing 3 inflammasome activation. MRS5980 also attenuated TBI-induced CD4+ and CD8+ T cell influx. Moreover, when measured 4–5 weeks after closed head weight-drop TBI, male mice treated with MRS5980 (1 mg/kg) performed significantly better in novel object-placement retention tests (NOPRT) and T maze trials than untreated mice with TBI without altered locomotor activity or increased anxiety. Conclusion Our results provide support for the beneficial effects of small molecule A3AR agonists to mitigate secondary tissue injury and cognitive impairment following TBI.
Collapse
Affiliation(s)
- Susan A Farr
- Veterans Affairs Medical Center, 915 N Grand Blvd, St. Louis, MO, 63106, USA.,Department of Internal Medicine, Division of Geriatric Medicine, Saint Louis University School of Medicine, 1402 S. Grand Blvd, St. Louis, MO, 63104, USA.,Department of Pharmacology and Physiology, Saint Louis University School of Medicine, 1402 S. Grand Blvd, St. Louis, MO, 63104, USA.,Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, 1402 S. Grand Blvd, St. Louis, MO, 63104, USA
| | - Salvatore Cuzzocrea
- Department of Clinical and Experimental Medicine and Pharmacology, University of Messina, 98122, Messina, Italy
| | - Emanuela Esposito
- Department of Clinical and Experimental Medicine and Pharmacology, University of Messina, 98122, Messina, Italy
| | - Michela Campolo
- Department of Clinical and Experimental Medicine and Pharmacology, University of Messina, 98122, Messina, Italy
| | - Michael L Niehoff
- Department of Internal Medicine, Division of Geriatric Medicine, Saint Louis University School of Medicine, 1402 S. Grand Blvd, St. Louis, MO, 63104, USA
| | - Timothy M Doyle
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, 1402 S. Grand Blvd, St. Louis, MO, 63104, USA.,Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, 1402 S. Grand Blvd, St. Louis, MO, 63104, USA
| | - Daniela Salvemini
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, 1402 S. Grand Blvd, St. Louis, MO, 63104, USA. .,Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, 1402 S. Grand Blvd, St. Louis, MO, 63104, USA.
| |
Collapse
|
15
|
de Oliveira M, Mathias LS, de Sibio MT, Noronha-Matos JB, Costa MA, Nogueira CR, Correia-de-Sá P. Pitfalls and challenges of the purinergic signaling cascade in obesity. Biochem Pharmacol 2020; 182:114214. [PMID: 32905795 DOI: 10.1016/j.bcp.2020.114214] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/31/2020] [Accepted: 09/01/2020] [Indexed: 12/15/2022]
Abstract
Obesity is a worldwide health problem which have reached pandemic proportions, now also including low and middle-income countries. Excessive or abnormal fat deposition in the abdomen especially in the visceral compartment is tightly associated with a high metabolic risk for arterial hypertension, type II diabetes, cardiovascular diseases, musculoskeletal disorders (especially articular degeneration) and some cancers. Contrariwise, accumulation of fat in the subcutaneous compartment has been associated with a neutral metabolic impact, favoring a lower risk of insulin resistance. Obesity results more often from an avoidable imbalance between food consumption and energy expenditure. There are several recommended strategies for dealing with obesity, including pharmacological therapies, but their success remains incomplete and may not compensate the associated adverse effects. Purinergic signaling operated by ATP and its metabolite, adenosine, has attracted increasing attention in obesity. The extracellular levels of purines often reflect the energy status of a given cell population. Adenine nucleotides and nucleosides fine tuning control adipogenesis and mature adipocytes function via the activation of P2 and P1 purinoceptors, respectively. These features make the purinergic signaling cascade a putative target for therapeutic intervention in obesity and related metabolic syndromes. There are, however, gaps in our knowledge regarding the role of purines in adipocyte precursors differentiation and mature adipocytes functions, as well as their impact among distinct adipose tissue deposits (e.g. white vs. brown, visceral vs. subcutaneous), which warrants further investigations before translation to clinical trials can be made.
Collapse
Affiliation(s)
- Miriane de Oliveira
- São Paulo State University (UNESP), Botucatu Medical School, District of Rubião Jr, s/n, 18618-000, Botucatu, São Paulo, Brazil
| | - Lucas Solla Mathias
- São Paulo State University (UNESP), Botucatu Medical School, District of Rubião Jr, s/n, 18618-000, Botucatu, São Paulo, Brazil
| | - Maria Teresa de Sibio
- São Paulo State University (UNESP), Botucatu Medical School, District of Rubião Jr, s/n, 18618-000, Botucatu, São Paulo, Brazil
| | - José Bernardo Noronha-Matos
- Laboratório de Farmacologia e Neurobiologia, Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP); Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP)
| | - Maria Adelina Costa
- Laboratório de Farmacologia e Neurobiologia, Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP); Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP); Departamento de Química, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP)
| | - Célia Regina Nogueira
- São Paulo State University (UNESP), Botucatu Medical School, District of Rubião Jr, s/n, 18618-000, Botucatu, São Paulo, Brazil
| | - Paulo Correia-de-Sá
- Laboratório de Farmacologia e Neurobiologia, Departamento de Imuno-Fisiologia e Farmacologia, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP); Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP).
| |
Collapse
|
16
|
Clinical value of detecting autoantibodies against β 1-, β 2,- and α 1-adrenergic receptors in carvedilol treatment of patients with heart failure. JOURNAL OF GERIATRIC CARDIOLOGY : JGC 2020; 17:305-312. [PMID: 32670360 PMCID: PMC7338933 DOI: 10.11909/j.issn.1671-5411.2020.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Objective To determine the possible association of anti-β1-adrenergic receptors (anti-β1-AR), anti-β2-AR and anti-α1-AR with carvedilol treatment in patients with heart failure (HF). Methods A total of 267 HF patients were prospectively enrolled. Blood samples were measured by an enzyme-linked immunosorbent assay. All of the patients received carvedilol for their HF. Each patient was followed up for six months and their cardiac function was measured. Results The final analysis encompassed 137 patients comprising 65 patients with three autoantibodies (positive group) and 72 patients without all three autoantibodies but with one or two autoantibodies (negative group). The frequency and geometric mean titer of anti-β1-AR, anti-β2-AR, and anti-α1-AR were significantly lower in the group without all three autoantibodies after six months of carvedilol treatment (all P < 0.01; from 100% to 57%, 50%, and 49%, respectively; and from 1: 118, 1: 138, and 1: 130 to 1: 72, 1: 61, and 1: 67, respectively). Furthermore, 28 patients in the positive group demonstrated complete ablation of autoantibodies. In addition, left ventricular remodelling and function was significantly improved by the use of carvedilol combined with the standard treatment regime for six months in the positive group (P < 0.01) when compared to the negative group (P < 0.05). Conclusions Carvedilol treatment significantly decreases frequency and geometric mean titer in patients with all three autoantibodies, even up to complete ablation, and significantly improved cardiac function and remodelling. The effect of carvedilol is probably correlated to the presence of all three autoantibodies.
Collapse
|
17
|
Doyle TM, Largent-Milnes TM, Chen Z, Staikopoulos V, Esposito E, Dalgarno R, Fan C, Tosh DK, Cuzzocrea S, Jacobson KA, Trang T, Hutchinson MR, Bennett GJ, Vanderah TW, Salvemini D. Chronic Morphine-Induced Changes in Signaling at the A 3 Adenosine Receptor Contribute to Morphine-Induced Hyperalgesia, Tolerance, and Withdrawal. J Pharmacol Exp Ther 2020; 374:331-341. [PMID: 32434943 DOI: 10.1124/jpet.120.000004] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 04/27/2020] [Indexed: 12/11/2022] Open
Abstract
Treating chronic pain by using opioids, such as morphine, is hampered by the development of opioid-induced hyperalgesia (OIH; increased pain sensitivity), antinociceptive tolerance, and withdrawal, which can contribute to dependence and abuse. In the central nervous system, the purine nucleoside adenosine has been implicated in beneficial and detrimental actions of morphine, but the extent of their interaction remains poorly understood. Here, we demonstrate that morphine-induced OIH and antinociceptive tolerance in rats is associated with a twofold increase in adenosine kinase (ADK) expression in the dorsal horn of the spinal cord. Blocking ADK activity in the spinal cord provided greater than 90% attenuation of OIH and antinociceptive tolerance through A3 adenosine receptor (A3AR) signaling. Supplementing adenosine signaling with selective A3AR agonists blocked OIH and antinociceptive tolerance in rodents of both sexes. Engagement of A3AR in the spinal cord with an ADK inhibitor or A3AR agonist was associated with reduced dorsal horn of the spinal cord expression of the NOD-like receptor pyrin domain-containing 3 (60%-75%), cleaved caspase 1 (40%-60%), interleukin (IL)-1β (76%-80%), and tumor necrosis factor (50%-60%). In contrast, the neuroinhibitory and anti-inflammatory cytokine IL-10 increased twofold. In mice, A3AR agonists prevented the development of tolerance in a model of neuropathic pain and reduced naloxone-dependent withdrawal behaviors by greater than 50%. These findings suggest A3AR-dependent adenosine signaling is compromised during sustained morphine to allow the development of morphine-induced adverse effects. These findings raise the intriguing possibility that A3AR agonists may be useful adjunct to opioids to manage their unwanted effects. SIGNIFICANCE STATEMENT: The development of hyperalgesia and antinociceptive tolerance during prolonged opioid use are noteworthy opioid-induced adverse effects that reduce opioid efficacy for treating chronic pain and increase the risk of dependence and abuse. We report that in rodents, these adverse effects are due to reduced adenosine signaling at the A3AR, resulting in NOD-like receptor pyrin domain-containing 3-interleukin-1β neuroinflammation in spinal cord. These effects are attenuated by A3AR agonists, suggesting that A3AR may be a target for therapeutic intervention with selective A3AR agonist as opioid adjuncts.
Collapse
Affiliation(s)
- Timothy M Doyle
- Department of Pharmacology and Physiology and Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, St. Louis, Missouri (T.M.D., Z.C., D.S.); Department of Pharmacology, University of Arizona College of Medicine, Tucson, Arizona (T.M.L.-M., T.W.V.); Discipline of Physiology, Institute for Photonics and Advanced Sensing, ARC Centre of Excellence for Nanoscale BioPhotonics, University of Adelaide, Adelaide, Australia (V.S., M.R.H.); Department of Clinical and Experimental Medicine and Pharmacology, University of Messina, Messina, Italy (E.E., S.C.); Departments of Comparative Biology and Experimental Medicine and Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Canada (R.D., C.F., T.T.); Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland (D.K.T., K.A.J.); and Department of Anesthesiology, University of California San Diego, La Jolla, California (G.J.B.)
| | - Tally M Largent-Milnes
- Department of Pharmacology and Physiology and Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, St. Louis, Missouri (T.M.D., Z.C., D.S.); Department of Pharmacology, University of Arizona College of Medicine, Tucson, Arizona (T.M.L.-M., T.W.V.); Discipline of Physiology, Institute for Photonics and Advanced Sensing, ARC Centre of Excellence for Nanoscale BioPhotonics, University of Adelaide, Adelaide, Australia (V.S., M.R.H.); Department of Clinical and Experimental Medicine and Pharmacology, University of Messina, Messina, Italy (E.E., S.C.); Departments of Comparative Biology and Experimental Medicine and Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Canada (R.D., C.F., T.T.); Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland (D.K.T., K.A.J.); and Department of Anesthesiology, University of California San Diego, La Jolla, California (G.J.B.)
| | - Zhoumou Chen
- Department of Pharmacology and Physiology and Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, St. Louis, Missouri (T.M.D., Z.C., D.S.); Department of Pharmacology, University of Arizona College of Medicine, Tucson, Arizona (T.M.L.-M., T.W.V.); Discipline of Physiology, Institute for Photonics and Advanced Sensing, ARC Centre of Excellence for Nanoscale BioPhotonics, University of Adelaide, Adelaide, Australia (V.S., M.R.H.); Department of Clinical and Experimental Medicine and Pharmacology, University of Messina, Messina, Italy (E.E., S.C.); Departments of Comparative Biology and Experimental Medicine and Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Canada (R.D., C.F., T.T.); Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland (D.K.T., K.A.J.); and Department of Anesthesiology, University of California San Diego, La Jolla, California (G.J.B.)
| | - Vasiliki Staikopoulos
- Department of Pharmacology and Physiology and Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, St. Louis, Missouri (T.M.D., Z.C., D.S.); Department of Pharmacology, University of Arizona College of Medicine, Tucson, Arizona (T.M.L.-M., T.W.V.); Discipline of Physiology, Institute for Photonics and Advanced Sensing, ARC Centre of Excellence for Nanoscale BioPhotonics, University of Adelaide, Adelaide, Australia (V.S., M.R.H.); Department of Clinical and Experimental Medicine and Pharmacology, University of Messina, Messina, Italy (E.E., S.C.); Departments of Comparative Biology and Experimental Medicine and Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Canada (R.D., C.F., T.T.); Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland (D.K.T., K.A.J.); and Department of Anesthesiology, University of California San Diego, La Jolla, California (G.J.B.)
| | - Emanuela Esposito
- Department of Pharmacology and Physiology and Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, St. Louis, Missouri (T.M.D., Z.C., D.S.); Department of Pharmacology, University of Arizona College of Medicine, Tucson, Arizona (T.M.L.-M., T.W.V.); Discipline of Physiology, Institute for Photonics and Advanced Sensing, ARC Centre of Excellence for Nanoscale BioPhotonics, University of Adelaide, Adelaide, Australia (V.S., M.R.H.); Department of Clinical and Experimental Medicine and Pharmacology, University of Messina, Messina, Italy (E.E., S.C.); Departments of Comparative Biology and Experimental Medicine and Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Canada (R.D., C.F., T.T.); Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland (D.K.T., K.A.J.); and Department of Anesthesiology, University of California San Diego, La Jolla, California (G.J.B.)
| | - Rebecca Dalgarno
- Department of Pharmacology and Physiology and Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, St. Louis, Missouri (T.M.D., Z.C., D.S.); Department of Pharmacology, University of Arizona College of Medicine, Tucson, Arizona (T.M.L.-M., T.W.V.); Discipline of Physiology, Institute for Photonics and Advanced Sensing, ARC Centre of Excellence for Nanoscale BioPhotonics, University of Adelaide, Adelaide, Australia (V.S., M.R.H.); Department of Clinical and Experimental Medicine and Pharmacology, University of Messina, Messina, Italy (E.E., S.C.); Departments of Comparative Biology and Experimental Medicine and Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Canada (R.D., C.F., T.T.); Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland (D.K.T., K.A.J.); and Department of Anesthesiology, University of California San Diego, La Jolla, California (G.J.B.)
| | - Churmy Fan
- Department of Pharmacology and Physiology and Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, St. Louis, Missouri (T.M.D., Z.C., D.S.); Department of Pharmacology, University of Arizona College of Medicine, Tucson, Arizona (T.M.L.-M., T.W.V.); Discipline of Physiology, Institute for Photonics and Advanced Sensing, ARC Centre of Excellence for Nanoscale BioPhotonics, University of Adelaide, Adelaide, Australia (V.S., M.R.H.); Department of Clinical and Experimental Medicine and Pharmacology, University of Messina, Messina, Italy (E.E., S.C.); Departments of Comparative Biology and Experimental Medicine and Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Canada (R.D., C.F., T.T.); Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland (D.K.T., K.A.J.); and Department of Anesthesiology, University of California San Diego, La Jolla, California (G.J.B.)
| | - Dilip K Tosh
- Department of Pharmacology and Physiology and Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, St. Louis, Missouri (T.M.D., Z.C., D.S.); Department of Pharmacology, University of Arizona College of Medicine, Tucson, Arizona (T.M.L.-M., T.W.V.); Discipline of Physiology, Institute for Photonics and Advanced Sensing, ARC Centre of Excellence for Nanoscale BioPhotonics, University of Adelaide, Adelaide, Australia (V.S., M.R.H.); Department of Clinical and Experimental Medicine and Pharmacology, University of Messina, Messina, Italy (E.E., S.C.); Departments of Comparative Biology and Experimental Medicine and Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Canada (R.D., C.F., T.T.); Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland (D.K.T., K.A.J.); and Department of Anesthesiology, University of California San Diego, La Jolla, California (G.J.B.)
| | - Salvatore Cuzzocrea
- Department of Pharmacology and Physiology and Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, St. Louis, Missouri (T.M.D., Z.C., D.S.); Department of Pharmacology, University of Arizona College of Medicine, Tucson, Arizona (T.M.L.-M., T.W.V.); Discipline of Physiology, Institute for Photonics and Advanced Sensing, ARC Centre of Excellence for Nanoscale BioPhotonics, University of Adelaide, Adelaide, Australia (V.S., M.R.H.); Department of Clinical and Experimental Medicine and Pharmacology, University of Messina, Messina, Italy (E.E., S.C.); Departments of Comparative Biology and Experimental Medicine and Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Canada (R.D., C.F., T.T.); Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland (D.K.T., K.A.J.); and Department of Anesthesiology, University of California San Diego, La Jolla, California (G.J.B.)
| | - Kenneth A Jacobson
- Department of Pharmacology and Physiology and Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, St. Louis, Missouri (T.M.D., Z.C., D.S.); Department of Pharmacology, University of Arizona College of Medicine, Tucson, Arizona (T.M.L.-M., T.W.V.); Discipline of Physiology, Institute for Photonics and Advanced Sensing, ARC Centre of Excellence for Nanoscale BioPhotonics, University of Adelaide, Adelaide, Australia (V.S., M.R.H.); Department of Clinical and Experimental Medicine and Pharmacology, University of Messina, Messina, Italy (E.E., S.C.); Departments of Comparative Biology and Experimental Medicine and Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Canada (R.D., C.F., T.T.); Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland (D.K.T., K.A.J.); and Department of Anesthesiology, University of California San Diego, La Jolla, California (G.J.B.)
| | - Tuan Trang
- Department of Pharmacology and Physiology and Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, St. Louis, Missouri (T.M.D., Z.C., D.S.); Department of Pharmacology, University of Arizona College of Medicine, Tucson, Arizona (T.M.L.-M., T.W.V.); Discipline of Physiology, Institute for Photonics and Advanced Sensing, ARC Centre of Excellence for Nanoscale BioPhotonics, University of Adelaide, Adelaide, Australia (V.S., M.R.H.); Department of Clinical and Experimental Medicine and Pharmacology, University of Messina, Messina, Italy (E.E., S.C.); Departments of Comparative Biology and Experimental Medicine and Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Canada (R.D., C.F., T.T.); Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland (D.K.T., K.A.J.); and Department of Anesthesiology, University of California San Diego, La Jolla, California (G.J.B.)
| | - Mark R Hutchinson
- Department of Pharmacology and Physiology and Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, St. Louis, Missouri (T.M.D., Z.C., D.S.); Department of Pharmacology, University of Arizona College of Medicine, Tucson, Arizona (T.M.L.-M., T.W.V.); Discipline of Physiology, Institute for Photonics and Advanced Sensing, ARC Centre of Excellence for Nanoscale BioPhotonics, University of Adelaide, Adelaide, Australia (V.S., M.R.H.); Department of Clinical and Experimental Medicine and Pharmacology, University of Messina, Messina, Italy (E.E., S.C.); Departments of Comparative Biology and Experimental Medicine and Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Canada (R.D., C.F., T.T.); Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland (D.K.T., K.A.J.); and Department of Anesthesiology, University of California San Diego, La Jolla, California (G.J.B.)
| | - Gary J Bennett
- Department of Pharmacology and Physiology and Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, St. Louis, Missouri (T.M.D., Z.C., D.S.); Department of Pharmacology, University of Arizona College of Medicine, Tucson, Arizona (T.M.L.-M., T.W.V.); Discipline of Physiology, Institute for Photonics and Advanced Sensing, ARC Centre of Excellence for Nanoscale BioPhotonics, University of Adelaide, Adelaide, Australia (V.S., M.R.H.); Department of Clinical and Experimental Medicine and Pharmacology, University of Messina, Messina, Italy (E.E., S.C.); Departments of Comparative Biology and Experimental Medicine and Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Canada (R.D., C.F., T.T.); Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland (D.K.T., K.A.J.); and Department of Anesthesiology, University of California San Diego, La Jolla, California (G.J.B.)
| | - Todd W Vanderah
- Department of Pharmacology and Physiology and Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, St. Louis, Missouri (T.M.D., Z.C., D.S.); Department of Pharmacology, University of Arizona College of Medicine, Tucson, Arizona (T.M.L.-M., T.W.V.); Discipline of Physiology, Institute for Photonics and Advanced Sensing, ARC Centre of Excellence for Nanoscale BioPhotonics, University of Adelaide, Adelaide, Australia (V.S., M.R.H.); Department of Clinical and Experimental Medicine and Pharmacology, University of Messina, Messina, Italy (E.E., S.C.); Departments of Comparative Biology and Experimental Medicine and Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Canada (R.D., C.F., T.T.); Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland (D.K.T., K.A.J.); and Department of Anesthesiology, University of California San Diego, La Jolla, California (G.J.B.)
| | - Daniela Salvemini
- Department of Pharmacology and Physiology and Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, St. Louis, Missouri (T.M.D., Z.C., D.S.); Department of Pharmacology, University of Arizona College of Medicine, Tucson, Arizona (T.M.L.-M., T.W.V.); Discipline of Physiology, Institute for Photonics and Advanced Sensing, ARC Centre of Excellence for Nanoscale BioPhotonics, University of Adelaide, Adelaide, Australia (V.S., M.R.H.); Department of Clinical and Experimental Medicine and Pharmacology, University of Messina, Messina, Italy (E.E., S.C.); Departments of Comparative Biology and Experimental Medicine and Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Canada (R.D., C.F., T.T.); Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland (D.K.T., K.A.J.); and Department of Anesthesiology, University of California San Diego, La Jolla, California (G.J.B.)
| |
Collapse
|
18
|
The Detrimental Action of Adenosine on Glutamate-Induced Cytotoxicity in PC12 Cells Can Be Shifted towards a Neuroprotective Role through A 1AR Positive Allosteric Modulation. Cells 2020; 9:cells9051242. [PMID: 32443448 PMCID: PMC7290574 DOI: 10.3390/cells9051242] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/14/2020] [Accepted: 05/15/2020] [Indexed: 12/22/2022] Open
Abstract
Glutamate cytotoxicity is implicated in neuronal death in different neurological disorders including stroke, traumatic brain injury, and neurodegenerative diseases. Adenosine is a nucleoside that plays an important role in modulating neuronal activity and its receptors have been identified as promising therapeutic targets for glutamate cytotoxicity. The purpose of this study is to elucidate the role of adenosine and its receptors on glutamate-induced injury in PC12 cells and to verify the protective effect of the novel A1 adenosine receptor positive allosteric modulator, TRR469. Flow cytometry experiments to detect apoptosis revealed that adenosine has a dual role in glutamate cytotoxicity, with A2A and A2B adenosine receptor (AR) activation exacerbating and A1 AR activation improving glutamate-induced cell injury. The overall effect of endogenous adenosine in PC12 cells resulted in a facilitating action on glutamate cytotoxicity, as demonstrated by the use of adenosine deaminase and selective antagonists. However, enhancing the action of endogenous adenosine on A1ARs by TRR469 completely abrogated glutamate-mediated cell death, caspase 3/7 activation, ROS production, and mitochondrial membrane potential loss. Our results indicate a novel potential therapeutic strategy against glutamate cytotoxicity based on the positive allosteric modulation of A1ARs.
Collapse
|
19
|
New Insights into Key Determinants for Adenosine 1 Receptor Antagonists Selectivity Using Supervised Molecular Dynamics Simulations. Biomolecules 2020; 10:biom10050732. [PMID: 32392873 PMCID: PMC7278174 DOI: 10.3390/biom10050732] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 04/27/2020] [Accepted: 04/28/2020] [Indexed: 12/13/2022] Open
Abstract
Adenosine receptors (ARs), like many otherGprotein-coupledreceptors (GPCRs), are targets of primary interest indrug design. However, one of the main limits for the development of drugs for this class of GPCRs is the complex selectivity profile usually displayed by ligands. Numerous efforts have been madefor clarifying the selectivity of ARs, leading to the development of many ligand-based models. The structure of the AR subtype A1 (A1AR) has been recently solved, providing important structural insights. In the present work, we rationalized the selectivity profile of two selective A1AR and A2AAR antagonists, investigating their recognition trajectories obtained by Supervised Molecular Dynamics from an unbound state and monitoring the role of the water molecules in the binding site.
Collapse
|
20
|
Deb PK, Deka S, Borah P, Abed SN, Klotz KN. Medicinal Chemistry and Therapeutic Potential of Agonists, Antagonists and Allosteric Modulators of A1 Adenosine Receptor: Current Status and Perspectives. Curr Pharm Des 2020; 25:2697-2715. [PMID: 31333094 DOI: 10.2174/1381612825666190716100509] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 07/01/2019] [Indexed: 12/28/2022]
Abstract
Adenosine is a purine nucleoside, responsible for the regulation of a wide range of physiological and pathophysiological conditions by binding with four G-protein-coupled receptors (GPCRs), namely A1, A2A, A2B and A3 adenosine receptors (ARs). In particular, A1 AR is ubiquitously present, mediating a variety of physiological processes throughout the body, thus represents a promising drug target for the management of various pathological conditions. Agonists of A1 AR are found to be useful for the treatment of atrial arrhythmia, angina, type-2 diabetes, glaucoma, neuropathic pain, epilepsy, depression and Huntington's disease, whereas antagonists are being investigated for the treatment of diuresis, congestive heart failure, asthma, COPD, anxiety and dementia. However, treatment with full A1 AR agonists has been associated with numerous challenges like cardiovascular side effects, off-target activation as well as desensitization of A1 AR leading to tachyphylaxis. In this regard, partial agonists of A1 AR have been found to be beneficial in enhancing insulin sensitivity and subsequently reducing blood glucose level, while avoiding severe CVS side effects and tachyphylaxis. Allosteric enhancer of A1 AR is found to be potent for the treatment of neuropathic pain, culminating the side effects related to off-target tissue activation of A1 AR. This review provides an overview of the medicinal chemistry and therapeutic potential of various agonists/partial agonists, antagonists and allosteric modulators of A1 AR, with a particular emphasis on their current status and future perspectives in clinical settings.
Collapse
Affiliation(s)
- Pran Kishore Deb
- Faculty of Pharmacy, Philadelphia University, PO Box - 1, 19392, Amman, Jordan
| | - Satyendra Deka
- Pratiksha Institute of Pharmaceutical Sciences, Chandrapur Road, Panikhaiti, Guwahati-26, Assam, India
| | - Pobitra Borah
- Pratiksha Institute of Pharmaceutical Sciences, Chandrapur Road, Panikhaiti, Guwahati-26, Assam, India
| | - Sara N Abed
- Faculty of Pharmacy, Philadelphia University, PO Box - 1, 19392, Amman, Jordan
| | - Karl-Norbert Klotz
- University of Würzburg, Department of Pharmacology and Toxicology Versbacher Str. 9, D-97078 Würzburg, Germany
| |
Collapse
|
21
|
Effendi WI, Nagano T, Kobayashi K, Nishimura Y. Focusing on Adenosine Receptors as a Potential Targeted Therapy in Human Diseases. Cells 2020; 9:E785. [PMID: 32213945 PMCID: PMC7140859 DOI: 10.3390/cells9030785] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 03/21/2020] [Accepted: 03/23/2020] [Indexed: 02/07/2023] Open
Abstract
Adenosine is involved in a range of physiological and pathological effects through membrane-bound receptors linked to G proteins. There are four subtypes of adenosine receptors, described as A1AR, A2AAR, A2BAR, and A3AR, which are the center of cAMP signal pathway-based drug development. Several types of agonists, partial agonists or antagonists, and allosteric substances have been synthesized from these receptors as new therapeutic drug candidates. Research efforts surrounding A1AR and A2AAR are perhaps the most enticing because of their concentration and affinity; however, as a consequence of distressing conditions, both A2BAR and A3AR levels might accumulate. This review focuses on the biological features of each adenosine receptor as the basis of ligand production and describes clinical studies of adenosine receptor-associated pharmaceuticals in human diseases.
Collapse
Affiliation(s)
- Wiwin Is Effendi
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan; (W.I.E.); (K.K.); (Y.N.)
- Department of Pulmonology and Respiratory Medicine, Medical Faculty of Airlangga University, Surabaya 60131, Indonesia
| | - Tatsuya Nagano
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan; (W.I.E.); (K.K.); (Y.N.)
| | - Kazuyuki Kobayashi
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan; (W.I.E.); (K.K.); (Y.N.)
| | - Yoshihiro Nishimura
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan; (W.I.E.); (K.K.); (Y.N.)
| |
Collapse
|
22
|
He X, Wedekind F, Kroll T, Oskamp A, Beer S, Drzezga A, Ermert J, Neumaier B, Bauer A, Elmenhorst D. Image-Derived Input Functions for Quantification of A 1 Adenosine Receptors Availability in Mice Brains Using PET and [ 18F]CPFPX. Front Physiol 2020; 10:1617. [PMID: 32063864 PMCID: PMC7000659 DOI: 10.3389/fphys.2019.01617] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 12/23/2019] [Indexed: 12/28/2022] Open
Abstract
Purpose In vivo imaging for the A1 adenosine receptors (A1ARs) with positron emission tomography (PET) using 8-cyclopentyl-3-(3-[18F]fluoropropyl)-1-propylxan- thine ([18F]CPFPX) has become an important tool for studying physiological processes quantitatively in mice. However, the measurement of arterial input functions (AIFs) on mice is a method with restricted applicability because of the small total blood volume and the related difficulties in withdrawing blood. Therefore, the aim of this study was to extract an appropriate [18F]CPFPX image-derived input function (IDIF) from dynamic PET images of mice. Procedures In this study, five mice were scanned with [18F]CPFPX for 60 min. Arterial blood samples (n = 7 per animal) were collected from the femoral artery and corrected for metabolites. To generate IDIFs, three different approaches were selected: (A) volume of interest (VOI) placed over the heart (cube, 10 mm); (B) VOI set over abdominal vena cava/aorta region with a cuboid (5 × 5 × 15 mm); and (C) with 1 × 1 × 1 mm voxels on five consecutive slices. A calculated scaling factor (α) was used to correct for partial volume effect; the method of obtaining the total metabolite correction of [18F]CPFPX for IDIFs was developed. Three IDIFs were validated by comparison with AIF. Validation included the following: visual performance; computing area under the curve (AUC) ratios (IDIF/AIF) of whole-blood curves and parent curves; and the mean distribution volume (VT) ratios (IDIF/AIF) of A1ARs calculated by Logan plot and two-tissue compartment model. Results Compared with the AIF, the IDIF with VOI over heart showed the best performance among the three IDIFs after scaling by 1.77 (α) in terms of visual analysis, AUC ratios (IDIF/AIF; whole-blood AUC ratio, 1.03 ± 0.06; parent curve AUC ratio, 1.01 ± 0.10) and VT ratios (IDIF/AIF; Logan VT ratio, 1.00 ± 0.17; two-tissue compartment model VT ratio, 1.00 ± 0.13) evaluation. The A1ARs distribution of average parametric images was in good accordance to autoradiography of the mouse brain. Conclusion The proposed study provides evidence that IDIF with VOI over heart can replace AIF effectively for quantification of A1ARs using PET and [18F]CPFPX in mice brains.
Collapse
Affiliation(s)
- Xuan He
- Institut für Neurowissenschaften und Medizin (INM-2), Forschungszentrum Jülich, Jülich, Germany.,Department of Neurophysiology, Institute of Zoology (Bio-II), RWTH Aachen University, Aachen, Germany
| | - Franziska Wedekind
- Institut für Neurowissenschaften und Medizin (INM-2), Forschungszentrum Jülich, Jülich, Germany
| | - Tina Kroll
- Institut für Neurowissenschaften und Medizin (INM-2), Forschungszentrum Jülich, Jülich, Germany
| | - Angela Oskamp
- Institut für Neurowissenschaften und Medizin (INM-2), Forschungszentrum Jülich, Jülich, Germany
| | - Simone Beer
- Institut für Neurowissenschaften und Medizin (INM-2), Forschungszentrum Jülich, Jülich, Germany
| | - Alexander Drzezga
- Institut für Neurowissenschaften und Medizin (INM-2), Forschungszentrum Jülich, Jülich, Germany.,Department of Nuclear Medicine, University Hospital of Cologne, Cologne, Germany
| | - Johannes Ermert
- Institut für Neurowissenschaften und Medizin (INM-5), Forschungszentrum Jülich, Jülich, Germany
| | - Bernd Neumaier
- Institut für Neurowissenschaften und Medizin (INM-5), Forschungszentrum Jülich, Jülich, Germany
| | - Andreas Bauer
- Institut für Neurowissenschaften und Medizin (INM-2), Forschungszentrum Jülich, Jülich, Germany.,Neurological Department, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - David Elmenhorst
- Institut für Neurowissenschaften und Medizin (INM-2), Forschungszentrum Jülich, Jülich, Germany.,Division of Medical Psychology, University of Bonn, Bonn, Germany
| |
Collapse
|
23
|
Dal Ben D, Lambertucci C, Buccioni M, Martí Navia A, Marucci G, Spinaci A, Volpini R. Non-Nucleoside Agonists of the Adenosine Receptors: An Overview. Pharmaceuticals (Basel) 2019; 12:E150. [PMID: 31597388 PMCID: PMC6958362 DOI: 10.3390/ph12040150] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 10/03/2019] [Accepted: 10/05/2019] [Indexed: 12/17/2022] Open
Abstract
Potent and selective adenosine receptor (AR) agonists are of pharmacological interest for the treatment of a wide range of diseases and conditions. Among these derivatives, nucleoside-based agonists represent the great majority of molecules developed and reported to date. However, the limited availability of compounds selective for a specific AR subtype (i.e., A2BAR) and a generally long and complex synthetic route for largely substituted nucleosides are the main drawbacks of this category of molecules. Non-nucleoside agonists represent an alternative set of compounds able to stimulate the AR function and based on simplified structures. This review provides an updated overview on the structural classes of non-nucleoside AR agonists and their biological activities, with emphasis on the main derivatives reported in the literature. A focus is also given to the synthetic routes employed to develop these derivatives and on molecular modeling studies simulating their interaction with ARs.
Collapse
Affiliation(s)
- Diego Dal Ben
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, 62032 Camerino (MC), Italy.
| | - Catia Lambertucci
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, 62032 Camerino (MC), Italy.
| | - Michela Buccioni
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, 62032 Camerino (MC), Italy.
| | - Aleix Martí Navia
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, 62032 Camerino (MC), Italy.
| | - Gabriella Marucci
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, 62032 Camerino (MC), Italy.
| | - Andrea Spinaci
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, 62032 Camerino (MC), Italy.
| | - Rosaria Volpini
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, 62032 Camerino (MC), Italy.
| |
Collapse
|
24
|
Deb PK. Progress in the Development of Agonists, Antagonists and Allosteric Modulators of Adenosine Receptors. Curr Pharm Des 2019; 25:2695-2696. [DOI: 10.2174/138161282525190916100149] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Pran Kishore Deb
- Faculty of Pharmacy, Philadelphia University, PO Box 1, 19392, Amman, Jordan
| |
Collapse
|
25
|
Olivés J, Mestres J. Closing the Gap Between Therapeutic Use and Mode of Action in Remedial Herbs. Front Pharmacol 2019; 10:1132. [PMID: 31632273 PMCID: PMC6785637 DOI: 10.3389/fphar.2019.01132] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 08/30/2019] [Indexed: 12/17/2022] Open
Abstract
The ancient tradition of taking parts of a plant or preparing plant extracts for treating certain discomforts and maladies has long been lacking a scientific rationale to support its preparation and still widespread use in several parts of the world. In an attempt to address this challenge, we collected and integrated data connecting metabolites, plants, diseases, and proteins. A mechanistic hypothesis is generated when a metabolite is known to be present in a given plant, that plant is known to be used to treat a certain disease, that disease is known to be linked to the function of a given protein, and that protein is finally known or predicted to interact with the original metabolite. The construction of plant–protein networks from mutually connected metabolites and diseases facilitated the identification of plausible mechanisms of action for plants being used to treat analgesia, hypercholesterolemia, diarrhea, catarrh, and cough. Additional concrete examples using both experimentally known and computationally predicted, and subsequently experimentally confirmed, metabolite–protein interactions to close the connection circle between metabolites, plants, diseases, and proteins offered further proof of concept for the validity and scope of the approach to generate mode of action hypotheses for some of the therapeutic uses of remedial herbs.
Collapse
Affiliation(s)
- Joaquim Olivés
- Research Group on Systems Pharmacology, Research Programme on Biomedical Informatics (GRIB), IMIM Hospital del Mar Medical Research Institute, Barcelona, Spain
| | - Jordi Mestres
- Research Group on Systems Pharmacology, Research Programme on Biomedical Informatics (GRIB), IMIM Hospital del Mar Medical Research Institute, Barcelona, Spain.,Department of Experimental and Health Sciences, University Pompeu Fabra, Barcelona, Spain
| |
Collapse
|
26
|
New A 2A adenosine receptor antagonists: a structure-based upside-down interaction in the receptor cavity. Bioorg Chem 2019; 92:103183. [PMID: 31446240 DOI: 10.1016/j.bioorg.2019.103183] [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: 05/02/2019] [Revised: 07/16/2019] [Accepted: 08/01/2019] [Indexed: 02/07/2023]
Abstract
Adenosine receptor antagonists are generally based on heterocyclic core structures presenting substituents of various volumes and chemical-physical profiles. Adenine and purine-based adenosine receptor antagonists have been reported in literature. In this work we combined various substituents in the 2, 6, and 8-positions of 9-ethylpurine to depict a structure-affinity relationship analysis at the human adenosine receptors. Compounds were rationally designed trough molecular modeling analysis and then synthesized and evaluated at radioligand binding studies at human adenosine receptors. The new compounds showed affinity for the human adenosine receptors, with some derivatives endowed with low nanomolar Ki data, in particular at the A2AAR subtype. The purine core proves to be a versatile core structure for the development of novel adenosine receptor antagonists with nanomolar affinity for these membrane proteins.
Collapse
|
27
|
Shaik K, Deb PK, Mailavaram RP, Chandrasekaran B, Kachler S, Klotz K, Jaber AMY. 7‐Amino‐2‐aryl/hetero‐aryl‐5‐oxo‐5,8‐dihydro[1,2,4]triazolo[1,5‐a]pyridine‐6‐carbonitriles: Synthesis and adenosine receptor binding studies. Chem Biol Drug Des 2019; 94:1568-1573. [DOI: 10.1111/cbdd.13528] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 02/26/2019] [Accepted: 03/17/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Khasim Shaik
- Pharmaceutical Chemistry Department Shri Vishnu College of Pharmacy Bhimavaram, West Godavari India
| | | | - Raghu Prasad Mailavaram
- Pharmaceutical Chemistry Department Shri Vishnu College of Pharmacy Bhimavaram, West Godavari India
| | | | - Sonja Kachler
- Department of Pharmacology and Toxicology University of Würzburg Würzburg Germany
| | - Karl‐Norbert Klotz
- Department of Pharmacology and Toxicology University of Würzburg Würzburg Germany
| | | |
Collapse
|
28
|
Jacobson KA, Tosh DK, Jain S, Gao ZG. Historical and Current Adenosine Receptor Agonists in Preclinical and Clinical Development. Front Cell Neurosci 2019; 13:124. [PMID: 30983976 PMCID: PMC6447611 DOI: 10.3389/fncel.2019.00124] [Citation(s) in RCA: 136] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 03/13/2019] [Indexed: 12/22/2022] Open
Abstract
Adenosine receptors (ARs) function in the body’s response to conditions of pathology and stress associated with a functional imbalance, such as in the supply and demand of energy/oxygen/nutrients. Extracellular adenosine concentrations vary widely to raise or lower the basal activation of four subtypes of ARs. Endogenous adenosine can correct an energy imbalance during hypoxia and other stress, for example, by slowing the heart rate by A1AR activation or increasing the blood supply to heart muscle by the A2AAR. Moreover, exogenous AR agonists, antagonists, or allosteric modulators can be applied for therapeutic benefit, and medicinal chemists working toward that goal have reported thousands of such agents. Thus, numerous clinical trials have ensued, using promising agents to modulate adenosinergic signaling, most of which have not succeeded. Currently, short-acting, parenteral agonists, adenosine and Regadenoson, are the only AR agonists approved for human use. However, new concepts and compounds are currently being developed and applied toward preclinical and clinical evaluation, and initial results are encouraging. This review focuses on key compounds as AR agonists and positive allosteric modulators (PAMs) for disease treatment or diagnosis. AR agonists for treating inflammation, pain, cancer, non-alcoholic steatohepatitis, angina, sickle cell disease, ischemic conditions and diabetes have been under development. Multiple clinical trials with two A3AR agonists are ongoing.
Collapse
Affiliation(s)
- Kenneth A Jacobson
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Dilip K Tosh
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Shanu Jain
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Zhan-Guo Gao
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
29
|
Tosh DK, Rao H, Bitant A, Salmaso V, Mannes P, Lieberman DI, Vaughan KL, Mattison JA, Rothwell AC, Auchampach JA, Ciancetta A, Liu N, Cui Z, Gao ZG, Reitman ML, Gavrilova O, Jacobson KA. Design and in Vivo Characterization of A 1 Adenosine Receptor Agonists in the Native Ribose and Conformationally Constrained (N)-Methanocarba Series. J Med Chem 2019; 62:1502-1522. [PMID: 30605331 PMCID: PMC6467784 DOI: 10.1021/acs.jmedchem.8b01662] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
(N)-Methanocarba ([3.1.0]bicyclohexyl) adenosines and corresponding ribosides were synthesized to identify novel A1 adenosine receptor (A1AR) agonists for CNS or peripheral applications. Human and mouse AR binding was determined to assess the constrained ring system's A1AR compatibility. N6-Dicyclobutylmethyl ribose agonist (9, MRS7469, >2000-fold selective for A1AR) and known truncated N6-dicyclopropylmethyl methanocarba 7 (MRS5474) were drug-like. The pure diastereoisomer of known riboside 4 displayed high hA1AR selectivity. Methanocarba modification reduced A1AR selectivity of N6-dicyclopropylmethyl and endo-norbornyladenosines but increased ribavirin selectivity. Most analogues tested (ip) were inactive or weak in inducing mouse hypothermia, despite mA1AR full agonism and variable mA3AR efficacy, but strong hypothermia by 9 depended on A1AR, which reflects CNS activity (determined using A1AR or A3AR null mice). Conserved hA1AR interactions were preserved in modeling of 9 and methanocarba equivalent 24 (∼400-fold A1AR-selective). Thus, we identified, and characterized in vivo, ribose and methanocarba nucleosides, including with A1AR-enhancing N6-dicyclobutylmethyl-adenine and 1,2,4-triazole-3-carboxamide (40, MRS7451) nucleobases.
Collapse
Affiliation(s)
- Dilip K. Tosh
- Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Disease, National Institutes of Health, 9000 Rockville Pike, Bethesda, MA, USA 20892
| | - Harsha Rao
- Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Disease, National Institutes of Health, 9000 Rockville Pike, Bethesda, MA, USA 20892
| | - Amelia Bitant
- Department of Pharmacology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, USA 53226
| | - Veronica Salmaso
- Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Disease, National Institutes of Health, 9000 Rockville Pike, Bethesda, MA, USA 20892
| | - Philip Mannes
- Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Disease, National Institutes of Health, 9000 Rockville Pike, Bethesda, MA, USA 20892
| | - David I. Lieberman
- Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Disease, National Institutes of Health, 9000 Rockville Pike, Bethesda, MA, USA 20892
| | - Kelli L. Vaughan
- SoBran BioSciences, SoBran, Inc., 4000 Blackburn Lane, Burtonsville, MD, USA 20866
- Translational Gerontology Branch, National Institute on Aging Intramural Research Program, 16701 Elmer School Rd., Bldg. 103, Dickerson, MD, USA 20842
| | - Julie A. Mattison
- Translational Gerontology Branch, National Institute on Aging Intramural Research Program, 16701 Elmer School Rd., Bldg. 103, Dickerson, MD, USA 20842
| | - Amy C. Rothwell
- Department of Pharmacology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, USA 53226
| | - John A. Auchampach
- Department of Pharmacology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, USA 53226
| | - Antonella Ciancetta
- Queen’s University Belfast, School of Pharmacy, 96 Lisburn Rd, Belfast BT9 7BL, UK
| | - Naili Liu
- Mouse Metabolism Core, National Institute of Diabetes and Digestive and Kidney Disease, National Institutes of Health, 9000 Rockville Pike, Bethesda, MA, USA 20892
| | - Zhenzhong Cui
- Mouse Metabolism Core, National Institute of Diabetes and Digestive and Kidney Disease, National Institutes of Health, 9000 Rockville Pike, Bethesda, MA, USA 20892
| | - Zhan-Guo Gao
- Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Disease, National Institutes of Health, 9000 Rockville Pike, Bethesda, MA, USA 20892
| | - Marc L. Reitman
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Disease, National Institutes of Health, 9000 Rockville Pike, Bethesda, MA, USA 20892
| | - Oksana Gavrilova
- Mouse Metabolism Core, National Institute of Diabetes and Digestive and Kidney Disease, National Institutes of Health, 9000 Rockville Pike, Bethesda, MA, USA 20892
| | - Kenneth A. Jacobson
- Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Disease, National Institutes of Health, 9000 Rockville Pike, Bethesda, MA, USA 20892
| |
Collapse
|
30
|
Jacobson KA, Tosh DK, Jain S, Gao ZG. Historical and Current Adenosine Receptor Agonists in Preclinical and Clinical Development. Front Cell Neurosci 2019. [PMID: 30983976 DOI: 10.3389/fncel.2019.00124/bibtex] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
Adenosine receptors (ARs) function in the body's response to conditions of pathology and stress associated with a functional imbalance, such as in the supply and demand of energy/oxygen/nutrients. Extracellular adenosine concentrations vary widely to raise or lower the basal activation of four subtypes of ARs. Endogenous adenosine can correct an energy imbalance during hypoxia and other stress, for example, by slowing the heart rate by A1AR activation or increasing the blood supply to heart muscle by the A2AAR. Moreover, exogenous AR agonists, antagonists, or allosteric modulators can be applied for therapeutic benefit, and medicinal chemists working toward that goal have reported thousands of such agents. Thus, numerous clinical trials have ensued, using promising agents to modulate adenosinergic signaling, most of which have not succeeded. Currently, short-acting, parenteral agonists, adenosine and Regadenoson, are the only AR agonists approved for human use. However, new concepts and compounds are currently being developed and applied toward preclinical and clinical evaluation, and initial results are encouraging. This review focuses on key compounds as AR agonists and positive allosteric modulators (PAMs) for disease treatment or diagnosis. AR agonists for treating inflammation, pain, cancer, non-alcoholic steatohepatitis, angina, sickle cell disease, ischemic conditions and diabetes have been under development. Multiple clinical trials with two A3AR agonists are ongoing.
Collapse
Affiliation(s)
- Kenneth A Jacobson
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Dilip K Tosh
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Shanu Jain
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Zhan-Guo Gao
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
31
|
Miao Y, Bhattarai A, Nguyen ATN, Christopoulos A, May LT. Structural Basis for Binding of Allosteric Drug Leads in the Adenosine A 1 Receptor. Sci Rep 2018; 8:16836. [PMID: 30442899 PMCID: PMC6237911 DOI: 10.1038/s41598-018-35266-x] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 11/02/2018] [Indexed: 12/12/2022] Open
Abstract
Despite intense interest in designing positive allosteric modulators (PAMs) as selective drugs of the adenosine A1 receptor (A1AR), structural binding modes of the receptor PAMs remain unknown. Using the first X-ray structure of the A1AR, we have performed all-atom simulations using a robust Gaussian accelerated molecular dynamics (GaMD) technique to determine binding modes of the A1AR allosteric drug leads. Two prototypical PAMs, PD81723 and VCP171, were selected. Each PAM was initially placed at least 20 Å away from the receptor. Extensive GaMD simulations using the AMBER and NAMD simulation packages at different acceleration levels captured spontaneous binding of PAMs to the A1AR. The simulations allowed us to identify low-energy binding modes of the PAMs at an allosteric site formed by the receptor extracellular loop 2 (ECL2), which are highly consistent with mutagenesis experimental data. Furthermore, the PAMs stabilized agonist binding in the receptor. In the absence of PAMs at the ECL2 allosteric site, the agonist sampled a significantly larger conformational space and even dissociated from the A1AR alone. In summary, the GaMD simulations elucidated structural binding modes of the PAMs and provided important insights into allostery in the A1AR, which will greatly facilitate the receptor structure-based drug design.
Collapse
Affiliation(s)
- Yinglong Miao
- Center for Computational Biology and Department of Molecular Biosciences, University of Kansas, Lawrence, KS, 66047, USA.
| | - Apurba Bhattarai
- Center for Computational Biology and Department of Molecular Biosciences, University of Kansas, Lawrence, KS, 66047, USA
| | - Anh T N Nguyen
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, VIC, 3052, Australia
| | - Arthur Christopoulos
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, VIC, 3052, Australia
| | - Lauren T May
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, VIC, 3052, Australia
| |
Collapse
|
32
|
Abstract
INTRODUCTION Understanding pathways and mechanisms of drug binding to receptors is important for rational drug design. Remarkable advances in supercomputing and methodological developments have opened a new era for application of computer simulations in predicting drug-receptor interactions at an atomistic level. Gaussian accelerated molecular dynamics (GaMD) is a computational enhanced sampling technique that works by adding a harmonic boost potential to reduce energy barriers. GaMD enables free energy calculations without the requirement of predefined collective variables. GaMD has proven useful in biomolecular simulations, in particular, the prediction of drug-receptor interactions. Areas covered: Herein, the authors review recent GaMD simulation studies that elucidated pathways of drug binding to proteins including the G-protein-coupled receptors and HIV protease. Expert opinion: GaMD is advantageous for enhanced simulations of, amongst many biological processes, drug binding to target receptors. Compared with conventional molecular dynamics, GaMD speeds up biomolecular simulations by orders of magnitude. GaMD enables routine drug binding simulations using personal computers with GPUs or common computing clusters. GaMD and, more broadly, enhanced sampling simulations are expected to dramatically increase our capabilities to determine the mechanisms of drug binding to a wide range of receptors in the near future. This will greatly facilitate computer-aided drug design.
Collapse
Affiliation(s)
- Apurba Bhattarai
- Center for Computational Biology and Department of Molecular Biosciences, University of Kansas, Lawrence, KS 66047, USA,
| | - Yinglong Miao
- Center for Computational Biology and Department of Molecular Biosciences, University of Kansas, Lawrence, KS 66047, USA,
| |
Collapse
|
33
|
Lambertucci C, Marucci G, Dal Ben D, Buccioni M, Spinaci A, Kachler S, Klotz KN, Volpini R. New potent and selective A 1 adenosine receptor antagonists as potential tools for the treatment of gastrointestinal diseases. Eur J Med Chem 2018; 151:199-213. [PMID: 29614417 DOI: 10.1016/j.ejmech.2018.03.067] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 03/21/2018] [Accepted: 03/22/2018] [Indexed: 01/31/2023]
Abstract
The synthesis of 9-alkyl substituted adenine derivatives presenting aromatic groups and cycloalkyl rings in 8- and N6-position, respectively, is reported. The compounds were tested with radioligand binding studies showing, in some cases, a low nanomolar A1 adenosine receptor affinity and a very good selectivity versus the other adenosine receptor subtypes. Functional assays at human adenosine receptors and at a mouse ileum tissue preparation clearly demonstrate the antagonist profile of these molecules, with inhibitory potency at nanomolar level. A molecular modeling study, consisting in docking analysis at the recently reported A1 adenosine receptor crystal structure, was performed for the interpretation of the obtained pharmacological results. The N6-cyclopentyl-9-methyl-8-phenyladenine (17), resulting the most active derivative of the series (Ki = 2.8 nM and IC50 = 14 nM), was also very efficacious in counteracting the effect of the agonist CCPA on mouse ileum contractility. This new compound represents a tool for the development of new agents for the treatment of intestinal diseases as constipation and postoperative ileus.
Collapse
Affiliation(s)
- Catia Lambertucci
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, 62032 Camerino, MC, Italy
| | - Gabriella Marucci
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, 62032 Camerino, MC, Italy
| | - Diego Dal Ben
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, 62032 Camerino, MC, Italy
| | - Michela Buccioni
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, 62032 Camerino, MC, Italy
| | - Andrea Spinaci
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, 62032 Camerino, MC, Italy
| | - Sonja Kachler
- Institut für Pharmakologie und Toxikologie, Universität Würzburg, Versbacher Str. 9, 97078 Würzburg, Germany
| | - Karl-Norbert Klotz
- Institut für Pharmakologie und Toxikologie, Universität Würzburg, Versbacher Str. 9, 97078 Würzburg, Germany
| | - Rosaria Volpini
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, 62032 Camerino, MC, Italy.
| |
Collapse
|
34
|
Dal Ben D, Antonioli L, Lambertucci C, Fornai M, Blandizzi C, Volpini R. Purinergic Ligands as Potential Therapeutic Tools for the Treatment of Inflammation-Related Intestinal Diseases. Front Pharmacol 2018; 9:212. [PMID: 29593540 PMCID: PMC5861216 DOI: 10.3389/fphar.2018.00212] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 02/26/2018] [Indexed: 12/13/2022] Open
Abstract
Inflammation-related intestinal diseases are a set of various conditions presenting an overactive enteric immune system. A continuous overproduction of pro-inflammatory cytokines and a decreased production of anti-inflammatory modulators are generally observed, while morpho-functional alterations of the enteric nervous system lead to intestinal secretory and motor dysfunctions. The factors at the basis of these conditions are still to be totally identified and current therapeutic strategies are aimed only at achieving and maintaining remission states, by using therapeutic tools like aminosalicylates, corticosteroids, immunomodulators, biological drugs (i.e., monoclonal antibodies), and eventually surgery. Recent reports described a key role of purinergic mediators (i.e., adenosine and its nucleotides ATP and ADP) in the regulation of the activity of immune cells and enteric nervous system, showing also that alterations of the purinergic signaling are linked to pathological conditions of the intestinal tract. These data prompted to a series of investigations to test the therapeutic potential for inflammation-related intestinal conditions of compounds able to restore or modulate an altered purinergic signaling within the gut. This review provides an overview on these investigations, describing the results of preclinical and/or clinical evaluation of compounds able to stimulate or inhibit specific P2 (i.e., P2X7) or P1 (i.e., A2A or A3) receptor signaling and to modify the adenosine levels through the modulation of enzymes activity (i.e., Adenosine Deaminase) or nucleoside transporters. Recent developments in the field are also reported and the most promising purine-based therapeutic strategies for the treatment of inflammation-related gastrointestinal disorders are schematically summarized.
Collapse
Affiliation(s)
- Diego Dal Ben
- Medicinal Chemistry Unit, School of Pharmacy, University of Camerino, Camerino, Italy
| | - Luca Antonioli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Catia Lambertucci
- Medicinal Chemistry Unit, School of Pharmacy, University of Camerino, Camerino, Italy
| | - Matteo Fornai
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Corrado Blandizzi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Rosaria Volpini
- Medicinal Chemistry Unit, School of Pharmacy, University of Camerino, Camerino, Italy
| |
Collapse
|
35
|
Varani K, Vincenzi F, Merighi S, Gessi S, Borea PA. Biochemical and Pharmacological Role of A1 Adenosine Receptors and Their Modulation as Novel Therapeutic Strategy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1051:193-232. [DOI: 10.1007/5584_2017_61] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
36
|
Methodical Challenges and a Possible Resolution in the Assessment of Receptor Reserve for Adenosine, an Agonist with Short Half-Life. Molecules 2017; 22:molecules22050839. [PMID: 28534854 PMCID: PMC6154002 DOI: 10.3390/molecules22050839] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 05/05/2017] [Accepted: 05/15/2017] [Indexed: 02/03/2023] Open
Abstract
The term receptor reserve, first introduced and used in the traditional receptor theory, is an integrative measure of response-inducing ability of the interaction between an agonist and a receptor system (consisting of a receptor and its downstream signaling). The underlying phenomenon, i.e., stimulation of a submaximal fraction of receptors can apparently elicit the maximal effect (in certain cases), provides an opportunity to assess the receptor reserve. However, determining receptor reserve is challenging for agonists with short half-lives, such as adenosine. Although adenosine metabolism can be inhibited several ways (in order to prevent the rapid elimination of adenosine administered to construct concentration–effect (E/c) curves for the determination), the consequent accumulation of endogenous adenosine biases the results. To address this problem, we previously proposed a method, by means of which this bias can be mathematically corrected (utilizing a traditional receptor theory-independent approach). In the present investigation, we have offered in silico validation of this method by simulating E/c curves with the use of the operational model of agonism and then by evaluating them using our method. We have found that our method is suitable to reliably assess the receptor reserve for adenosine in our recently published experimental setting, suggesting that it may be capable for a qualitative determination of receptor reserve for rapidly eliminating agonists in general. In addition, we have disclosed a possible interference between FSCPX (8-cyclopentyl-N3-[3-(4-(fluorosulfonyl)benzoyloxy)propyl]-N1-propylxanthine), an irreversible A1 adenosine receptor antagonist, and NBTI (S-(2-hydroxy-5-nitrobenzyl)-6-thioinosine), a nucleoside transport inhibitor, i.e., FSCPX may blunt the effect of NBTI.
Collapse
|
37
|
Role of adenosine receptors in the adipocyte-macrophage interaction during obesity. ACTA ACUST UNITED AC 2017; 64:317-327. [PMID: 28604342 DOI: 10.1016/j.endinu.2017.03.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 03/14/2017] [Accepted: 03/29/2017] [Indexed: 02/04/2023]
Abstract
Lipoinflamation is the inflammation generated in the adipose tissue. It can contribute to the development of insulin resistance. The lipoinflammation-associated mechanisms are related to the function of adipocytes and macrophages present in the adipose tissue. In this regard, the level of nucleoside adenosine is increased in individuals with obesity. Causes or consequences of this increase are unknown. Although, adenosine activating its receptors (A1, A2A, A2B and A3) is able to differentially modulate the function of adipocytes and macrophages, in order to avoid the reduction of insulin sensitivity and generate an anti-inflammatory state in subject with obesity. In this review we propose that adenosine could be a key element in the development of new strategies for limit lipoinflammation and regulate metabolic homeostasis through modulation of adipocyte-macrophage dialogue.
Collapse
|
38
|
Meibom D, Albrecht-Küpper B, Diedrichs N, Hübsch W, Kast R, Krämer T, Krenz U, Lerchen HG, Mittendorf J, Nell PG, Süssmeier F, Vakalopoulos A, Zimmermann K. Neladenoson Bialanate Hydrochloride: A Prodrug of a Partial Adenosine A1Receptor Agonist for the Chronic Treatment of Heart Diseases. ChemMedChem 2017; 12:728-737. [DOI: 10.1002/cmdc.201700151] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 04/12/2017] [Indexed: 11/09/2022]
Affiliation(s)
- Daniel Meibom
- Medicinal Chemistry Wuppertal; Bayer AG; 42113 Wuppertal Germany
| | | | - Nicole Diedrichs
- Project Management, Development; Bayer AG; 42113 Wuppertal Germany
| | - Walter Hübsch
- Medicinal Chemistry Wuppertal; Bayer AG; 42113 Wuppertal Germany
| | - Raimund Kast
- Department of Cardiology Research Wuppertal; Bayer AG; 42113 Wuppertal Germany
| | - Thomas Krämer
- Medicinal Chemistry Wuppertal; Bayer AG; 42113 Wuppertal Germany
| | - Ursula Krenz
- Medicinal Chemistry Wuppertal; Bayer AG; 42113 Wuppertal Germany
| | | | | | - Peter G. Nell
- Medicinal Chemistry Wuppertal; Bayer AG; 42113 Wuppertal Germany
| | - Frank Süssmeier
- Medicinal Chemistry Wuppertal; Bayer AG; 42113 Wuppertal Germany
| | | | | |
Collapse
|
39
|
Schmidt J, Ferk P. Safety issues of compounds acting on adenosinergic signalling. ACTA ACUST UNITED AC 2017; 69:790-806. [PMID: 28397249 DOI: 10.1111/jphp.12720] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 03/04/2017] [Indexed: 12/20/2022]
Abstract
OBJECTIVES Much research has been performed on the field of identifying the roles of adenosine and adenosinergic signalling, but a relatively low number of marketing authorizations have been granted for adenosine receptor (AdR) ligands. In part, this could be related to their safety issues; therefore, our aim was to examine the toxicological and adverse effects data of different compounds acting on adenosinergic signalling, including different AdR ligands and compounds resembling the structure of adenosine. We also wanted to present recent pharmaceutical developments of experimental compounds that showed promising results in clinical trial setting. KEY FINDINGS Safety issues of compounds modulating adenosinergic signalling were investigated, and different mechanisms were presented. Structurally different classes of compounds act on AdRs, the most important being adenosine, adenosine derivatives and other non-nucleoside compounds. Many of them are either not selective enough or are targeting other targets of adenosinergic signalling such as metabolizing enzymes that regulate adenosine levels. Many other targets are also involved that are not part of adenosinergic signalling system such as GABA receptors, different channels, enzymes and others. Some synthetic AdR ligands even showed to be genotoxic. SUMMARY Current review presents safety data of adenosine, adenosine derivatives and other non-nucleoside compounds that modulate adenosinergic signalling. We have presented different mechanisms that participate to an adverse effect or toxic outcome. A separate section also deals with possible organ-specific toxic effects on different in-vitro and in-vivo models.
Collapse
Affiliation(s)
- Jan Schmidt
- Faculty of Chemistry and Chemical Engineering, University of Maribor, Maribor, Slovenia.,Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Polonca Ferk
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
| |
Collapse
|
40
|
Vincenzi F, Ravani A, Pasquini S, Merighi S, Gessi S, Romagnoli R, Baraldi PG, Borea PA, Varani K. Positive allosteric modulation of A1 adenosine receptors as a novel and promising therapeutic strategy for anxiety. Neuropharmacology 2016; 111:283-292. [DOI: 10.1016/j.neuropharm.2016.09.015] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 09/05/2016] [Accepted: 09/13/2016] [Indexed: 11/16/2022]
|
41
|
Burbiel JC, Ghattas W, Küppers P, Köse M, Lacher S, Herzner AM, Kombu RS, Akkinepally RR, Hockemeyer J, Müller CE. 2-Amino[1,2,4]triazolo[1,5-c]quinazolines and Derived Novel Heterocycles: Syntheses and Structure-Activity Relationships of Potent Adenosine Receptor Antagonists. ChemMedChem 2016; 11:2272-2286. [PMID: 27531666 DOI: 10.1002/cmdc.201600255] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 07/29/2016] [Indexed: 11/06/2022]
Abstract
2-Amino[1,2,4]triazolo[1,5-c]quinazolines were identified as potent adenosine receptor (AR) antagonists. Synthetic strategies were devised to gain access to a broad range of derivatives including novel polyheterocyclic compounds. Potent and selective A3 AR antagonists were discovered, including 3,5-diphenyl[1,2,4]triazolo[4,3-c]quinazoline (17, Ki human A3 AR 1.16 nm) and 5'-phenyl-1,2-dihydro-3'H-spiro[indole-3,2'-[1,2,4]triazolo[1,5-c]quinazolin]-2-one (20, Ki human A3 AR 6.94 nm). In addition, multitarget antagonists were obtained, such as the dual A1 /A3 antagonist 2,5-diphenyl[1,2,4]triazolo[1,5-c]quinazoline (13 b, Ki human A1 AR 51.6 nm, human A3 AR 11.1 nm), and the balanced pan-AR antagonists 5-(2-thienyl)[1,2,4]triazolo[1,5-c]quinazolin-2-amine (11 c, Ki human A1 AR 131 nm, A2A AR 32.7 nm, A2B AR 150 nm, A3 AR 47.5 nm) and 9-bromo-5-phenyl[1,2,4]triazolo[1,5-c]quinazolin-2-amine (11 q, Ki human A1 AR 67.7 nm, A2A AR 13.6 nm, A2B AR 75.0 nm, A3 AR 703 nm). In many cases, significantly different affinities for human and rat receptors were observed, which emphasizes the need for caution in extrapolating conclusions between different species.
Collapse
Affiliation(s)
- Joachim C Burbiel
- Pharmazeutische Chemie I, Universität Bonn, Pharma-Zentrum Bonn, Pharmazeutisches Institut, An der Immenburg 4, 53121, Bonn, Germany
| | - Wadih Ghattas
- Pharmazeutische Chemie I, Universität Bonn, Pharma-Zentrum Bonn, Pharmazeutisches Institut, An der Immenburg 4, 53121, Bonn, Germany
| | - Petra Küppers
- Pharmazeutische Chemie I, Universität Bonn, Pharma-Zentrum Bonn, Pharmazeutisches Institut, An der Immenburg 4, 53121, Bonn, Germany
| | - Meryem Köse
- Pharmazeutische Chemie I, Universität Bonn, Pharma-Zentrum Bonn, Pharmazeutisches Institut, An der Immenburg 4, 53121, Bonn, Germany
| | - Svenja Lacher
- Pharmazeutische Chemie I, Universität Bonn, Pharma-Zentrum Bonn, Pharmazeutisches Institut, An der Immenburg 4, 53121, Bonn, Germany
| | - Anna-Maria Herzner
- Pharmazeutische Chemie I, Universität Bonn, Pharma-Zentrum Bonn, Pharmazeutisches Institut, An der Immenburg 4, 53121, Bonn, Germany
| | - Rajan Subramanian Kombu
- University College of Pharmaceutical Sciences, Kakatiya University, 506 009, Warangal, India
| | - Raghuram Rao Akkinepally
- Pharmazeutische Chemie I, Universität Bonn, Pharma-Zentrum Bonn, Pharmazeutisches Institut, An der Immenburg 4, 53121, Bonn, Germany.,University College of Pharmaceutical Sciences, Kakatiya University, 506 009, Warangal, India
| | - Jörg Hockemeyer
- Pharmazeutische Chemie I, Universität Bonn, Pharma-Zentrum Bonn, Pharmazeutisches Institut, An der Immenburg 4, 53121, Bonn, Germany
| | - Christa E Müller
- Pharmazeutische Chemie I, Universität Bonn, Pharma-Zentrum Bonn, Pharmazeutisches Institut, An der Immenburg 4, 53121, Bonn, Germany.
| |
Collapse
|
42
|
Guo D, Heitman LH, IJzerman AP. Kinetic Aspects of the Interaction between Ligand and G Protein-Coupled Receptor: The Case of the Adenosine Receptors. Chem Rev 2016; 117:38-66. [DOI: 10.1021/acs.chemrev.6b00025] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Dong Guo
- Division of Medicinal Chemistry,
Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands
| | - Laura H. Heitman
- Division of Medicinal Chemistry,
Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands
| | - Adriaan P. IJzerman
- Division of Medicinal Chemistry,
Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands
| |
Collapse
|
43
|
Janes K, Symons-Liguori AM, Jacobson KA, Salvemini D. Identification of A3 adenosine receptor agonists as novel non-narcotic analgesics. Br J Pharmacol 2016; 173:1253-67. [PMID: 26804983 DOI: 10.1111/bph.13446] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 11/09/2015] [Accepted: 11/22/2015] [Indexed: 12/26/2022] Open
Abstract
Chronic pain negatively impacts the quality of life in a variety of patient populations. The current therapeutic repertoire is inadequate in managing patient pain and warrants the development of new therapeutics. Adenosine and its four cognate receptors (A1 , A2A , A2B and A3 ) have important roles in physiological and pathophysiological states, including chronic pain. Preclinical and clinical studies have revealed that while adenosine and agonists of the A1 and A2A receptors have antinociceptive properties, their therapeutic utility is limited by adverse cardiovascular side effects. In contrast, our understanding of the A3 receptor is only in its infancy, but exciting preclinical observations of A3 receptor antinociception, which have been bolstered by clinical trials of A3 receptor agonists in other disease states, suggest pain relief without cardiovascular side effects and with sufficient tolerability. Our goal herein is to briefly discuss adenosine and its receptors in the context of pathological pain and to consider the current data regarding A3 receptor-mediated antinociception. We will highlight recent findings regarding the impact of the A3 receptor on pain pathways and examine the current state of selective A3 receptor agonists used for these studies. The adenosine-to-A3 receptor pathway represents an important endogenous system that can be targeted to provide safe, effective pain relief from chronic pain.
Collapse
Affiliation(s)
- K Janes
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - A M Symons-Liguori
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - K A Jacobson
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - D Salvemini
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
44
|
Cagide F, Reis J, Gaspar A, Chavarria D, Kachler S, Klotz KN, Gomes LR, Low JN, Vilar S, Hripcsak G, Borges F. Discovery of the first A 1 adenosine receptor ligand based on the chromone scaffold. RSC Adv 2016. [DOI: 10.1039/c6ra02347a] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The first potent and selective hA1AR ligand based on the chromone scaffold is reported in this work.
Collapse
|
45
|
He W, Wilder T, Cronstein BN. Rolofylline, an adenosine A1 receptor antagonist, inhibits osteoclast differentiation as an inverse agonist. Br J Pharmacol 2014; 170:1167-76. [PMID: 23962057 DOI: 10.1111/bph.12342] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Revised: 06/07/2013] [Accepted: 06/19/2013] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND AND PURPOSE Adenosine may be generated by hydrolysis of extracellular nucleotides by ectonucleotidases, including ectonucleoside triphosphate diphosphohydrolase 1 (CD39), ecto-5'-nucleotidase (CD73), nucleotide pyrophosphatase phosphodiesterase 1 (NPP-1) and tissue non-specific alkaline phosphatase (TNAP). Previous work from our laboratory has uncovered a critical role for adenosine A1 receptors (A1 R) in osteoclastogenesis; blockade or deletion of these receptors diminishes osteoclast differentiation. Interestingly, selective A1 R agonists neither affect basal osteoclastogenesis nor do they reverse A1 R antagonist-mediated inhibition of osteoclastogenesis. In this study, we determined whether ectonucleotidase-mediated adenosine production was required for A1 R antagonist-mediated inhibition, and, when we saw no effect, determined whether A1 R was constitutively activated and the antagonist was acting as an inverse agonist to diminish osteoclast differentiation. EXPERIMENTAL APPROACH Osteoclast formation derived from wild-type, CD39 knockout (KO), CD73 KO, NPP-1 KO and TNAP KO mice was examined by tartrate-resistant acid phosphatase staining of receptor activator of NF-κB ligand-macrophage colony-stimulating factor-stimulated osteoclasts and osteoclast gene expression (Ctsk, Acp5, MMP-9 and NFATc1). Intracellular cAMP concentration was determined by elisa. KEY RESULTS Rolofylline inhibited osteoclast formation in a dose-dependent manner (IC50 = 20-70 nM) in mice lacking all four of these phosphatases, although baseline osteoclast formation was significantly less in precursors from CD73 KO mice. Rolofylline (1 μM) stimulates cAMP production in bone marrow macrophages by 10.23 ± 0.89-fold. CONCLUSIONS AND IMPLICATIONS Based on these findings, we hypothesize that the A1 R is constitutively activated in osteoclast precursors, thereby diminishing basal AC activity, and that A1 R antagonists act as inverse agonists to release A1 R-mediated inhibition of basal AC activity and permit osteoclast differentiation. The constitutive activity of A1 R promotes osteoclast formation and down-regulation of this activity blocks osteoclast formation.
Collapse
Affiliation(s)
- Wenjie He
- New York University School of Medicine, New York, NY, USA
| | | | | |
Collapse
|
46
|
Cortés A, Gracia E, Moreno E, Mallol J, Lluís C, Canela EI, Casadó V. Moonlighting Adenosine Deaminase: A Target Protein for Drug Development. Med Res Rev 2014; 35:85-125. [DOI: 10.1002/med.21324] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Antoni Cortés
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED); Institute of Biomedicine of the University of Barcelona (IBUB); Department of Biochemistry and Molecular Biology; Faculty of Biology; University of Barcelona; Barcelona Spain
| | - Eduard Gracia
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED); Institute of Biomedicine of the University of Barcelona (IBUB); Department of Biochemistry and Molecular Biology; Faculty of Biology; University of Barcelona; Barcelona Spain
| | - Estefania Moreno
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED); Institute of Biomedicine of the University of Barcelona (IBUB); Department of Biochemistry and Molecular Biology; Faculty of Biology; University of Barcelona; Barcelona Spain
| | - Josefa Mallol
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED); Institute of Biomedicine of the University of Barcelona (IBUB); Department of Biochemistry and Molecular Biology; Faculty of Biology; University of Barcelona; Barcelona Spain
| | - Carme Lluís
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED); Institute of Biomedicine of the University of Barcelona (IBUB); Department of Biochemistry and Molecular Biology; Faculty of Biology; University of Barcelona; Barcelona Spain
| | - Enric I. Canela
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED); Institute of Biomedicine of the University of Barcelona (IBUB); Department of Biochemistry and Molecular Biology; Faculty of Biology; University of Barcelona; Barcelona Spain
| | - Vicent Casadó
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED); Institute of Biomedicine of the University of Barcelona (IBUB); Department of Biochemistry and Molecular Biology; Faculty of Biology; University of Barcelona; Barcelona Spain
| |
Collapse
|
47
|
Vincenzi F, Targa M, Romagnoli R, Merighi S, Gessi S, Baraldi PG, Borea PA, Varani K. TRR469, a potent A(1) adenosine receptor allosteric modulator, exhibits anti-nociceptive properties in acute and neuropathic pain models in mice. Neuropharmacology 2014; 81:6-14. [PMID: 24486382 DOI: 10.1016/j.neuropharm.2014.01.028] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Revised: 01/14/2014] [Accepted: 01/20/2014] [Indexed: 01/21/2023]
Abstract
A(1) adenosine receptors (ARs) have been identified as a potential target for the development of anti-nociceptive compounds. The present study explores the analgesic effects of a novel A(1)AR positive allosteric modulator, TRR469, in different models of acute and chronic pain in mice. To evaluate the allosteric enhancement, in vitro binding experiments were performed. The anti-nociceptive properties were investigated in formalin and writhing tests, and in the streptozotocin-induced diabetic neuropathic pain model. Rotarod and catalepsy tests were used to identify potential side effects, while the functional effect of TRR469 was studied using [(3)H]-d-aspartate release from synaptosomes. TRR469 effectively inhibited nociceptive responses in the formalin and writhing tests, with effects comparable to those of the reference analgesic morphine. Isobolographic analysis of the combination of TRR469 and morphine revealed an additive interaction. TRR469 was anti-allodynic in the neuropathic pain model and did not display locomotor or cataleptic side effects. TRR469 enhanced the binding of the agonist radioligand [(3)H]-CCPA and induced a 33-fold increase of adenosine affinity in spinal cord membranes. In mouse spinal cord synaptosomes, TRR469 enhanced the inhibitory effect of A(1)AR activation on [(3)H]-d-aspartate release, a non-metabolizable analogue of glutamate. In conclusion, this research demonstrates the anti-nociceptive effect of the novel compound TRR469, one of the most potent and effective A(1)AR positive allosteric modulators so far synthesized. The use of TRR469 allows for the possibility of exploiting analgesic properties of endogenous adenosine, with a minor potential to develop the various side effects often associated with the use of direct receptor agonists.
Collapse
Affiliation(s)
- Fabrizio Vincenzi
- Department of Medical Sciences, Pharmacology Section, University of Ferrara, via Fossato di Mortara 17/19, 44121 Ferrara, Italy
| | - Martina Targa
- Department of Medical Sciences, Pharmacology Section, University of Ferrara, via Fossato di Mortara 17/19, 44121 Ferrara, Italy
| | - Romeo Romagnoli
- Department of Pharmaceutical Sciences, University of Ferrara, via Fossato di Mortara 17/19, 44121 Ferrara, Italy
| | - Stefania Merighi
- Department of Medical Sciences, Pharmacology Section, University of Ferrara, via Fossato di Mortara 17/19, 44121 Ferrara, Italy
| | - Stefania Gessi
- Department of Medical Sciences, Pharmacology Section, University of Ferrara, via Fossato di Mortara 17/19, 44121 Ferrara, Italy
| | - Pier Giovanni Baraldi
- Department of Pharmaceutical Sciences, University of Ferrara, via Fossato di Mortara 17/19, 44121 Ferrara, Italy
| | - Pier Andrea Borea
- Department of Medical Sciences, Pharmacology Section, University of Ferrara, via Fossato di Mortara 17/19, 44121 Ferrara, Italy
| | - Katia Varani
- Department of Medical Sciences, Pharmacology Section, University of Ferrara, via Fossato di Mortara 17/19, 44121 Ferrara, Italy.
| |
Collapse
|
48
|
Dal Ben D, Buccioni M, Lambertucci C, Kachler S, Falgner N, Marucci G, Thomas A, Cristalli G, Volpini R, Klotz KN. Different efficacy of adenosine and NECA derivatives at the human A3 adenosine receptor: insight into the receptor activation switch. Biochem Pharmacol 2013; 87:321-31. [PMID: 24161786 DOI: 10.1016/j.bcp.2013.10.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Revised: 10/15/2013] [Accepted: 10/15/2013] [Indexed: 01/15/2023]
Abstract
A3 Adenosine receptors are promising drug targets for a number of diseases and intense efforts are dedicated to develop selective agonists and antagonists of these receptors. A series of adenosine derivatives with 2-(ar)-alkynyl chains, with high affinity and different degrees of selectivity for human A3 adenosine receptors was tested for the ability to inhibit forskolin-stimulated adenylyl cyclase. All these derivatives are partial agonists at A3 adenosine receptors; their efficacy is not significantly modified by the introduction of small alkyl substituents in the N(6)-position. In contrast, the adenosine-5'-N-ethyluronamide (NECA) analogs of 2-(ar)-alkynyladenosine derivatives are full A3 agonists. Molecular modeling analyses were performed considering both the conformational behavior of the ligands and the impact of 2- and 5'-substituents on ligand-target interaction. The results suggest an explanation for the different agonistic behavior of adenosine and NECA derivatives, respectively. A sub-pocket of the binding site was analyzed as a crucial interaction domain for receptor activation.
Collapse
Affiliation(s)
- Diego Dal Ben
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, I-62032 Camerino, Italy
| | - Michela Buccioni
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, I-62032 Camerino, Italy
| | - Catia Lambertucci
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, I-62032 Camerino, Italy
| | - Sonja Kachler
- Universität Würzburg, Institut für Pharmakologie und Toxikologie, Versbacher Str. 9, D-97078, Würzburg, Germany
| | - Nico Falgner
- Universität Würzburg, Institut für Pharmakologie und Toxikologie, Versbacher Str. 9, D-97078, Würzburg, Germany
| | - Gabriella Marucci
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, I-62032 Camerino, Italy
| | - Ajiroghene Thomas
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, I-62032 Camerino, Italy
| | - Gloria Cristalli
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, I-62032 Camerino, Italy
| | - Rosaria Volpini
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, I-62032 Camerino, Italy
| | - Karl-Norbert Klotz
- Universität Würzburg, Institut für Pharmakologie und Toxikologie, Versbacher Str. 9, D-97078, Würzburg, Germany.
| |
Collapse
|
49
|
Abstract
The pancreas is an organ with a central role in nutrient breakdown, nutrient sensing and release of hormones regulating whole body nutrient homeostasis. In diabetes mellitus, the balance is broken-cells can be starving in the midst of plenty. There are indications that the incidence of diabetes type 1 and 2, and possibly pancreatogenic diabetes, is rising globally. Events leading to insulin secretion and action are complex, but there is emerging evidence that intracellular nucleotides and nucleotides are not only important as intracellular energy molecules but also as extracellular signalling molecules in purinergic signalling cascades. This signalling takes place at the level of the pancreas, where the close apposition of various cells-endocrine, exocrine, stromal and immune cells-contributes to the integrated function. Following an introduction to diabetes, the pancreas and purinergic signalling, we will focus on the role of purinergic signalling and its changes associated with diabetes in the pancreas and selected tissues/organ systems affected by hyperglycaemia and other stress molecules of diabetes. Since this is the first review of this kind, a comprehensive historical angle is taken, and common and divergent roles of receptors for nucleotides and nucleosides in different organ systems will be given. This integrated picture will aid our understanding of the challenges of the potential and currently used drugs targeted to specific organ/cells or disorders associated with diabetes.
Collapse
Affiliation(s)
- Geoffrey Burnstock
- Autonomic Neuroscience Centre, University College Medical School, Rowland Hill Street, London, NW3 2PF UK
- Department of Pharmacology, Melbourne University, Melbourne, Australia
| | - Ivana Novak
- Molecular and Integrative Physiology, Department of Biology, University of Copenhagen, August Krogh Building, Universitetsparken 13, 2100 Copenhagen Ø, Denmark
| |
Collapse
|
50
|
Elmenhorst D, Kroll T, Wedekind F, Weisshaupt A, Beer S, Bauer A. In vivo kinetic and steady-state quantification of 18F-CPFPX binding to rat cerebral A1 adenosine receptors: validation by displacement and autoradiographic experiments. J Nucl Med 2013; 54:1411-9. [PMID: 23740103 DOI: 10.2967/jnumed.112.115576] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED In vivo imaging of the A1 adenosine receptor (A1AR) using (18)F-8-cyclopentyl-3-(3-fluoropropyl)-1-propylxanthine ((18)F-CPFPX) and PET has become an important tool for studying physiologic and pathologic states of the human brain. However, dedicated experimental settings for small-animal studies are still lacking. The aim of the present study was therefore to develop and evaluate suitable pharmacokinetic models for the quantification of the cerebral A1AR in high-resolution PET. METHODS On a dedicated animal PET scanner, 15 rats underwent (18)F-CPFPX PET scans of 120-min duration. In all animals, arterial blood samples were drawn and corrected for metabolites. The radioligand was injected either as a bolus or as a bolus plus constant infusion. For the definition of unspecific binding, the A1AR selective antagonist 8-cyclopentyl-1,3-dipropylxanthine (DPCPX) was applied. After PET, the brains of 9 animals were dissected and in vitro saturation binding was performed using high-resolution (3)H-DPCPX autoradiography. RESULTS The kinetics of (18)F-CPFPX were well described by either compartmental or noncompartmental models based on arterial input function. The resulting distribution volume ratio correlated with a low bias toward identity with the binding potential derived from a reference region (olfactory bulb) approach. Furthermore, PET quantification correlated significantly with autoradiographic in vitro data. Blockade of the A1AR with DPCPX identified specific binding of about 45% in the reference region olfactory bulb. CONCLUSION The present study provides evidence that (18)F-CPFPX PET based on a reference tissue approach can be performed quantitatively in rodents in selected applications. Specific binding in the reference region needs careful consideration for quantitative investigations.
Collapse
Affiliation(s)
- David Elmenhorst
- Institute of Neuroscience and Medicine, INM-2, Forschungszentrum Jülich, Jülich, Germany.
| | | | | | | | | | | |
Collapse
|