1
|
Ash J, Curtis K, Bhowmik S, Odoh SO, Kang JY. Hydropersulfide (RSSH)-Mediated Hydrothiolation of Alkenes. J Org Chem 2025. [PMID: 39749719 DOI: 10.1021/acs.joc.4c02309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
A hydropersulfide-mediated hydrothiolation reaction of alkenes has been developed for C-S bond formation with Markovnikov selectivity. This new approach is a transition-metal-, additive-, and solvent-free reaction under mild conditions. The reaction is postulated to proceed by an ionic mechanism with the release of elemental sulfur based on our control experiments and density functional theory calculations.
Collapse
Affiliation(s)
- Jeffrey Ash
- Department of Chemistry and Biochemistry, University of Nevada Las Vegas, 4505 South Maryland Parkway, Las Vegas, Nevada 89154-4003, United States
| | - Kevin Curtis
- Department of Chemistry, University of Nevada Reno, 1664 North Virginia Street, Reno, Nevada 89557-0705, United States
| | - Shishir Bhowmik
- Department of Chemistry and Biochemistry, University of Nevada Las Vegas, 4505 South Maryland Parkway, Las Vegas, Nevada 89154-4003, United States
| | - Samuel O Odoh
- Department of Chemistry, University of Nevada Reno, 1664 North Virginia Street, Reno, Nevada 89557-0705, United States
| | - Jun Yong Kang
- Department of Chemistry and Biochemistry, University of Nevada Las Vegas, 4505 South Maryland Parkway, Las Vegas, Nevada 89154-4003, United States
| |
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
|
Synthesis of 2-chloropurine ribosides with chiral amino acid amides at C6 and their evaluation as A1 adenosine receptor agonists. Bioorg Chem 2022; 126:105878. [DOI: 10.1016/j.bioorg.2022.105878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 05/11/2022] [Accepted: 05/14/2022] [Indexed: 11/23/2022]
|
4
|
Xu C, Zhou W, Dong G, Qiao H, Peng J, Jia P, Li Y, Liu H, Sun K, Zhao W. Novel [1,2,3]triazolo[4,5-d]pyrimidine derivatives containing hydrazone fragment as potent and selective anticancer agents. Bioorg Chem 2020; 105:104424. [PMID: 33161253 DOI: 10.1016/j.bioorg.2020.104424] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 09/18/2020] [Accepted: 10/20/2020] [Indexed: 01/22/2023]
Abstract
In this paper, based on molecular hybridization, a series of [1,2,3]triazolo[4,5-d]pyrimidine derivatives containing hydrazine was synthesized and their antiproliferative activities against 5 cancer cell lines (MGC-803, PC3, PC9, EC9706 and SMMC-7721) were evaluated. We found that most of them exhibited obvious growth inhibition effects on these tested cancer cells, especially compound 34 on PC3 cells (IC50 = 26.25 ± 0.28 nM). Meanwhile, compound 34 displayed best selectivity on PC3, compared with the other cancer cell lines, as well as excellent selectivity towards normal cell lines (Het-1A, L02 and GES-1). Further investigations demonstrated that 34 could significantly inhibit PC3 cells' colony formation, increase cellular ROS content, suppress EGFR expression and induce apoptosis. Our findings indicate that 34 may serve as a novel lead compound for the discovery of more triazolopyrimidine derivatives with improved anticancer potency and selectivity.
Collapse
Affiliation(s)
- Chenhao Xu
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University School of Pharmaceutical Sciences and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Wenjuan Zhou
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University School of Pharmaceutical Sciences and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Department of Pathology, Oslo University Hospital, Faculty of Medicine, University of Oslo, Oslo 0379, Norway
| | - Guanjun Dong
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University School of Pharmaceutical Sciences and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Hui Qiao
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University School of Pharmaceutical Sciences and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Jiadi Peng
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University School of Pharmaceutical Sciences and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Pengfei Jia
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University School of Pharmaceutical Sciences and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Yuhao Li
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University School of Pharmaceutical Sciences and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Hongmin Liu
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University School of Pharmaceutical Sciences and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Kai Sun
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University School of Pharmaceutical Sciences and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China.
| | - Wen Zhao
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou University School of Pharmaceutical Sciences and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China.
| |
Collapse
|
5
|
Heckmann CM, Gourlay LJ, Dominguez B, Paradisi F. An ( R)-Selective Transaminase From Thermomyces stellatus: Stabilizing the Tetrameric Form. Front Bioeng Biotechnol 2020; 8:707. [PMID: 32793563 PMCID: PMC7387707 DOI: 10.3389/fbioe.2020.00707] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 06/05/2020] [Indexed: 12/17/2022] Open
Abstract
The identification and 3D structural characterization of a homolog of the (R)-selective transaminase (RTA) from Aspergillus terreus (AtRTA), from the thermotolerant fungus Thermomyces stellatus (TsRTA) is here reported. The thermostability of TsRTA (40% retained activity after 7 days at 40°C) was initially attributed to its tetrameric form in solution, however subsequent studies of AtRTA revealed it also exists predominantly as a tetramer yet, at 40°C, it is inactivated within 48 h. The engineering of a cysteine residue to promote disulfide bond formation across the dimer-dimer interface stabilized both enzymes, with TsRTA_G205C retaining almost full activity after incubation at 50°C for 7 days. Thus, the role of this mutation was elucidated and the importance of stabilizing the tetramer for overall stability of RTAs is highlighted. TsRTA accepts the common amine donors (R)-methylbenzylamine, isopropylamine, and d-alanine as well as aromatic and aliphatic ketones and aldehydes.
Collapse
Affiliation(s)
| | - Louise J. Gourlay
- Department of Biosciences, Università degli Studi di Milano, Milan, Italy
| | | | - Francesca Paradisi
- School of Chemistry, University of Nottingham, Nottingham, United Kingdom
- Department of Chemistry and Biochemistry, University of Bern, Bern, Switzerland
| |
Collapse
|
6
|
The adenosine A1 receptor agonist WAG 994 suppresses acute kainic acid-induced status epilepticus in vivo. Neuropharmacology 2020; 176:108213. [PMID: 32615188 DOI: 10.1016/j.neuropharm.2020.108213] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 06/10/2020] [Accepted: 06/12/2020] [Indexed: 12/22/2022]
Abstract
Status epilepticus (SE) is a neurological emergency characterized by continuous seizure activity lasting longer than 5 min, often with no recovery between seizures (Trinka et al., 2015). SE is refractory to benzodiazepine and second-line treatments in about 30% cases. Novel treatment approaches are urgently needed as refractory SE is associated with mortality rates of up to 70%. Robust adenosinergic anticonvulsant effects have been known for decades, but translation into seizure treatments was hampered by cardiovascular side effects. However, the selective adenosine A1 receptor agonist SDZ WAG 994 (WAG) displays diminished cardiovascular side effects compared to classic A1R agonists and was safely administered systemically in human clinical trials. Here, we investigate the anticonvulsant efficacy of WAG in vitro and in vivo. WAG robustly inhibited high-K+-induced continuous epileptiform activity in rat hippocampal slices (IC50 = 52.5 nM). Importantly, WAG acutely suppressed SE in vivo induced by kainic acid (20 mg/kg i.p.) in mice. After SE was established, mice received three i.p. injections of WAG or diazepam (DIA, 5 mg/kg). Interestingly, DIA did not attenuate SE while the majority of WAG-treated mice (1 mg/kg) were seizure-free after three injections. Anticonvulsant effects were retained when a lower dose of WAG (0.3 mg/kg) was used. Importantly, all WAG-treated mice survived kainic acid induced SE. In summary, we report for the first time that an A1R agonist with an acceptable human side-effect profile can acutely suppress established SE in vivo. Our results suggest that WAG stops or vastly attenuates SE while DIA fails to mitigate SE in this model.
Collapse
|
7
|
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: 6.6] [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
|
8
|
Chen FF, Zhang YH, Zhang ZJ, Liu L, Wu JP, Xu JH, Zheng GW. An Ammonium-Formate-Driven Trienzymatic Cascade for ω-Transaminase-Catalyzed (R)-Selective Amination. J Org Chem 2019; 84:14987-14993. [DOI: 10.1021/acs.joc.9b02445] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Fei-Fei Chen
- State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Yu-Hui Zhang
- State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Zhi-Jun Zhang
- State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Lei Liu
- State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Jian-Ping Wu
- Institute of Bioengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China
| | - Jian-He Xu
- State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Gao-Wei Zheng
- State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| |
Collapse
|
9
|
Wang Y, Wang K, Cao W, Liu X, Feng X. Diastereo- and Enantioselective 1,6-Conjugate Addition of 2-Azaarylacetamides to para-Quinone Methides. Org Lett 2019; 21:6063-6067. [PMID: 31334665 DOI: 10.1021/acs.orglett.9b02215] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
A chiral Mg(II)/N,N'-dioxide complex was found to be highly effective in promoting the diastereo- and enantioselective 1,6-conjugate addition of various azaarylacetamides to p-quinone methides under mild reaction conditions. Various chiral azaarene derivatives containing gem(1,1)-diaryl skeletons were obtained in good yields, dr, and excellent ee values. Meanwhile, on the basis of insight gained from the control experiments as well as the single-crystal structure of the Mg(II)/N,N'-dioxide complex and product, a possible transition state was proposed to explain the origin of coordination and stereoinduction.
Collapse
Affiliation(s)
- Yan Wang
- Key Laboratory of Green Chemistry & Technology, Ministry of Education, College of Chemistry , Sichuan University , Chengdu 610064 , China
| | - Kaixuan Wang
- Key Laboratory of Green Chemistry & Technology, Ministry of Education, College of Chemistry , Sichuan University , Chengdu 610064 , China
| | - Weidi Cao
- Key Laboratory of Green Chemistry & Technology, Ministry of Education, College of Chemistry , Sichuan University , Chengdu 610064 , China
| | - Xiaohua Liu
- Key Laboratory of Green Chemistry & Technology, Ministry of Education, College of Chemistry , Sichuan University , Chengdu 610064 , China
| | - Xiaoming Feng
- Key Laboratory of Green Chemistry & Technology, Ministry of Education, College of Chemistry , Sichuan University , Chengdu 610064 , China
| |
Collapse
|
10
|
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: 22.7] [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
|
11
|
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: 3.3] [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
|
12
|
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
|
13
|
Zafar S, Jabeen I. Structure, Function, and Modulation of γ-Aminobutyric Acid Transporter 1 (GAT1) in Neurological Disorders: A Pharmacoinformatic Prospective. Front Chem 2018; 6:397. [PMID: 30255012 PMCID: PMC6141625 DOI: 10.3389/fchem.2018.00397] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 08/20/2018] [Indexed: 02/03/2023] Open
Abstract
γ-Aminobutyric acid (GABA) Transporters (GATs) belong to sodium and chloride dependent-transporter family and are widely expressed throughout the brain. Notably, GAT1 is accountable for sustaining 75% of the synaptic GABA concentration and entails its transport to the GABAA receptors to initiate the receptor-mediated inhibition of post-synaptic neurons. Imbalance in ion homeostasis has been associated with several neurological disorders related to the GABAergic system. However, inhibition of the GABA uptake by these transporters has been accepted as an effective approach to enhance GABAergic inhibitory neurotransmission in the treatment of seizures in epileptic and other neurological disorders. Here, we reviewed computational methodologies including molecular modeling, docking, and molecular dynamic simulations studies to underscore the structure and function of GAT1 in the GABAergic system. Additionally, various SAR and QSAR methodologies have been reviewed to probe the 3D structural features of inhibitors required to modulate GATs activity. Overall, present review provides an overview of crucial role of GAT1 in GABAergic system and its modulation to evade neurological disorders.
Collapse
Affiliation(s)
| | - Ishrat Jabeen
- Research Center for Modeling and Simulation, National University of Sciences and Technology, Islamabad, Pakistan
| |
Collapse
|
14
|
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: 1.6] [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
|
15
|
Petrelli R, Scortichini M, Belardo C, Boccella S, Luongo L, Capone F, Kachler S, Vita P, Del Bello F, Maione S, Lavecchia A, Klotz KN, Cappellacci L. Structure-Based Design, Synthesis, and In Vivo Antinociceptive Effects of Selective A1 Adenosine Receptor Agonists. J Med Chem 2018; 61:305-318. [DOI: 10.1021/acs.jmedchem.7b01399] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Riccardo Petrelli
- School
of Pharmacy, Medicinal Chemistry Unit, University of Camerino, 62032 Camerino, Italy
| | - Mirko Scortichini
- School
of Pharmacy, Medicinal Chemistry Unit, University of Camerino, 62032 Camerino, Italy
| | - Carmela Belardo
- Section
of Pharmacology “L. Donatelli”, Department of Experimental
Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy
| | - Serena Boccella
- Section
of Pharmacology “L. Donatelli”, Department of Experimental
Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy
| | - Livio Luongo
- Section
of Pharmacology “L. Donatelli”, Department of Experimental
Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy
| | - Fabio Capone
- Department
of Pharmacy, “Drug Discovery” Laboratory, University of Naples Federico II, 80131 Naples, Italy
| | - Sonja Kachler
- Institut
für Pharmakologie and Toxikologie, Universität Würzburg, D-97078 Würzburg, Germany
| | - Patrizia Vita
- School
of Pharmacy, Medicinal Chemistry Unit, University of Camerino, 62032 Camerino, Italy
| | - Fabio Del Bello
- School
of Pharmacy, Medicinal Chemistry Unit, University of Camerino, 62032 Camerino, Italy
| | - Sabatino Maione
- Section
of Pharmacology “L. Donatelli”, Department of Experimental
Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy
| | - Antonio Lavecchia
- Department
of Pharmacy, “Drug Discovery” Laboratory, University of Naples Federico II, 80131 Naples, Italy
| | - Karl-Norbert Klotz
- Institut
für Pharmakologie and Toxikologie, Universität Würzburg, D-97078 Würzburg, Germany
| | - Loredana Cappellacci
- School
of Pharmacy, Medicinal Chemistry Unit, University of Camerino, 62032 Camerino, Italy
| |
Collapse
|
16
|
Lepitre T, Denhez C, Sanselme M, Othman M, Lawson AM, Daïch A. Original Mitsunobu-Triggered Sequence Involved in a One-Pot Domino Process toward Tetracyclic Systems Bearing a Bis-N,O-acetal Junction. J Org Chem 2016; 81:8837-8849. [DOI: 10.1021/acs.joc.6b01523] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Thomas Lepitre
- Normandie Univ,
France, UNILEHAVRE, URCOM, EA 3221, FR 3038 CNRS, F-76600 Le Havre, France
| | - Clement Denhez
- Université
de Reims Champagne Ardenne, Institut de Chimie Moléculaire
de Reims, CNRS UMR 7312, UFR de Pharmacie, 51 rue Cognacq-Jay, 51096 Cedex Reims, France
- Multiscale
Molecular Modeling Platform, Université de Reims Champagne-Ardenne, UFR Sciences Exactes et Naturelles, F-51687 Cedex 2 Reims, France
| | - Morgane Sanselme
- Normandie Univ,
France; UNIROUEN, Laboratoire SMS EA3233, 1 rue Tesniere, France, F-76821 Mont Saint Aignan, France
| | - Mohamed Othman
- Normandie Univ,
France, UNILEHAVRE, URCOM, EA 3221, FR 3038 CNRS, F-76600 Le Havre, France
| | - Ata Martin Lawson
- Normandie Univ,
France, UNILEHAVRE, URCOM, EA 3221, FR 3038 CNRS, F-76600 Le Havre, France
| | - Adam Daïch
- Normandie Univ,
France, UNILEHAVRE, URCOM, EA 3221, FR 3038 CNRS, F-76600 Le Havre, France
| |
Collapse
|
17
|
Knight A, Hemmings JL, Winfield I, Leuenberger M, Frattini E, Frenguelli BG, Dowell SJ, Lochner M, Ladds G. Discovery of Novel Adenosine Receptor Agonists That Exhibit Subtype Selectivity. J Med Chem 2016; 59:947-64. [DOI: 10.1021/acs.jmedchem.5b01402] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Anthony Knight
- Systems
Biology Doctoral Training Centre, University of Warwick, Coventry CV4 7AL, U.K
| | - Jennifer L. Hemmings
- Department
of Chemistry and Biochemistry, University of Bern, 3012 Bern, Switzerland
| | - Ian Winfield
- Division
of Biomedical Cell Biology, Warwick Medical School, University of Warwick, Coventry CV4 7AL, U.K
- Department
of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, U.K
| | - Michele Leuenberger
- Department
of Chemistry and Biochemistry, University of Bern, 3012 Bern, Switzerland
| | - Eugenia Frattini
- Department
of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, U.K
| | | | - Simon J. Dowell
- Department
of Platform Technology and Science, GlaxoSmithKline, Hertfordshire SG1 2NY, U.K
| | - Martin Lochner
- Department
of Chemistry and Biochemistry, University of Bern, 3012 Bern, Switzerland
| | - Graham Ladds
- Department
of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, U.K
| |
Collapse
|
18
|
Jacobson KA, Gao ZG, Paoletta S, Kiselev E, Chakraborty S, Jayasekara PS, Balasubramanian R, Tosh DK. John Daly Lecture: Structure-guided Drug Design for Adenosine and P2Y Receptors. Comput Struct Biotechnol J 2014; 13:286-98. [PMID: 25973142 PMCID: PMC4423517 DOI: 10.1016/j.csbj.2014.10.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 10/08/2014] [Accepted: 10/13/2014] [Indexed: 02/02/2023] Open
Abstract
We establish structure activity relationships of extracellular nucleosides and nucleotides at G protein-coupled receptors (GPCRs), e.g. adenosine receptors (ARs) and P2Y receptors (P2YRs), respectively. We synthesize selective agents for use as pharmacological probes and potential therapeutic agents (e.g. A3AR agonists for neuropathic pain). Detailed structural information derived from the X-ray crystallographic structures within these families enables the design of novel ligands, guides modification of known agonists and antagonists, and helps predict polypharmacology. Structures were recently reported for the P2Y12 receptor (P2Y12R), an anti-thrombotic target. Comparison of agonist-bound and antagonist-bound P2Y12R indicates unprecedented structural plasticity in the outer portions of the transmembrane (TM) domains and the extracellular loops. Nonphosphate-containing ligands of the P2YRs, such as the selective P2Y14R antagonist PPTN, are desired for bioavailability and increased stability. Also, A2AAR structures are effectively applied to homology modeling of closely related A1AR and A3AR, which are not yet crystallized. Conformational constraint of normally flexible ribose with bicyclic analogues increased the ligand selectivity. Comparison of rigid A3AR agonist congeners allows the exploration of interaction of specific regions of the nucleoside analogues with the target and off-target GPCRs, such as biogenic amine receptors. Molecular modeling predicts plasticity of the A3AR at TM2 to accommodate highly rigidified ligands. Novel fluorescent derivatives of high affinity GPCR ligands are useful tool compounds for characterization of receptors and their oligomeric assemblies. Fluorescent probes are useful for characterization of GPCRs in living cells by flow cytometry and other methods. Thus, 3D knowledge of receptor binding and activation facilitates drug discovery.
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 20892 USA
| | - Zhan-Guo Gao
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892 USA
| | - Silvia Paoletta
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892 USA
| | - Evgeny Kiselev
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892 USA
| | - Saibal Chakraborty
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892 USA
| | - P Suresh Jayasekara
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892 USA
| | - Ramachandran Balasubramanian
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892 USA
| | - 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 20892 USA
| |
Collapse
|
19
|
Talwar D, Poyatos Salguero N, Robertson CM, Xiao J. Primary amines by transfer hydrogenative reductive amination of ketones by using cyclometalated Ir(III) catalysts. Chemistry 2014; 20:245-52. [PMID: 24516890 DOI: 10.1002/chem.201303541] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Cyclometalated iridium complexes are found to be versatile catalysts for the direct reductive amination (DRA) of carbonyls to give primary amines under transfer-hydrogenation conditions with ammonium formate as both the nitrogen and hydrogen source. These complexes are easy to synthesise and their ligands can be easily tuned. The activity and chemoselectivity of the catalyst towards primary amines is excellent, with a substrate to catalyst ratio (S/C) of 1000 being feasible. Both aromatic and aliphatic primary amines were obtained in high yields. Moreover, a first example of homogeneously catalysed transfer-hydrogenative DRA has been realised for β-keto ethers, leading to the corresponding β-amino ethers. In addition, non-natural α-amino acids could also be obtained in excellent yields with this method.
Collapse
|
20
|
Tosh DK, Paoletta S, Deflorian F, Phan K, Moss SM, Gao ZG, Jiang X, Jacobson KA. Structural sweet spot for A1 adenosine receptor activation by truncated (N)-methanocarba nucleosides: receptor docking and potent anticonvulsant activity. J Med Chem 2012; 55:8075-90. [PMID: 22921089 PMCID: PMC3463139 DOI: 10.1021/jm300965a] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
A(1) adenosine receptor (AR) agonists display antiischemic and antiepileptic neuroprotective activity, but peripheral cardiovascular side effects impeded their development. SAR study of N(6)-cycloalkylmethyl 4'-truncated (N)-methanocarba-adenosines identified 10 (MRS5474, N(6)-dicyclopropylmethyl, K(i) = 47.9 nM) as a moderately A(1)AR-selective full agonist. Two stereochemically defined N(6)-methynyl group substituents displayed narrow SAR; groups larger than cyclobutyl greatly reduced AR affinity, and those larger or smaller than cyclopropyl reduced A(1)AR selectivity. Nucleoside docking to A(1)AR homology model characterized distinct hydrophobic cyclopropyl subpockets, the larger "A" forming contacts with Thr270 (7.35), Tyr271 (7.36), Ile274 (7.39), and carbon chains of glutamates (EL2) and the smaller subpocket "B" forming contacts between TM6 and TM7. 10 suppressed minimal clonic seizures (6 Hz mouse model) without typical rotarod impairment of A(1)AR agonists. Truncated nucleosides, an appealing preclinical approach, have more druglike physicochemical properties than other A(1)AR agonists. Thus, we identified highly restricted regions for substitution around N(6) suitable for an A(1)AR agonist with anticonvulsant activity.
Collapse
Affiliation(s)
- Dilip K. Tosh
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Silvia Paoletta
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Francesca Deflorian
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Khai Phan
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Steven M. Moss
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Zhan-Guo Gao
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Xiaohui Jiang
- Anticonvulsant Screening Program, Office of Translational Research, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Kenneth A. Jacobson
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
21
|
Müller CE, Jacobson KA. Recent developments in adenosine receptor ligands and their potential as novel drugs. BIOCHIMICA ET BIOPHYSICA ACTA 2011; 1808:1290-308. [PMID: 21185259 PMCID: PMC3437328 DOI: 10.1016/j.bbamem.2010.12.017] [Citation(s) in RCA: 334] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2010] [Revised: 12/14/2010] [Accepted: 12/15/2010] [Indexed: 01/16/2023]
Abstract
Medicinal chemical approaches have been applied to all four of the adenosine receptor (AR) subtypes (A(1), A(2A), A(2B), and A(3)) to create selective agonists and antagonists for each. The most recent class of selective AR ligands to be reported is the class of A(2B)AR agonists. The availability of these selective ligands has facilitated research on therapeutic applications of modulating the ARs and in some cases has provided clinical candidates. Prodrug approaches have been developed which improve the bioavailability of the drugs, reduce side-effects, and/or may lead to site-selective effects. The A(2A) agonist regadenoson (Lexiscan®), a diagnostic drug for myocardial perfusion imaging, is the first selective AR agonist to be approved. Other selective agonists and antagonists are or were undergoing clinical trials for a broad range of indications, including capadenoson and tecadenoson (A(1) agonists) for atrial fibrillation, or paroxysmal supraventricular tachycardia, respectively, apadenoson and binodenoson (A(2A) agonists) for myocardial perfusion imaging, preladenant (A(2A) antagonist) for the treatment of Parkinson's disease, and CF101 and CF102 (A(3) agonists) for inflammatory diseases and cancer, respectively.
Collapse
|
22
|
Guo HM, Zhang Y, Niu HY, Wang DC, Chu ZL, Qu GR. Microwave promoted C6-alkylation of purines through S(N)Ar-based reaction of 6-chloropurines with 3-alkyl-acetylacetone. Org Biomol Chem 2011; 9:2065-8. [PMID: 21340084 DOI: 10.1039/c0ob01213k] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
C6-Alkylated purine analogues were obtained in good to excellent isolated yields by S(N)Ar reaction of 6-chloropurine derivatives with 3-alkyl-acetylacetone. 3-Alkyl-acetylacetones were employed as alkylating agents and C6-alkylated purines were obtained highly selectively within short reaction time under microwave irradiation conditions. This work is complementary to the classical coupling reactions for the synthesis of C6-alkylated purine analogues.
Collapse
Affiliation(s)
- Hai-Ming Guo
- College of Chemistry and Environmental Science, Key Laboratory of Green Chemical Media and Reactions of Ministry of Education, Henan Normal University, Xinxiang 453007, Henan, China.
| | | | | | | | | | | |
Collapse
|
23
|
Swamy KCK, Kumar NNB, Balaraman E, Kumar KVPP. Mitsunobu and Related Reactions: Advances and Applications. Chem Rev 2009; 109:2551-651. [PMID: 19382806 DOI: 10.1021/cr800278z] [Citation(s) in RCA: 877] [Impact Index Per Article: 54.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- K. C. Kumara Swamy
- School of Chemistry, University of Hyderabad, Hyderabad − 500046, A. P., India
| | - N. N. Bhuvan Kumar
- School of Chemistry, University of Hyderabad, Hyderabad − 500046, A. P., India
| | - E. Balaraman
- School of Chemistry, University of Hyderabad, Hyderabad − 500046, A. P., India
| | | |
Collapse
|
24
|
Nell PG, Albrecht-Küpper B. The adenosine A1 receptor and its ligands. PROGRESS IN MEDICINAL CHEMISTRY 2009; 47:163-201. [PMID: 19328291 DOI: 10.1016/s0079-6468(08)00204-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Peter G Nell
- Global Drug Discovery - Operations, Bayer HealthCare AG, Bayer Schering Pharma, Müllerstrasse 178, 13353 Berlin, Germany
| | | |
Collapse
|
25
|
Abstract
The development of potent and selective agonists and antagonists of adenosine receptors (ARs) has been a target of medicinal chemistry research for several decades, and recently the US Food and Drug Administration has approved Lexiscan, an adenosine derivative substituted at the 2 position, for use as a pharmacologic stress agent in radionuclide myocardial perfusion imaging. Currently, some other adenosine A(2A) receptor (A(2A)AR) agonists and antagonists are undergoing preclinical testing and clinical trials. While agonists are potent antiinflammatory agents also showing hypotensive effects, antagonists are being developed for the treatment of Parkinson's disease.However, since there are still major problems in this field, including side effects, low brain penetration (for the targeting of CNS diseases), short half-life, or lack of in vivo effects, the design and development of new AR ligands is a hot research topic.This review presents an update on the medicinal chemistry of A(2A)AR agonists and antagonists, and stresses the strong need for more selective ligands at the human A(2A)AR subtype, in particular in the case of agonists.
Collapse
Affiliation(s)
- Gloria Cristalli
- Dipartimento di Scienze Chimiche, Università di Camerino, 62032 Camerino (MC), Italy.
| | | | | |
Collapse
|
26
|
Abstract
Adenosine acts as a cytoprotective modulator in response to stress to an organ or tissue. Although short-lived in the circulation, it can activate four subtypes of G protein-coupled adenosine receptors (ARs): A(1), A(2A), A(2B), and A(3). The alkylxanthines caffeine and theophylline are the prototypical antagonists of ARs, and their stimulant actions occur primarily through this mechanism. For each of the four AR subtypes, selective agonists and antagonists have been introduced and used to develop new therapeutic drug concepts. ARs are notable among the GPCR family in the number and variety of agonist therapeutic candidates that have been proposed. The selective and potent synthetic AR agonists, which are typically much longer lasting in the body than adenosine, have potential therapeutic applications based on their anti-inflammatory (A(2A) and A(3)), cardioprotective (preconditioning by A(1) and A(3) and postconditioning by A(2B)), cerebroprotective (A(1) and A(3)), and antinociceptive (A(1)) properties. Potent and selective AR antagonists display therapeutic potential as kidney protective (A(1)), antifibrotic (A(2A)), neuroprotective (A(2A)), and antiglaucoma (A(3)) agents. AR agonists for cardiac imaging and positron-emitting AR antagonists are in development for diagnostic applications. Allosteric modulators of A(1) and A(3) ARs have been described. In addition to the use of selective agonists/antagonists as pharmacological tools, mouse strains in which an AR has been genetically deleted have aided in developing novel drug concepts based on the modulation of ARs.
Collapse
Affiliation(s)
- Kenneth A Jacobson
- Molecular Recognition Section, Laboratory of Biooorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892-0810, USA.
| |
Collapse
|
27
|
Koszelewski D, Lavandera I, Clay D, Guebitz GM, Rozzell D, Kroutil W. Formal asymmetric biocatalytic reductive amination. Angew Chem Int Ed Engl 2008; 47:9337-40. [PMID: 18972473 DOI: 10.1002/anie.200803763] [Citation(s) in RCA: 177] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Dominik Koszelewski
- Research Centre Applied Biocatalysis, c/o Department of Chemistry, Organic and Bioorganic Chemistry, University of Graz, Heinrichstrasse 28, 8010 Graz, Austria
| | | | | | | | | | | |
Collapse
|
28
|
Koszelewski D, Lavandera I, Clay D, Guebitz G, Rozzell D, Kroutil W. Formal Asymmetric Biocatalytic Reductive Amination. Angew Chem Int Ed Engl 2008. [DOI: 10.1002/ange.200803763] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Dominik Koszelewski
- Research Centre Applied Biocatalysis, c/o Department of Chemistry, Organic and Bioorganic Chemistry, University of Graz, Heinrichstrasse 28, 8010 Graz (Austria), Fax: (+43) 316‐380‐9840
| | - Iván Lavandera
- Research Centre Applied Biocatalysis, c/o Department of Chemistry, Organic and Bioorganic Chemistry, University of Graz, Heinrichstrasse 28, 8010 Graz (Austria), Fax: (+43) 316‐380‐9840
| | - Dorina Clay
- Research Centre Applied Biocatalysis, c/o Department of Chemistry, Organic and Bioorganic Chemistry, University of Graz, Heinrichstrasse 28, 8010 Graz (Austria), Fax: (+43) 316‐380‐9840
| | - Georg M. Guebitz
- Institute of Environmental Biotechnology, Research Centre Applied Biocatalysis, University of Technology, Petersgasse 12, 8010 Graz (Austria)
| | | | - Wolfgang Kroutil
- Research Centre Applied Biocatalysis, c/o Department of Chemistry, Organic and Bioorganic Chemistry, University of Graz, Heinrichstrasse 28, 8010 Graz (Austria), Fax: (+43) 316‐380‐9840
| |
Collapse
|
29
|
Giorgi I, Nieri P. Therapeutic potential of A1adenosine receptor ligands: a survey of recent patent literature. Expert Opin Ther Pat 2008. [DOI: 10.1517/13543776.18.7.677] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
30
|
Bromberg KD, Iyengar R, He JC. Regulation of neurite outgrowth by G(i/o) signaling pathways. FRONT BIOSCI-LANDMRK 2008; 13:4544-57. [PMID: 18508528 DOI: 10.2741/3022] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Neurogenesis is a long and winding journey. A neural progenitor cell migrates long distances, differentiates by forming a single axon and multiple dendrites, undergoes maturation, and ultimately survives. The initial formation of neurites during neuronal differentiation, commonly referred to as "neurite outgrowth," can be induced by a large repertoire of signals that stimulate an array of receptors and downstream signaling pathways. The G(i/o) family of heterotrimeric G-proteins are abundantly expressed in the brain and enriched at neuronal growth cones. Recent evidence has uncovered several G(i/o)-coupled receptors that induce neurite outgrowth and has begun to elucidate the underlying molecular mechanisms. Emerging data suggests that signals from several G(i/o)-coupled receptors converge at the transcription factor STAT3 to regulate neurite outgrowth and at Rac1 and Cdc42 to regulate cytoskeletal reorganization. Physiologically, signaling through G(i/o)-coupled cannabinoid receptors is critical for pro percentral nervous system development. As the mechanisms by which G(i/o)-coupled receptors regulate neurite outgrowth are clarified, it is becoming evident that modulating signals from G(i/o) and their receptors has great potential for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Kenneth D Bromberg
- Department of Pharmacology and Systems Therapeutics, Mount Sinai School of Medicine, New York, NY 10029, USA.
| | | | | |
Collapse
|
31
|
Synthesis of (R)-, (S)-, and (RS)-hydroxymethylmexiletine, one of the major metabolites of mexiletine. ACTA ACUST UNITED AC 2007. [DOI: 10.1016/j.tetasy.2007.10.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
32
|
Ivanov AA, Palyulin VA, Zefirov NS. Computer aided comparative analysis of the binding modes of the adenosine receptor agonists for all known subtypes of adenosine receptors. J Mol Graph Model 2007; 25:740-54. [PMID: 17095272 DOI: 10.1016/j.jmgm.2006.06.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2006] [Revised: 06/20/2006] [Accepted: 06/21/2006] [Indexed: 12/01/2022]
Abstract
Molecular models of all known subtypes (A1, A2A, A2B, and A3) of the human adenosine receptors were built in homology with bovine rhodopsin. These models include the transmembrane domain as well as all extracellular and intracellular hydrophilic loops and terminal domains. The molecular docking of adenosine and 46 selected derivatives was performed for each receptor subtype. A binding mode common for all studied agonists was proposed, and possible explanations for differences in the ligand activities were suggested.
Collapse
Affiliation(s)
- Andrei A Ivanov
- Department of Chemistry, M.V. Lomonosov Moscow State University, 119992 Moscow, Russian Federation
| | | | | |
Collapse
|
33
|
Duong HT, Gao ZG, Jacobson KA. Nucleoside modification and concerted mutagenesis of the human A3 adenosine receptor to probe interactions between the 2-position of adenosine analogs and Gln167 in the second extracellular loop. NUCLEOSIDES NUCLEOTIDES & NUCLEIC ACIDS 2006; 24:1507-17. [PMID: 16438031 PMCID: PMC6954878 DOI: 10.1080/15257770500265778] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Residues of the second extracellular loop are believed to be important for ligand recognition in adenosine receptors. Molecular modeling studies have suggested that one such residue, Gln167 of the human A3 receptor, is in proximity to the C2 moiety of some adenosine analogs when bound. Here this putative interaction was systematically explored using a neoceptor strategy, i.e., by site-directed mutagenesis and examination of the affinities of nucleosides modified to have complementary functionality. Gln167 was mutated to Ala, Glu, and Arg, while the 2-position of several adenosine analogs was substituted with amine or carboxylic acid groups. All compounds tested lost affinity to the mutant receptors in comparison to the wild type. However, comparing affinities among the mutant receptors, several compounds bearing charge at the 2-position demonstrated preferential affinity for the mutant receptor bearing a residue of complementary charge. 13, with a positively-charged C2 moiety, displayed an 8.5-fold increase in affinity at the Q167E mutant receptor versus the Q167R mutant receptor Preferential affinity for specific mutant receptors was also observed for 8 and 12. The data suggests that a direct contact is made between the C2 substituent of some charged ligands and the mutant receptor bearing the opposite charge at position 167.
Collapse
Affiliation(s)
- Heng T Duong
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892-0810, USA
| | | | | |
Collapse
|
34
|
Abstract
Adenosine receptors are major targets of caffeine, the most commonly consumed drug in the world. There is growing evidence that they could also be promising therapeutic targets in a wide range of conditions, including cerebral and cardiac ischaemic diseases, sleep disorders, immune and inflammatory disorders and cancer. After more than three decades of medicinal chemistry research, a considerable number of selective agonists and antagonists of adenosine receptors have been discovered, and some have been clinically evaluated, although none has yet received regulatory approval. However, recent advances in the understanding of the roles of the various adenosine receptor subtypes, and in the development of selective and potent ligands, as discussed in this review, have brought the goal of therapeutic application of adenosine receptor modulators considerably closer.
Collapse
Affiliation(s)
- Kenneth A Jacobson
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892-0810, USA.
| | | |
Collapse
|
35
|
Fossa P, Mosti L, Bondavalli F, Schenone S, Ranise A, Casolino C, Forina M. Affinity prediction on A1 adenosine receptor agonists: the chemometric approach. Bioorg Med Chem 2005; 14:1348-63. [PMID: 16263293 DOI: 10.1016/j.bmc.2005.09.058] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2005] [Revised: 09/08/2005] [Accepted: 09/27/2005] [Indexed: 10/25/2022]
Abstract
In this paper, we are presenting a quantitative-structure-activity relationship (QSAR) study performed on 21 selective A(1) adenosine receptor agonists plus the endogenous substrate, adenosine, so as to identify those predictors which play a key role in describing the binding of the ligand with the A(1) receptor. A large number of molecular descriptors plus a calculated receptor-agonist binding energy and atomic charges were taken into account to derive different QSAR models, using different regression techniques. The results obtained both with linear and nonlinear approaches converge to the selection of the same informative parameters, highlighting the correlation of these descriptors with the biological Response. The evaluation 'a priori' of these predictors could therefore represent a useful tool in the screening of large libraries of compounds and in the rational design of new selective agonists.
Collapse
Affiliation(s)
- Paola Fossa
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Genova, Genoa, Italy.
| | | | | | | | | | | | | |
Collapse
|
36
|
Yan L, Burbiel JC, Maass A, Müller CE. Adenosine receptor agonists: from basic medicinal chemistry to clinical development. Expert Opin Emerg Drugs 2005; 8:537-76. [PMID: 14662005 DOI: 10.1517/14728214.8.2.537] [Citation(s) in RCA: 104] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Adenosine is a physiological nucleoside which acts as an autocoid and activates G protein-coupled membrane receptors, designated A(1), A(2A), A(2B) and A(3). Adenosine plays an important role in many (patho)physiological conditions in the CNS as well as in peripheral organs and tissues. Adenosine receptors are present on virtually every cell. However, receptor subtype distribution and densities vary greatly. Adenosine itself is used as a therapeutic agent for the treatment of supraventricular paroxysmal tachycardia and arrhythmias and as a vasodilatatory agent in cardiac imaging. During the past 20 years, a number of selective agonists for A(1), A(2A) and A(3) adenosine receptors have been developed, all of them structurally derived from adenosine. Several such compounds are currently undergoing clinical trials for the treatment of cardiovascular diseases (A(1)and A(2A)), pain (A(1)), wound healing (A(2A)), diabetic foot ulcers (A(2A)), colorectal cancer (A(3)) and rheumatoid arthritis (A(3)). Clinical evaluation of some A(1) and A(2A) adenosine receptor agonists has been discontinued. Major problems include side effects due to the wide distribution of adenosine receptors; low brain penetration, which is important for the targeting of CNS diseases; short half-lifes of compounds; or a lack of effects, in some cases perhaps due to receptor desensitisation or to low receptor density in the targeted tissue. Partial agonists, inhibitors of adenosine metabolism (adenosine kinase and deaminase inhibitors) or allosteric activators of adenosine receptors may be advantageous for certain indications, as they may exhibit fewer side effects.
Collapse
Affiliation(s)
- Luo Yan
- University of Bonn, Pharmaceutical Institute Poppelsdorf, Kreuzbergweg 26, D-53115 Bonn, Germany
| | | | | | | |
Collapse
|
37
|
Jacobson KA, Ohno M, Duong HT, Kim SK, Tchilibon S, Cesnek M, Holý A, Gao ZG. A neoceptor approach to unraveling microscopic interactions between the human A2A adenosine receptor and its agonists. CHEMISTRY & BIOLOGY 2005; 12:237-47. [PMID: 15734651 PMCID: PMC3122079 DOI: 10.1016/j.chembiol.2004.12.010] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2004] [Revised: 11/10/2004] [Accepted: 12/14/2004] [Indexed: 12/01/2022]
Abstract
Strategically mutated neoceptors, e.g., with anionic residues in TMs 3 and 7 intended for pairing with positively charged amine-modified nucleosides, were derived from the antiinflammatory A(2A) adenosine receptor (AR). Adenosine derivatives functionalized at the 5', 2, and N(6) positions were synthesized. The T88D mutation selectively enhanced the binding of the chain-length-optimized 5'-(2-aminoethyl)uronamide but not 5'-(2-hydroxyethyl)uronamide, suggesting a critical role of the positively charged amine. Combination of this modification with the N(6)-(2-methylbenzyl) group enhanced affinity at the Q89D- and N181D- but not the T88D-A(2A)AR. Amino groups placed near the 2- or N(6)-position only slightly affected the binding to mutant receptors. The 5'-hydrazide MRS3412 was 670- and 161-fold enhanced, in binding and functionally, respectively, at the Q89D-A(2A)AR compared to the wild-type. Thus, we identified and modeled pairs of A(2A)AR-derived neoceptor-neoligand, which are pharmacologically orthogonal with respect to the native species.
Collapse
Affiliation(s)
- Kenneth A Jacobson
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
An integrated approach to the study of drug-receptor interactions has been applied to adenosine receptors (ARs) and P2Y nucleotide receptors. This approach includes probing the receptor structure through site-directed mutagenesis and molecular modeling, in concert with altering the structure of the agonist ligands. Goals of this structural approach are to generate a testable hypothesis for location of the binding site and subsequently to enable the rational design of new agonists and antagonists. In this manner, receptor subtype selectivity has been increased, and agonists have been converted into partial agonists and antagonists. An approach to receptor engineering (neoceptors) has been explored, in which synthetic small molecule agonists (neoligands) are specifically tailored to activate only receptors in which the putative binding sites have been modified. This orthogonal approach to receptor activation, intended for eventual gene therapy, has been demonstrated for A3 and A2A ARs.
Collapse
Affiliation(s)
- Kenneth A Jacobson
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | |
Collapse
|
39
|
de Ligt RAF, van der Klein PAM, von Frijtag Drabbe Künzel JK, Lorenzen A, Ait El Maate F, Fujikawa S, van Westhoven R, van den Hoven T, Brussee J, IJzerman AP. Synthesis and biological evaluation of disubstituted N6-cyclopentyladenine analogues: the search for a neutral antagonist with high affinity for the adenosine A1 receptor. Bioorg Med Chem 2004; 12:139-49. [PMID: 14697779 DOI: 10.1016/j.bmc.2003.10.023] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Novel 3,8- and 8,9-disubstituted N(6)-cyclopentyladenine derivatives were synthesised in moderate overall yield from 6-chloropurine. The derivatives were made in an attempt to find a new neutral antagonist with high affinity for adenosine A(1) receptors. N(6)-Cyclopentyl-9-methyladenine (N-0840) was used as a lead compound. Binding affinities of the new analogues were determined for human adenosine A(1) and A(3) receptors. Their intrinsic activity was assessed in [35S]GTPgammaS binding experiments. Elongation of the 9-methyl of N-0840 to a 9-propyl substituent was very well tolerated. A 9-benzyl group, on the other hand, caused a decrease in adenosine A(1) receptor affinity. Next, the 8-position was examined in detail, and affinity was increased with appropriate substitution. Most derivatives were A(1)-selective and 20 of the new compounds (6-9, 15-21, 23-26, 28, 31, 33, 35, and 36) had higher adenosine A(1) receptor affinity than the reference substance, N-0840. Compound 31 (N(6)-cyclopentyl-8-(N-methylisopropylamino)-9-methyladenine, LUF 5608) had the highest adenosine A(1) receptor affinity, 7.7 nM. In the [35S]GTPgammaS binding experiments, derivatives 5, 14, 22, 23, 25, 26, 33 and 34 did not significantly change basal [35S]GTPgammaS binding, thus behaving as neutral antagonists. Moreover, four of these compounds (23, 25, 26, and 33) displayed a 4- to 10-fold increased adenosine A(1) receptor affinity (75-206 nM) compared to N-0840 (852 nM). In summary, we synthesised a range of N-0840 analogues with higher affinity for adenosine A(1) receptors. In addition, four new derivatives, LUF 5666 (23), LUF 5668 (25), LUF 5669 (26) and LUF 5674 (33), behaved as neutral antagonists when tested in [35S]GTPgammaS binding studies. Thus, these compounds have improved characteristics as neutral adenosine A(1) receptor antagonists.
Collapse
Affiliation(s)
- Rianne A F de Ligt
- Division of Medicinal Chemistry, Leiden/Amsterdam Center for Drug Research, Leiden University, PO Box 9502, 2300 RA, Leiden, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Etherington LAV, Frenguelli BG. Endogenous adenosine modulates epileptiform activity in rat hippocampus in a receptor subtype-dependent manner. Eur J Neurosci 2004; 19:2539-50. [PMID: 15128407 DOI: 10.1111/j.0953-816x.2004.03355.x] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The purine nucleoside adenosine is released during seizure activity and exerts an anticonvulsant influence through inhibition of glutamate release and hyperpolarization of neurons via adenosine A(1) receptors. However, activation of adenosine A(2A) and A(3) receptors may counteract the inhibitory effects of A(1) receptors. We have therefore examined the extent to which endogenous adenosine released during seizure activity activates the different adenosine receptor subtypes and the implications for seizure activity in the rat hippocampus in vitro. Brief trains of high-frequency stimulation in nominally Mg(2+)-free artificial cerebrospinal fluid evoked epileptiform activity and resulted in a transient depression of the simultaneously recorded CA1 field excitatory postsynaptic potential. In the presence of 8-cyclopentyl-1,3-dimethylxanthine (CPT), an adenosine A(1) receptor antagonist, the occurrence of spontaneous seizure activity was greatly increased as was the duration and intensity of evoked seizures, whilst the postictal depression of basal synaptic transmission was greatly attenuated. Application of ZM 241385, an adenosine A(2A) receptor antagonist, shortened the duration of epileptiform activity, whereas administration of MRS 1191, an adenosine A(3) receptor antagonist, both decreased the duration and intensity of seizures. Combined application of the A(2A) and A(3) receptor antagonists also resulted in a reduction in seizure duration and intensity. However, no evidence was found for a role for protein kinase C in the regulation of seizure activity by endogenous adenosine. Our data confirm the dominant anticonvulsant role that endogenous and tonic adenosine play via the A(1) receptor, and suggest that the additional adenosine receptor subtypes may compromise this anticonvulsant property through promotion of seizure activity.
Collapse
Affiliation(s)
- Lori-An V Etherington
- Neurosciences Institute, Division of Pathology and Neuroscience, University of Dundee, Ninewells Hospital, Dundee DD1 9SY, Scotland, UK
| | | |
Collapse
|
41
|
Tchilibon S, Kim SK, Gao ZG, Harris BA, Blaustein JB, Gross AS, Duong HT, Melman N, Jacobson KA. Exploring distal regions of the A3 adenosine receptor binding site: sterically constrained N6-(2-phenylethyl)adenosine derivatives as potent ligands. Bioorg Med Chem 2004; 12:2021-34. [PMID: 15080906 PMCID: PMC4667970 DOI: 10.1016/j.bmc.2004.02.037] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2003] [Revised: 02/11/2004] [Accepted: 02/28/2004] [Indexed: 10/26/2022]
Abstract
We synthesized phenyl ring-substituted analogues of N(6)-(1S,2R)-(2-phenyl-1-cyclopropyl)adenosine, which is highly potent in binding to the human A(3)AR with a Ki value of 0.63 nM. The effects of these structural changes on affinity at human and rat adenosine receptors and on intrinsic efficacy at the hA(3)AR were measured. A 3-nitrophenyl analogue was resolved chromatographically into pure diastereomers, which displayed 10-fold stereoselectivity in A(3)AR binding in favor of the 1S,2R isomer. A molecular model defined a hydrophobic region (Phe168) in the putative A(3)AR binding site around the phenyl moiety. A heteroaromatic group (3-thienyl) could substitute for the phenyl moiety with retention of high affinity of A(3)AR binding. Other related N(6)-substituted adenosine derivatives were included for comparison. Although the N(6)-(2-phenyl-1-cyclopropyl) derivatives were full A(3)AR agonists, several other derivatives had greatly reduced efficacy. N(6)-Cyclopropyladenosine was an A(3)AR antagonist, and adding either one or two phenyl rings at the 2-position of the cyclopropyl moiety restored efficacy. N(6)-(2,2-Diphenylethyl)adenosine was an A(3)AR antagonist, and either adding a bond between the two phenyl rings (N(6)-9-fluorenylmethyl) or shortening the ethyl moiety (N(6)-diphenylmethyl) restored efficacy. A QSAR study of the N(6) region provided a model that was complementary to the putative A(3)AR binding site in a rhodopsin-based homology model. Thus, a new series of high-affinity A(3)AR agonists and related nucleoside antagonists was explored through both empirical and theoretical approaches.
Collapse
Affiliation(s)
- Susanna Tchilibon
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health (NIH), DHHS, Bethesda, MD 20892-0810, USA
| | - Soo-Kyung Kim
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health (NIH), DHHS, Bethesda, MD 20892-0810, USA
| | - Zhan-Guo Gao
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health (NIH), DHHS, Bethesda, MD 20892-0810, USA
| | - Brian A. Harris
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health (NIH), DHHS, Bethesda, MD 20892-0810, USA
| | - Joshua B. Blaustein
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health (NIH), DHHS, Bethesda, MD 20892-0810, USA
| | - Ariel S. Gross
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health (NIH), DHHS, Bethesda, MD 20892-0810, USA
| | - Heng T. Duong
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health (NIH), DHHS, Bethesda, MD 20892-0810, USA
| | - Neli Melman
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health (NIH), DHHS, Bethesda, MD 20892-0810, USA
| | - Kenneth A. Jacobson
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health (NIH), DHHS, Bethesda, MD 20892-0810, USA
| |
Collapse
|
42
|
Jagtap PG, Chen Z, Szabó C, Klotz KN. 2-(N-Acyl) and 2-N-acyl-N6-substituted analogues of adenosine and their affinity at the human adenosine receptors. Bioorg Med Chem Lett 2004; 14:1495-8. [PMID: 15006389 DOI: 10.1016/j.bmcl.2004.01.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2003] [Revised: 12/18/2003] [Accepted: 01/09/2004] [Indexed: 11/17/2022]
Abstract
A series of 2-(N-acyl) and 2-(N-acyl)-N(6)-alkyladenosine analogues have been synthesized from the intermediate 2-amino-6-chloroadenosine derivatives (2b and 7) and evaluated for their affinity at the human A(1), A(2A), and A(3) receptors. We found that 2-(N-acyl) derivatives of adenosine showed relatively low affinity at A(2A) and A(3) receptors, while the N(6)-cyclopentyl substituent in 4h and 4i induced high potency [A(1) (K(i))=20.7 and 31.8 nM respectively] at the A(1) receptor and resulted therefore in increased selectivity for this subtype. The general synthetic methods and their binding studies are presented herein.
Collapse
Affiliation(s)
- Prakash G Jagtap
- Inotek Pharmaceuticals Corporation, 100 Cummings Center, Suite 419E, Beverly, MA 01915, USA.
| | | | | | | |
Collapse
|
43
|
Palle VP, Varkhedkar V, Ibrahim P, Ahmed H, Li Z, Gao Z, Ozeck M, Wu Y, Zeng D, Wu L, Leung K, Chu N, Zablocki JA. Affinity and intrinsic efficacy (IE) of 5′-carbamoyl adenosine analogues for the A1 adenosine receptor—efforts towards the discovery of a chronic ventricular rate control agent for the treatment of atrial fibrillation (AF). Bioorg Med Chem Lett 2004; 14:535-9. [PMID: 14698198 DOI: 10.1016/j.bmcl.2003.09.094] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The SAR for the affinity to the A(1) adenosine receptor and relative intrinsic efficacy (IE, [(35)S]-GTPgammaS binding) of a series of 5'-carbamate and 5'-thionocarbamate derivatives of tecadenoson is described. Based on this SAR, selected compounds were evaluated in guinea pig isolated hearts to determine whether they were partial or full agonists with respect to their negative dromotropism, an A(1) AdoR mediated effect. Progress towards obtaining a partial A(1) AdoR agonist to potentially control ventricular rate during atrial fibrillation has been made with the discovery of several potent partial A(1) AdoR agonists (compounds 13, 14, and 17).
Collapse
Affiliation(s)
- Venkata P Palle
- Department of Bioorganic Chemistry, CV Therapeutics, 3172 Porter Drive, Palo Alto, CA 94304, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
The activation of adenosine A1, A2 andA3 receptors can protect neurones against damage generated by mechanical or hypoxic/ischaemic insults as well as excitotoxins. A1 receptors are probably effective by suppressing transmitter release and producing neuronal hyperpolarisation. They are less likely to be of therapeutic importance due to the plethora of side effects resulting from A1 agonism, although the existence of receptor subtypes and recent synthetic chemistry efforts to increase ligand selectivity, may yet yield clinically viable compounds. Activation of A2A receptors can protect neurons, although there is much uncertainty as to whether agonists are acting centrally or via a peripheral mechanism such as altering blood flow or immune cell function. Selective antagonists at the A2A receptor, such as 4-(2-[7-amino-2-(2-furyl)(1,2,4)triazolo(2,3-a)(1,3,5)triazin-5-yl-amino]ethyl)phenol (ZM 241385) and 7-(2-phenylethyl)-5-amino-2-(2-furyl)-pyrazolo-[4,3e]-1,2,4-triazolo[1,5-c]pyrimidine (SCH 58261), can also protect against neuronal death produced by ischaemia or excitotoxicity. In addition, A2A receptor antagonists can reduce damage produced by combinations of subthreshold doses of the endogenous excitotoxin quinolinic acid and free radicals. Since the A2A receptors do not seem to be activated by normal endogenous levels of adenosine, their blockade should not generate significant side effects, so that A2A receptor antagonists appear to be promising candidates as new drugs for the prevention of neuronal damage. Adenosine A3 receptors have received less attention to date, but agonists are clearly able to afford protection against damage when administered chronically. Given the disappointing lack of success of NMDA receptor antagonists in human stroke patients, despite their early promise in animal models, it is possible that A2A receptor antagonists could have a far greater clinical utility.
Collapse
Affiliation(s)
- Trevor W Stone
- Division of Neuroscienec and Biomedical Systems, West Medical Bldg, University of Glasgow, Glasgow G12 8QQ, UK
| |
Collapse
|
45
|
Harrison PK, Bueters TJH, Ijzerman AP, van Helden HPM, Tattersall JEH. Partial adenosine A(1) receptor agonists inhibit sarin-induced epileptiform activity in the hippocampal slice. Eur J Pharmacol 2003; 471:97-104. [PMID: 12818696 DOI: 10.1016/s0014-2999(03)01783-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Organophosphate poisoning can result in seizures and subsequent neuropathology. One possible therapeutic approach would be to employ adenosine A(1) receptor agonists, which have already been shown to have protective effects against organophosphate poisoning. Using an in vitro model of organophosphate-induced seizures, we have investigated the ability of several adenosine A(1) receptor agonists to inhibit epileptiform activity induced by the organophosphate sarin, in the CA1 stratum pyramidale of the guinea pig hippocampal slice. Application of the adenosine A(1) receptor agonist N(6)-cyclopentyladenosine (CPA) or the partial adenosine A(1) receptor agonists 2-deoxy-N(6)-cyclopentyladenosine (2-deoxy-CPA) and 8-butylamino-N(6)-cyclopentyladenosine (8-butylamino-CPA) abolished epileptiform activity in a concentration-related manner. The rank order of potency was CPA (IC(50) 4-5 nM) >2-deoxy-CPA (IC(50) 113-119 nM)=8-butylamino-CPA (IC(50) 90-115 nM). These data suggest that partial adenosine A(1) receptor agonists, which have fewer cardiovascular effects, should be further evaluated in vivo as potential treatments for organophosphate poisoning.
Collapse
Affiliation(s)
- Patrick K Harrison
- Department of Biomedical Sciences, Dstl Chemical and Biological Sciences, Wiltshire, Salisbury SP4 0JQ, UK.
| | | | | | | | | |
Collapse
|
46
|
Blum D, Hourez R, Galas MC, Popoli P, Schiffmann SN. Adenosine receptors and Huntington's disease: implications for pathogenesis and therapeutics. Lancet Neurol 2003; 2:366-74. [PMID: 12849153 DOI: 10.1016/s1474-4422(03)00411-3] [Citation(s) in RCA: 104] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Huntington's disease (HD) is a devastating hereditary neurodegenerative disorder, the progression of which cannot be prevented by any neuroprotective approach, despite major advances in the understanding of its pathogenesis. The study of several animal models of the disease has led to the discovery of both loss-of-normal and gain-of-toxic functions of the mutated huntingtin protein and the elucidation of the mechanisms that underlie the formation of huntingtin aggregates and nuclear inclusions. Moreover, these models also provide good evidence of a role for excitotoxicity and mitochondrial metabolic impairments in striatal neuronal death. Adenosine has neuroprotective potential in both acute and chronic neurological disorders such as stroke or Parkinson's disease. Here we review experimental data on the role of A1 and A2A adenosine receptors in HD that warrant further investigation of the beneficial effects of A1 agonists and A2A antagonists in animal models of HD. Future pharmacological analysis of adenosine receptors could justify the use of A1 agonists and A2A antagonists for the treatment of HDin clinical trials.
Collapse
Affiliation(s)
- David Blum
- Laboratory of Neurophysiology, Université Libre de Bruxelles, Belgium.
| | | | | | | | | |
Collapse
|
47
|
Weeks SG, Silva C, Auer RN, Doig CJ, Gill MJ, Power C. Encephalopathy with staphylococcal endocarditis: multiple neuropathological findings. Can J Neurol Sci 2001; 28:260-4. [PMID: 11513347 DOI: 10.1017/s0317167100001438] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND Infective endocarditis is associated with serious neurological sequelae. OBJECTIVE Here, we report a patient with Staphylococcus aureus endocarditis, secondary to congenital heart disease, with subacute onset of multiple neurological complications. RESULTS Despite prompt antibiotic treatment with rapid sterilization of blood cultures, the patient died with brain herniation within 96 hours of admission. Neuropathological examination showed intraparenchymal hemorrhages, mycotic aneurysms, micro-abscesses and septic arteritis with accompanying infarction. Immunocytochemical studies revealed enhanced CD45 and GFAP immunoreactivity, together with adenosine A1 receptor detection on macrophages and microglia. CONCLUSIONS Infective endocarditis is associated with multiple neuropathological lesions, which may contribute to its poor clinical outcome and activation of cells of monocyte-microglial lineage throughout the brain.
Collapse
Affiliation(s)
- S G Weeks
- Department of Medicine, University of Calgary, AB, Canada
| | | | | | | | | | | |
Collapse
|
48
|
Chao HM, Osborne NN. Topically applied clonidine protects the rat retina from ischaemia/reperfusion by stimulating alpha(2)-adrenoceptors and not by an action on imidazoline receptors. Brain Res 2001; 904:126-36. [PMID: 11516418 DOI: 10.1016/s0006-8993(01)02499-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Ischaemia was induced to the rat retina by raising the intraocular pressure above the systolic blood pressure for 45 min. After a reperfusion period of 5 days, alterations in the localisation of choline acetyltransferase (ChAT) and calretinin immunoreactivities, a reduction in the thickness of the inner retinal layers and a decline in the b-wave amplitude of the electroretinogram were recorded. These changes were blunted when clonidine was injected intraperitoneally before or after ischaemia or when applied topically by a specific regime. Other alpha(2)-adrenoceptor agonists, brimonidine and apraclonidine, acted in a similar way to clonidine when applied topically but because of the number of experiments carried out a comparison between the effectiveness of the different alpha(2)-adrenoceptor agonists was not possible. The protective effect of clonidine was attenuated when the alpha(2)-adrenoceptor antagonists yohimbine or rauwolscine were co-administered, suggesting that the mechanism of action of the drug is to stimulate alpha(2)-adrenoceptors. In addition, the imidazoline receptor ligands, BU-226 and AGN-192403 did not blunt the effect of ischaemia/reperfusion, supporting the notion that the protective action of the alpha(2)-adrenoceptor agonists does not involve imidazoline sites but rather the activation of alpha(2)-adrenoceptors. The protective effect of 0.5% clonidine appeared to be greater when topically applied to the eye that received ischaemia than when applied by the same regime to the contralateral eye. These studies suggest that while most of topically applied clonidine reaches the retina by a systemic route one cannot rule out additional pathways.
Collapse
Affiliation(s)
- H M Chao
- Nuffield Laboratory of Ophthalmology, University of Oxford, Walton Street, OX2 6AW, Oxford, UK
| | | |
Collapse
|
49
|
Carruthers AM, Sellers LA, Jenkins DW, Jarvie EM, Feniuk W, Humphrey PP. Adenosine A(1) receptor-mediated inhibition of protein kinase A-induced calcitonin gene-related peptide release from rat trigeminal neurons. Mol Pharmacol 2001; 59:1533-41. [PMID: 11353815 DOI: 10.1124/mol.59.6.1533] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Calcitonin gene-related peptide (CGRP), a potent vasodilator, has been implicated in the pathogenesis of migraine. Its release from adult rat trigeminal neurons in culture was shown to be markedly increased by the activation of adenylate cyclase with forskolin. Modulation of this secretion was investigated by a number of agents with known inhibitory effects on cAMP generation mediated via receptor coupling to G(i/o) proteins. Significantly, forskolin-stimulated CGRP release could be closely correlated with the phosphorylation of the protein kinase A (PKA) substrate cyclic AMP response element-binding protein (CREB). Forskolin-stimulated CGRP release could be potently and effectively inhibited by the adenosine A(1) receptor-selective agonist GR79236X (pIC(50) = 7.7 +/- 0.1, maximal inhibition 65 +/- 2.5% at 300 nM), whereas the A(2A) (CGS21680) and the A(3) (2-chloro-N(6)-(3-iodobenzyl)-adenosine-5'-N-methyluronamide) receptor-selective agonists were without effect. GR79236X-mediated inhibition was abolished by the A(1) receptor antagonist 8-cyclopentyl-1,3-dipropylxanthine. Immunocytochemical studies and Western analysis revealed the presence of adenosine A(1) receptors on trigeminal neurons. However, despite the additional detection of 5-hydroxytryptamine (5-HT)(1B) receptors on these cells, the clinically effective antimigraine 5-HT(1B/1D) agonist sumatriptan did not inhibit forskolin-stimulated CGRP release nor did it show any effect on the concomitant CREB phosphorylation. In contrast, the mu-opioid agonist fentanyl elicited a 74 +/- 4% reduction in CGRP levels. Forskolin-stimulated CGRP release and CREB phosphorylation could be mimicked by incubation of the cells with chlorophenylthio-cAMP and blocked by pretreatment with the PKA inhibitor myrPKI(14-22). Taken together, the present data confirm the PKA-dependence of forskolin-stimulated CGRP release and suggest that A(1) adenosine agonists may warrant further investigation in models of migraine and neurogenic inflammation.
Collapse
Affiliation(s)
- A M Carruthers
- Glaxo Institute of Applied Pharmacology, Department of Pharmacology, University of Cambridge, Tennis Court Rd., Cambridge, CB2 1QJ, UK
| | | | | | | | | | | |
Collapse
|
50
|
Abstract
Agonist stimulation of adenosine A(1) receptors has been consistently shown to result in reduction of brain damage following experimentally induced global and focal brain ischaemia in animals. Unsurprisingly, the use of adenosine A(1) receptors as targets for the development of clinical therapeutics suitable for treatment of ischaemic brain disorders has been suggested by many authors. The latest studies of adenosine and its receptors indicate that adenosine-mediated actions might be far more complex than originally anticipated, casting some doubt about the rapid development of stroke treatment based on adenosine. This review discusses the possible role of adenosine receptor subtypes (A(1), A(2) and A(3)) in the context of their potential as therapeutics in stroke.
Collapse
Affiliation(s)
- D K von Lubitz
- Emergency Medicine Research Laboratories, Department of Emergency Medicine, University of Michigan Medical Center, TC/B1354/0303, 1500 E. Medical Center Drive, Ann Arbor, MI 48109-0303, USA.
| |
Collapse
|