1
|
Viswanath V, Mistry S, Cabrera-Ghayouri S, Leang R, Frail D, Donello J, Gil D. Sustained Alleviation of Autoimmunity by Activating α2B-adrenergic Receptors. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:435-441. [PMID: 38940628 DOI: 10.4049/jimmunol.2300893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 06/10/2024] [Indexed: 06/29/2024]
Abstract
Catecholamines binding to α- and β-adrenergic receptors on immune cells have recently been shown to play an important role in regulating immune responses. Although α2-adrenergic receptors are known to modulate the immune response in different ways, the therapeutic exploration of their utility has been limited by the lack of agonists selective for the three α2-adrenergic subtypes. We report in this study the identification of the agonist AGN-762, which activates α2B- and α2C-adrenergic subtypes, but not the α2A subtype. We show that AGN-762 reduced clinical disease in an experimental autoimmune encephalitis model of autoimmune disease via direct or indirect effects on T regulatory cells. The activity of AGN-762 was abrogated by depletion of T regulatory cells, which express the α2B-adrenergic receptor. Furthermore, a drug-induced shift to an anti-inflammatory phenotype was demonstrated in immune cells in the spleen of drug-treated experimental autoimmune encephalitis mice. AGN-762 does not display sedative and cardiovascular side effects associated with α2A subtype agonists. Immune modulation by selective α2-adrenergic agonists represents a novel, to our knowledge, approach for treating autoimmune disease.
Collapse
MESH Headings
- Animals
- Mice
- Receptors, Adrenergic, alpha-2/metabolism
- Receptors, Adrenergic, alpha-2/immunology
- Autoimmunity/immunology
- Adrenergic alpha-2 Receptor Agonists/pharmacology
- Adrenergic alpha-2 Receptor Agonists/therapeutic use
- Mice, Inbred C57BL
- T-Lymphocytes, Regulatory/immunology
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Humans
- Female
- Disease Models, Animal
Collapse
Affiliation(s)
| | - Shruti Mistry
- Ophthalmology Discovery Research, AbbVie Inc., Irvine, CA
| | | | - Ronika Leang
- Ophthalmology Discovery Research, AbbVie Inc., Irvine, CA
| | - Don Frail
- Alceptor Therapeutics, Newport Beach, CA
| | | | - Daniel Gil
- Alceptor Therapeutics, Newport Beach, CA
| |
Collapse
|
2
|
Lv X, Zhou P, Qiao X, Li Y, Yang X, Wang J, He X, Su R. Designing Chromane Derivatives as α 2A-Adrenoceptor Selective Agonists via Conformation Constraint. J Med Chem 2024; 67:11435-11449. [PMID: 38889119 DOI: 10.1021/acs.jmedchem.4c01239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Abstract
Enhancing the selectivity of alpha2-adrenoceptor (α2A-AR) agonists remains an unresolved issue. Herein, we reported the design of an α2A-AR agonist using the conformation constraint method, beginning with medetomidine. The structure-activity relationship indicated that the 8-substituent of chromane derivatives exerted the most pronounced effect on α2A-AR agonistic activity. Compounds A9 and B9 were identified as the most promising, exhibiting EC50 values of 0.78 and 0.23 nM, respectively. Their selectivity indexes surpassed dexmedetomidine (DMED) by 10-80 fold. In vivo studies demonstrated that both A9 and B9 dose-dependently increased the loss of righting reflex in mice, with ED50 values of 1.54 and 0.138 mg/kg, respectively. Binding mode calculations and mutation studies suggested the indispensability of the hydrogen bond between ASP1283.32 and α2A-AR agonist. In particular, A9 and B9 showed no dual reverse pharmacological effect, a characteristic exhibited by DMED in α2A-AR activation.
Collapse
Affiliation(s)
- Xucheng Lv
- Beijing Institute of Pharmacology and Toxicology, Haidian District, Beijing 100850, China
| | - Peilan Zhou
- Beijing Institute of Pharmacology and Toxicology, Haidian District, Beijing 100850, China
| | - Xuehong Qiao
- Beijing Institute of Pharmacology and Toxicology, Haidian District, Beijing 100850, China
- Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yulei Li
- Beijing Institute of Pharmacology and Toxicology, Haidian District, Beijing 100850, China
| | - Xingxing Yang
- Beijing Institute of Pharmacology and Toxicology, Haidian District, Beijing 100850, China
- Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jiaqi Wang
- Beijing Institute of Pharmacology and Toxicology, Haidian District, Beijing 100850, China
| | - Xinhua He
- Beijing Institute of Pharmacology and Toxicology, Haidian District, Beijing 100850, China
| | - Ruibin Su
- Beijing Institute of Pharmacology and Toxicology, Haidian District, Beijing 100850, China
| |
Collapse
|
3
|
Duan S, Zhou S. Dexmedetomidine and Perioperative Arrhythmias. J Cardiothorac Vasc Anesth 2024; 38:1221-1227. [PMID: 38443205 DOI: 10.1053/j.jvca.2024.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 12/13/2023] [Accepted: 01/08/2024] [Indexed: 03/07/2024]
Abstract
The highly selective α2-adrenoceptor agonist dexmedetomidine is a commonly used sedative drug for patients undergoing anesthesia and intensive care treatment. Several studies have indicated that dexmedetomidine may have a potential role in preventing and treating perioperative tachyarrhythmias. However, the specific effect and mechanism of action of dexmedetomidine in this context remain unclear. Dexmedetomidine is known to regulate the electrophysiologic function of the myocardium by inhibiting the function of the sinus node and atrioventricular node, as well as affecting myocardial repolarization. This paper aims to provide a theoretical basis for the prevention and treatment of perioperative arrhythmias by summarizing the effects of dexmedetomidine on myocardial electrophysiologic function and its impact on different types of arrhythmias.
Collapse
Affiliation(s)
- Shengji Duan
- Department of Anesthesiology, The Second People's Hospital of Yibin, Sichuan, China
| | - Shuzhi Zhou
- Department of Anesthesiology, Ya 'an People Hospital, Ya 'an, Sichuan, China.
| |
Collapse
|
4
|
Callaway CW, Flickinger KL, Weissman A, Guyette FX, DeMaio R, Jonsson A, Wu V, Monteleone JL, Prescott P, Birabaharan J, Buysse DJ, Empey PE, Nolin TD, West RE. Alpha-2-adrenergic agonists reduce resting energy expenditure in humans during external cooling. Temperature (Austin) 2024; 11:280-298. [PMID: 39193049 PMCID: PMC11346546 DOI: 10.1080/23328940.2024.2339781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 04/02/2024] [Accepted: 04/02/2024] [Indexed: 08/29/2024] Open
Abstract
Intravenous alpha-2-adrenergic receptor agonists reduce energy expenditure and lower the temperature when shivering begins in humans, allowing a decrease in core body temperature. Because there are few data about similar effects from oral drugs, we tested whether single oral doses of the sedative dexmedetomidine (1 µg/kg sublingual or 4 µg/kg swallowed) or the muscle relaxant tizanidine (8 mg or 16 mg), combined with surface cooling, reduce energy expenditure and core body temperature in humans. A total of 26 healthy participants completed 41 one-day laboratory studies measuring core body temperature using an ingested telemetry capsule and measuring energy expenditure using indirect calorimetry for up to 6 hours after drug ingestion. Dexmedetomidine induced a median 13% - 19% peak reduction and tizanidine induced a median 15% - 22% peak reduction in energy expenditure relative to baseline. Core body temperature decreased a median of 0.5°C - 0.6°C and 0.5°C - 0.7°C respectively. Decreases in temperature occurred after peak reductions in energy expenditure. Energy expenditure increased with a decrease in core temperature in control participants but did not occur after 4 µg/kg dexmedetomidine or 16 mg tizanidine. Plasma levels of dexmedetomidine but not tizanidine were related to mean temperature change. Decreases in heart rate, blood pressure, respiratory rate, cardiac stroke volume index, and cardiac index were associated with the change in metabolic rate after higher drug doses. We conclude that both oral dexmedetomidine and oral tizanidine reduce energy expenditure and allow decrease in core temperature in humans.
Collapse
Affiliation(s)
- Clifton W. Callaway
- Department of Emergency Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Katharyn L. Flickinger
- Department of Emergency Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Alexandra Weissman
- Department of Emergency Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Francis X. Guyette
- Department of Emergency Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Ryann DeMaio
- Department of Emergency Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Andrea Jonsson
- Department of Emergency Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Victor Wu
- Department of Emergency Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jenna L. Monteleone
- Department of Emergency Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Peter Prescott
- Department of Emergency Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jonathan Birabaharan
- Center for Clinical Pharmaceutical Sciences, Department of Pharmacy and Therapeutics, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, USA
| | - Daniel J. Buysse
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Philip E. Empey
- Center for Clinical Pharmaceutical Sciences, Department of Pharmacy and Therapeutics, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, USA
| | - Thomas D. Nolin
- Center for Clinical Pharmaceutical Sciences, Department of Pharmacy and Therapeutics, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, USA
| | - Raymond E. West
- Center for Clinical Pharmaceutical Sciences, Department of Pharmacy and Therapeutics, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, USA
| |
Collapse
|
5
|
Amna S, Øhlenschlaeger T, Saedder EA, Sigaard JV, Bergmann TK. Review of clinical pharmacokinetics and pharmacodynamics of clonidine as an adjunct to opioids in palliative care. Basic Clin Pharmacol Toxicol 2024; 134:485-497. [PMID: 38275186 DOI: 10.1111/bcpt.13979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 12/29/2023] [Accepted: 12/29/2023] [Indexed: 01/27/2024]
Abstract
Clonidine is an α-adrenoceptor agonist acting on receptors in the brain and peripheral tissues, leading to a reduction in sympathetic outflow and release of certain neurotransmitters. Clonidine has multiple uses across various medical conditions. One of its uses is as adjuvant to anaesthetic and analgesic agents specially opioids, mostly administered through intravenous and epidural routes. The opioids, effective in cancer pain management, are associated with various side effects such as sedation, pruritus, constipation, nausea, respiratory depression, tolerance and dependence. Combination of clonidine with opioids seems to help to achieve better pain management and less need of opioids. Use of clonidine in palliative care has been less common, but it is gradually gaining recognition for its potential benefits in managing symptoms like cancer pain and agitation. This combination approach has been explored in palliative care settings, including cancer pain and agitation, where patients experience complex and refractory symptoms. It seems to be well tolerated and gives better symptom relief. The available literature on clonidine's use in cancer pain and agitation management, especially in subcutaneous form, is limited and outdated. Therefore, the optimal dosing, safety profile and overall effectiveness of subcutaneous clonidine requires further exploration through prospective research studies.
Collapse
Affiliation(s)
- Sarwat Amna
- Department of Palliative Medicine, University Hospital of Southern Denmark, Esbjerg, Denmark
| | | | - Eva Aggerholm Saedder
- Department of Clinical Pharmacology, Aarhus University Hospital, Aarhus, Denmark
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Jarl Voss Sigaard
- Department of Palliative Medicine, University Hospital of Southern Denmark, Esbjerg, Denmark
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Troels Korshøj Bergmann
- Department of Clinical Pharmacology, Odense University Hospital, Odense, Denmark
- Department of Regional Health Research, University of Southern Denmark, Esbjerg, Denmark
| |
Collapse
|
6
|
Sun S, Li P, Wang J, Zhao D, Yang T, Zhou P, Su R, Zheng Z, Li S. Novel Scaffold Agonists of the α 2A Adrenergic Receptor Identified via Ensemble-Based Strategy. Molecules 2024; 29:1097. [PMID: 38474611 DOI: 10.3390/molecules29051097] [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: 02/07/2024] [Revised: 02/21/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
The α2A adrenergic receptor (α2A-AR) serves as a critical molecular target for sedatives and analgesics. However, α2A-AR ligands with an imidazole ring also interact with an imidazoline receptor as well as other proteins and lead to undesirable effects, motivating us to develop more novel scaffold α2A-AR ligands. For this purpose, we employed an ensemble-based ligand discovery strategy, integrating long-term molecular dynamics (MD) simulations and virtual screening, to identify new potential α2A-AR agonists with novel scaffold. Our results showed that compounds SY-15 and SY-17 exhibited significant biological effects in the preliminary evaluation of protein kinase A (PKA) redistribution assays. They also reduced levels of intracellular cyclic adenosine monophosphate (cAMP) in a dose-dependent manner. Upon treatment of the cells with 100 μM concentrations of SY-15 and SY-17, there was a respective decrease in the intracellular cAMP levels by 63.43% and 53.83%. Subsequent computational analysis was conducted to elucidate the binding interactions of SY-15 and SY-17 with the α2A-AR. The binding free energies of SY-15 and SY-17 calculated by MD simulations were -45.93 and -71.97 kcal/mol. MD simulations also revealed that both compounds act as bitopic agonists, occupying the orthosteric site and a novel exosite of the receptor simultaneously. Our findings of integrative computational and experimental approaches could offer the potential to enhance ligand affinity and selectivity through dual-site occupancy and provide a novel direction for the rational design of sedatives and analgesics.
Collapse
Affiliation(s)
- Shiyang Sun
- National Engineering Research Center for Strategic Drugs, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Pengyun Li
- National Engineering Research Center for Strategic Drugs, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Jiaqi Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Dongsheng Zhao
- Academy of Military Medical Sciences, Beijing 100850, China
| | - Tingting Yang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Peilan Zhou
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Ruibin Su
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Zhibing Zheng
- National Engineering Research Center for Strategic Drugs, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Song Li
- National Engineering Research Center for Strategic Drugs, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| |
Collapse
|
7
|
Wang X, Zhang R, Chen B, Zhang T, Jin X, Gao P. Preliminary evaluation of the efficacy and safety of brimonidine for deep sedation. Fundam Clin Pharmacol 2024; 38:139-151. [PMID: 37612481 DOI: 10.1111/fcp.12944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 05/16/2023] [Accepted: 07/20/2023] [Indexed: 08/25/2023]
Abstract
BACKGROUND Although brimonidine is currently used in the clinical treatment of glaucoma and rosacea, research of the deep sedative effect on animals after systemic administration is reported firstly and has shown promising results. METHODS The median effective dose (ED50 ), the median lethal dose (LD50 ), and the therapeutic index of brimonidine for deep sedation and formalin stimulation assay were determined by various animal experiments. The effect of synergistic anesthesia in rabbits with brimonidine and chloral hydrate was preliminarily evaluated. RESULTS The ED50 of brimonidine for highly effective sedation by intraperitoneal injection in rats was calculated to be 2.05 mg kg-1 with a 95% confidence interval (CI) of 1.87 to 2.25 mg kg-1 . The ED50 of brimonidine for deep sedation by intravenous and intrarectal injection in rabbits was calculated to be 0.087 mg kg-1 with a 95% CI of 0.084 to 0.091 mg kg-1 and 1.65 mg kg-1 with a 95% CI of 1.43 to 1.91 mg kg-1 , respectively. The LD50 of intraperitoneal brimonidine injection in rats was calculated to be 468 mg kg-1 with a 95% CI of 441 to 497 mg kg-1 and a therapeutic index of 228. Brimonidine has a certain analgesic and heart rate lowering effects. CONCLUSION The results confirmed that brimonidine has deep sedation and analgesic effects after systemic administration and has high safety. It can be used in combination with other types of sedative drugs to achieve better effects.
Collapse
Affiliation(s)
- Xiaohui Wang
- Laboratory of Traditional Chinese Medicine Preparations, Tianjin Institute of Medical & Pharmaceutical Sciences, Tianjin, China
| | - Rui Zhang
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Bin Chen
- Laboratory of Medical Biomaterials, Tianjin Institute of Medical & Pharmaceutical Sciences, Tianjin, China
| | - Ting Zhang
- Pharmacokinetics Laboratory, Tianjin Institute of Medical & Pharmaceutical Sciences, Tianjin, China
| | - Xinghua Jin
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Ping Gao
- Laboratory of Medical Biomaterials, Tianjin Institute of Medical & Pharmaceutical Sciences, Tianjin, China
| |
Collapse
|
8
|
Baker JG, Shaw DE. Asthma and COPD: A Focus on β-Agonists - Past, Present and Future. Handb Exp Pharmacol 2024; 285:369-451. [PMID: 37709918 DOI: 10.1007/164_2023_679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/16/2023]
Abstract
Asthma has been recognised as a respiratory disorder for millennia and the focus of targeted drug development for the last 120 years. Asthma is one of the most common chronic non-communicable diseases worldwide. Chronic obstructive pulmonary disease (COPD), a leading cause of morbidity and mortality worldwide, is caused by exposure to tobacco smoke and other noxious particles and exerts a substantial economic and social burden. This chapter reviews the development of the treatments of asthma and COPD particularly focussing on the β-agonists, from the isolation of adrenaline, through the development of generations of short- and long-acting β-agonists. It reviews asthma death epidemics, considers the intrinsic efficacy of clinical compounds, and charts the improvement in selectivity and duration of action that has led to our current medications. Important β2-agonist compounds no longer used are considered, including some with additional properties, and how the different pharmacological properties of current β2-agonists underpin their different places in treatment guidelines. Finally, it concludes with a look forward to future developments that could improve the β-agonists still further, including extending their availability to areas of the world with less readily accessible healthcare.
Collapse
Affiliation(s)
- Jillian G Baker
- Department of Respiratory Medicine, Queen's Medical Centre, Nottingham University Hospitals NHS Trust, Nottingham, UK.
- Cell Signalling, Medical School, Queen's Medical Centre, University of Nottingham, Nottingham, UK.
| | - Dominick E Shaw
- Nottingham NIHR Respiratory Biomedical Research Centre, University of Nottingham, Nottingham, UK
| |
Collapse
|
9
|
Mastos C, Xu X, Keen AC, Halls ML. Signalling of Adrenoceptors: Canonical Pathways and New Paradigms. Handb Exp Pharmacol 2024; 285:147-184. [PMID: 38227198 DOI: 10.1007/164_2023_704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2024]
Abstract
The concept of G protein-coupled receptors initially arose from studies of the β-adrenoceptor, adenylyl cyclase, and cAMP signalling pathway. Since then both canonical G protein-coupled receptor signalling pathways and emerging paradigms in receptor signalling have been defined by experiments focused on adrenoceptors. Here, we discuss the evidence for G protein coupling specificity of the nine adrenoceptor subtypes. We summarise the ability of each of the adrenoceptors to activate proximal signalling mediators including cAMP, calcium, mitogen-activated protein kinases, and protein kinase C pathways. Finally, we highlight the importance of precise spatial and temporal control of adrenoceptor signalling that is controlled by the localisation of receptors at intracellular membranes and in larger protein complexes.
Collapse
Affiliation(s)
- Chantel Mastos
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Xiaomeng Xu
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Alastair C Keen
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Michelle L Halls
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia.
| |
Collapse
|
10
|
Baker JG, Summers RJ. Adrenoceptors: Receptors, Ligands and Their Clinical Uses, Molecular Pharmacology and Assays. Handb Exp Pharmacol 2024; 285:55-145. [PMID: 38926158 DOI: 10.1007/164_2024_713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2024]
Abstract
The nine G protein-coupled adrenoceptor subtypes are where the endogenous catecholamines adrenaline and noradrenaline interact with cells. Since they are important therapeutic targets, over a century of effort has been put into developing drugs that modify their activity. This chapter provides an outline of how we have arrived at current knowledge of the receptors, their physiological roles and the methods used to develop ligands. Initial studies in vivo and in vitro with isolated organs and tissues progressed to cell-based techniques and the use of cloned adrenoceptor subtypes together with high-throughput assays that allow close examination of receptors and their signalling pathways. The crystal structures of many of the adrenoceptor subtypes have now been determined opening up new possibilities for drug development.
Collapse
Affiliation(s)
- Jillian G Baker
- Cell Signalling, Medical School, Queen's Medical Centre, University of Nottingham, Nottingham, UK.
- Department of Respiratory Medicine, Nottingham University Hospitals NHS Trust, Nottingham, UK.
| | - Roger J Summers
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia.
| |
Collapse
|
11
|
Zhang XW, Chen L, Chen CF, Cheng J, Zhang PP, Wang LC. Dexmedetomidine modulates neuronal activity of horizontal limbs of diagonal band via α2 adrenergic receptor in mice. BMC Anesthesiol 2023; 23:327. [PMID: 37784079 PMCID: PMC10544551 DOI: 10.1186/s12871-023-02278-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 09/11/2023] [Indexed: 10/04/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Dexmedetomidine (DEX) is widely used in clinical sedation which has little effect on cardiopulmonary inhibition, however the mechanism remains to be elucidated. The basal forebrain (BF) is a key nucleus that controls sleep-wake cycle. The horizontal limbs of diagonal bundle (HDB) is one subregions of the BF. The purpose of this study was to examine whether the possible mechanism of DEX is through the α2 adrenergic receptor of BF (HDB). METHODS In this study, we investigated the effects of DEX on the BF (HDB) by using whole cell patch clamp recordings. The threshold stimulus intensity, the inter-spike-intervals (ISIs) and the frequency of action potential firing in the BF (HDB) neurons were recorded by application of DEX (2 µM) and co-application of a α2 adrenergic receptor antagonist phentolamine (PHEN) (10 µM). RESULTS DEX (2 µM) increased the threshold stimulus intensity, inhibited the frequency of action potential firing and enlarged the inter-spike-interval (ISI) in the BF (HDB) neurons. These effects were reversed by co-application of PHEN (10 µM). CONCLUSION Taken together, our findings revealed DEX decreased the discharge activity of BF (HDB) neuron via α2 adrenergic receptors.
Collapse
Affiliation(s)
- Xia-Wei Zhang
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, 230032, Hefei, China
| | - Lei Chen
- Departments of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, 230031, Hefei, China
| | - Chang-Feng Chen
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, 230032, Hefei, China
| | - Juan Cheng
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, 230032, Hefei, China
| | - Ping-Ping Zhang
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, 230032, Hefei, China
| | - Lie-Cheng Wang
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, 230032, Hefei, China.
| |
Collapse
|
12
|
Fan S, Cheng X, Zhang P, Wang Y, Wang L, Cheng J. The α 2 Adrenoceptor Agonist and Sedative/Anaesthetic Dexmedetomidine Excites Diverse Neuronal Types in the Ventrolateral Preoptic Area of Male Mice. ASN Neuro 2023; 15:17590914231191016. [PMID: 37499170 PMCID: PMC10388635 DOI: 10.1177/17590914231191016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 06/30/2023] [Accepted: 07/13/2023] [Indexed: 07/29/2023] Open
Abstract
SUMMARY STATEMENT Dexmedetomidine is an important ICU sedative. The mechanism of dexmedetomidine is not fully understood. Activating NA(-) and NA(+) neurons in the VLPO by dexmedetomidine using polysomnography and electrophysiological recording, this may explain the unique sedative properties with rapid arousal.
Collapse
Affiliation(s)
- Sumei Fan
- Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Xinqi Cheng
- Department of Anesthesiology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Pingping Zhang
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Yuanyin Wang
- School of Stomatology, Anhui Medical University, Hefei, China
| | - Liecheng Wang
- Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Juan Cheng
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| |
Collapse
|