1
|
Holanda VAD, Oliveira MC, de Oliveira Torres CI, de Almeida Moura C, Belchior H, da Silva Junior ED, Gavioli EC. The alpha 1A antagonist tamsulosin impairs memory acquisition, consolidation and retrieval in a novel object recognition task in mice. Behav Brain Res 2024; 469:115027. [PMID: 38697302 DOI: 10.1016/j.bbr.2024.115027] [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: 03/13/2024] [Revised: 04/26/2024] [Accepted: 04/29/2024] [Indexed: 05/04/2024]
Abstract
Tamsulosin is an α1-adrenoceptor antagonist used to treat benign prostatic hyperplasia. This drug exhibits high affinity for α1A- and α1D-adrenoceptor subtypes, which are also expressed in the brain. While dementia symptoms have been reported after administration of tamsulosin in humans, studies on its effects on the rodent brain are still rare. The present study investigated the effects of tamsulosin (and biperiden, an amnesic drug) on cognitive performance in the object recognition task (ORT). Tamsulosin (0.001-0.01 mg/kg) was orally administrated in mice at three distinct time points: pre-training, post-training and pre-test session. Tamsulosin 0.01 mg/kg impaired object recognition regardless of when it was injected, whereas at lower doses did not affect mouse performance in the ORT. Biperiden also impaired acquisition and consolidation of object recognition in mice. Furthermore, the effects of tamsulosin on locomotion, motivation and anxiety were excluded as potential confounding factors. At all doses tested, tamsulosin did not alter distance moved, time spent exploring objects in the ORT, and anxiety-related behaviors in the elevated plus-maze test. By contrast, diazepam evoked a significant reduction of anxiety-like behaviours. In conclusion, tamsulosin impaired memory acquisition, consolidation and retrieval in an object recognition task in mice, thus affecting memory performance in a non-specific phase manner. These findings contribute to our understanding of the potential adverse effects of tamsulosin, and shed light on the role played by α1-adrenoceptors, particularly α1A- subtype, in cognitive processes.
Collapse
Affiliation(s)
- Victor A D Holanda
- Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte, Av. Senador Salgado Filho, Campus Universitário, Lagoa Nova, Natal 59078-900, Brazil
| | - Matheus C Oliveira
- Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte, Av. Senador Salgado Filho, Campus Universitário, Lagoa Nova, Natal 59078-900, Brazil
| | - Carina I de Oliveira Torres
- Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte, Av. Senador Salgado Filho, Campus Universitário, Lagoa Nova, Natal 59078-900, Brazil
| | - Clarissa de Almeida Moura
- Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte, Av. Senador Salgado Filho, Campus Universitário, Lagoa Nova, Natal 59078-900, Brazil
| | - Hindiael Belchior
- Department of Physical Education, Federal University of Rio Grande do Norte, Av. Senador Salgado Filho, Campus Universitário, Lagoa Nova, Natal 59078-900, Brazil
| | - Edilson D da Silva Junior
- Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte, Av. Senador Salgado Filho, Campus Universitário, Lagoa Nova, Natal 59078-900, Brazil
| | - Elaine C Gavioli
- Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte, Av. Senador Salgado Filho, Campus Universitário, Lagoa Nova, Natal 59078-900, Brazil.
| |
Collapse
|
2
|
Ozdemir E. Adrenergic receptor system as a pharmacological target in the treatment of epilepsy (Review). MEDICINE INTERNATIONAL 2024; 4:20. [PMID: 38476984 PMCID: PMC10928664 DOI: 10.3892/mi.2024.144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 02/16/2024] [Indexed: 03/14/2024]
Abstract
Epilepsy is a complex and common neurological disorder characterized by spontaneous and recurrent seizures, affecting ~75 million individuals worldwide. Numerous studies have been conducted to develop new pharmacological drugs for the effective treatment of epilepsy. In recent years, numerous experimental and clinical studies have focused on the role of the adrenergic receptor (AR) system in the regulation of epileptogenesis, seizure susceptibility and convulsions. α1-ARs (α1A, α1B and α1D), α2-ARs (α2A, α2B and α2C) and β-ARs (β1, β2 and β3), known to have convulsant or anticonvulsant effects, have been isolated. Norepinephrine (NE), the key endogenous agonist of ARs, is considered to play a crucial role in the pathophysiology of epileptic seizures. However, the effects of NE on different ARs have not been fully elucidated. Although the activation of some AR subtypes produces conflicting results, the activation of α1, α2 and β receptor subtypes, in particular, produces anticonvulsant effects. The present review focuses on NE and ARs involved in epileptic seizure formation and discusses therapeutic approaches.
Collapse
Affiliation(s)
- Ercan Ozdemir
- Department of Physiology, Faculty of Medicine, Sivas Cumhuriyet University, 58140 Sivas, Turkey
| |
Collapse
|
3
|
Abdul-Ridha A, de Zhang LA, Betrie AH, Deluigi M, Vaid TM, Whitehead A, Zhang Y, Davis B, Harris R, Simmonite H, Hubbard RE, Gooley PR, Plückthun A, Bathgate RA, Chalmers DK, Scott DJ. Identification of a Novel Subtype-Selective α 1B-Adrenoceptor Antagonist. ACS Chem Neurosci 2024; 15:671-684. [PMID: 38238043 PMCID: PMC10854767 DOI: 10.1021/acschemneuro.3c00767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 12/19/2023] [Accepted: 12/20/2023] [Indexed: 02/08/2024] Open
Abstract
α1A-, α1B-, and α1D-adrenoceptors (α1-ARs) are members of the adrenoceptor G protein-coupled receptor family that are activated by adrenaline (epinephrine) and noradrenaline. α1-ARs are clinically targeted using antagonists that have minimal subtype selectivity, such as prazosin and tamsulosin, to treat hypertension and benign prostatic hyperplasia, respectively. Abundant expression of α1-ARs in the heart and central nervous system (CNS) makes these receptors potential targets for the treatment of cardiovascular and CNS disorders, such as heart failure, epilepsy, and Alzheimer's disease. Our understanding of the precise physiological roles of α1-ARs, however, and their involvement in disease has been hindered by the lack of sufficiently subtype-selective tool compounds, especially for α1B-AR. Here, we report the discovery of 4-[(2-hydroxyethyl)amino]-6-methyl-2H-chromen-2-one (Cpd1), as an α1B-AR antagonist that has 10-15-fold selectivity over α1A-AR and α1D-AR. Through computational and site-directed mutagenesis studies, we have identified the binding site of Cpd1 in α1B-AR and propose the molecular basis of α1B-AR selectivity, where the nonconserved V19745.52 residue plays a major role, with contributions from L3146.55 within the α1B-AR pocket. By exploring the structure-activity relationships of Cpd1 at α1B-AR, we have also identified 3-[(cyclohexylamino)methyl]-6-methylquinolin-2(1H)-one (Cpd24), which has a stronger binding affinity than Cpd1, albeit with reduced selectivity for α1B-AR. Cpd1 and Cpd24 represent potential leads for α1B-AR-selective drug discovery and novel tool molecules to further study the physiology of α1-ARs.
Collapse
Affiliation(s)
- Alaa Abdul-Ridha
- The
Florey Institute, The University of Melbourne, 30 Royal Parade, Parkville, Victoria 3052, Australia
| | - Lazarus A. de Zhang
- The
Florey Institute, The University of Melbourne, 30 Royal Parade, Parkville, Victoria 3052, Australia
| | | | - Mattia Deluigi
- Department
of Biochemistry, University of Zurich, CH-8057 Zurich, Switzerland
| | - Tasneem M. Vaid
- The
Florey Institute, The University of Melbourne, 30 Royal Parade, Parkville, Victoria 3052, Australia
- The
Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, Victoria 3010, Australia
- The Bio21
Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Alice Whitehead
- The
Florey Institute, The University of Melbourne, 30 Royal Parade, Parkville, Victoria 3052, Australia
| | - Yifan Zhang
- The
Florey Institute, The University of Melbourne, 30 Royal Parade, Parkville, Victoria 3052, Australia
| | - Ben Davis
- Vernalis
(R&D) Ltd, Granta Park, Cambridge CB21 6GB, U.K.
| | - Richard Harris
- Vernalis
(R&D) Ltd, Granta Park, Cambridge CB21 6GB, U.K.
| | | | - Roderick E. Hubbard
- Vernalis
(R&D) Ltd, Granta Park, Cambridge CB21 6GB, U.K.
- Department
of Chemistry, University of York, York YO10 5DD, U.K.
| | - Paul R. Gooley
- The
Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, Victoria 3010, Australia
- The Bio21
Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Andreas Plückthun
- Department
of Biochemistry, University of Zurich, CH-8057 Zurich, Switzerland
| | - Ross A.D. Bathgate
- The
Florey Institute, The University of Melbourne, 30 Royal Parade, Parkville, Victoria 3052, Australia
- The
Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - David K. Chalmers
- Medicinal
Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Daniel J. Scott
- The
Florey Institute, The University of Melbourne, 30 Royal Parade, Parkville, Victoria 3052, Australia
- The
Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
4
|
Chodari L, Ghasemi M, Mehranfard N. Alterations in expression of α1-adrenergic receptors possibly are involved in prevention of age-associated apoptosis in rat hippocampus by treadmill exercise. JOURNAL OF COMPLEMENTARY & INTEGRATIVE MEDICINE 2023; 20:707-713. [PMID: 37428454 DOI: 10.1515/jcim-2023-0114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 06/23/2023] [Indexed: 07/11/2023]
Abstract
OBJECTIVES Exercise is assumed to attenuate age-related neuronal apoptosis, but the detailed mechanism(s) is not fully understood. α1-Adrenergic receptors (ARs) can either trigger or suppress apoptosis, therefore, here we determined the impact of treadmill exercise on the expression of the apoptosis regulatory proteins as well as α1-AR subtypes α1A- and α1B-ARs, in order to elucidate a possible association between apoptosis and the hippocampal expression of α1-ARs in aged male rats. METHODS Twenty-one male Wistar rats were divided into 3 groups (n=7): young control, aged sedentary, and aged + exercise. Western blot for α1A- and α1B-ARs as well as pro-(Bax and p53) and anti-apoptotic (Bcl2) proteins was conducted. An 8-week regular moderate-intensity treadmill exercise intervention was carried out in exercise group. RESULTS In aged rats, α1A-AR expression in the hippocampus was significantly increased, and exercise markedly prevented this event. While α1B-AR expression was no altered with aging, a marked reduction in α1B-AR level was detected in exercise group when compared to aged group. Furthermore, pro-apoptotic protein levels of Bax and p53 were upregulated and anti-apoptotic protein Bcl2 was downregulated in the aging hippocampus, but could be reversed by treadmill exercise. In the present research, exercise-induced reduction in α1A- and α1B-ARs was associated with an obvious downregulation of Bax/Bcl2 ratio in aged rats, suggesting that exercise may inhibit apoptosis through regulating α1-ARs, particularly α1A-AR. CONCLUSIONS Our study suggests that manipulations attenuating α1-AR activity, including nonselective α1-adrenergic antagonists, may protect against hippocampal neurodegeneration in aging brains.
Collapse
Affiliation(s)
- Leila Chodari
- Neurophysiology Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
- Department of Physiology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Maedeh Ghasemi
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nasrin Mehranfard
- Neurophysiology Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
5
|
Kaczor A, Knutelska J, Kucwaj-Brysz K, Zygmunt M, Żesławska E, Siwek A, Bednarski M, Podlewska S, Jastrzębska-Więsek M, Nitek W, Sapa J, Handzlik J. The Subtype Selectivity in Search of Potent Hypotensive Agents among 5,5-Dimethylhydantoin Derived α 1-Adrenoceptors Antagonists. Int J Mol Sci 2023; 24:16609. [PMID: 38068933 PMCID: PMC10706087 DOI: 10.3390/ijms242316609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 11/18/2023] [Accepted: 11/19/2023] [Indexed: 12/18/2023] Open
Abstract
In order to find new hypotensive drugs possessing higher activity and better selectivity, a new series of fifteen 5,5-dimethylhydantoin derivatives (1-15) was designed. Three-step syntheses, consisting of N-alkylations using standard procedures as well as microwaves, were carried out. Crystal structures were determined for compounds 7-9. All of the synthesized 5,5-dimethylhydantoins were tested for their affinity to α1-adrenergic receptors (α1-AR) using both in vitro and in silico methods. Most of them displayed higher affinity (Ki < 127.9 nM) to α1-adrenoceptor than urapidil in radioligand binding assay. Docking to two subtypes of adrenergic receptors, α1A and α1B, was conducted. Selected compounds were tested for their activity towards two α1-AR subtypes. All of them showed intrinsic antagonistic activity. Moreover, for two compounds (1 and 5), which possess o-methoxyphenylpiperazine fragments, strong activity (IC50 < 100 nM) was observed. Some representatives (3 and 5), which contain alkyl linker, proved selectivity towards α1A-AR, while two compounds with 2-hydroxypropyl linker (11 and 13) to α1B-AR. Finally, hypotensive activity was examined in rats. The most active compound (5) proved not only a lower effective dose than urapidil but also a stronger effect than prazosin.
Collapse
Affiliation(s)
- Aneta Kaczor
- Department of Technology and Biotechnology of Drugs, Medical College, Jagiellonian University, Medyczna 9, 30-688 Krakow, Poland; (A.K.); (K.K.-B.)
| | - Joanna Knutelska
- Department of Pharmacodynamics, Medical College, Jagiellonian University, Medyczna 9, 30-688 Krakow, Poland; (J.K.); (M.Z.); (M.B.); (J.S.)
| | - Katarzyna Kucwaj-Brysz
- Department of Technology and Biotechnology of Drugs, Medical College, Jagiellonian University, Medyczna 9, 30-688 Krakow, Poland; (A.K.); (K.K.-B.)
| | - Małgorzata Zygmunt
- Department of Pharmacodynamics, Medical College, Jagiellonian University, Medyczna 9, 30-688 Krakow, Poland; (J.K.); (M.Z.); (M.B.); (J.S.)
| | - Ewa Żesławska
- Institute of Biology and Earth Sciences, University of the National Education Commision, Podchorążych 2, 30-084 Krakow, Poland;
| | - Agata Siwek
- Department of Pharmacobiology, Medical College, Jagiellonian University, Medyczna 9, 30-688 Krakow, Poland;
| | - Marek Bednarski
- Department of Pharmacodynamics, Medical College, Jagiellonian University, Medyczna 9, 30-688 Krakow, Poland; (J.K.); (M.Z.); (M.B.); (J.S.)
| | - Sabina Podlewska
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Medicinal Chemistry, Smętna 12, 31-343 Krakow, Poland;
| | | | - Wojciech Nitek
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Krakow, Poland;
| | - Jacek Sapa
- Department of Pharmacodynamics, Medical College, Jagiellonian University, Medyczna 9, 30-688 Krakow, Poland; (J.K.); (M.Z.); (M.B.); (J.S.)
| | - Jadwiga Handzlik
- Department of Technology and Biotechnology of Drugs, Medical College, Jagiellonian University, Medyczna 9, 30-688 Krakow, Poland; (A.K.); (K.K.-B.)
| |
Collapse
|
6
|
Neubert E, Rassler B, Hoschke A, Raffort C, Salameh A. Effects of Normobaric Hypoxia and Adrenergic Blockade over 72 h on Cardiac Function in Rats. Int J Mol Sci 2023; 24:11417. [PMID: 37511176 PMCID: PMC10379660 DOI: 10.3390/ijms241411417] [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: 05/17/2023] [Revised: 07/06/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
In rats, acute normobaric hypoxia depressed left ventricular (LV) inotropic function. After 24 h of hypoxic exposure, a slight recovery of LV function occurred. We speculated that prolonged hypoxia (72 h) would induce acclimatization and, hence, recovery of LV function. Moreover, we investigated biomarkers of nitrosative stress and apoptosis as possible causes of hypoxic LV depression. To elucidate the role of hypoxic sympathetic activation, we studied whether adrenergic blockade would further deteriorate the general state of the animals and their cardiac function. Ninety-four rats were exposed over 72 h either to normal room air (N) or to normobaric hypoxia (H). The rodents received infusion (0.1 mL/h) with 0.9% NaCl or with different adrenergic blockers. Despite clear signs of acclimatization to hypoxia, the LV depression continued persistently after 72 h of hypoxia. Immunohistochemical analyses revealed significant increases in markers of nitrosative stress, adenosine triphosphate deficiency and apoptosis in the myocardium, which could provide a possible explanation for the absence of LV function recovery. Adrenergic blockade had a slightly deteriorative effect on the hypoxic LV function compared to the hypoxic group with maintained sympathetic efficacy. These findings show that hypoxic sympathetic activation compensates, at least partially, for the compromised function in hypoxic conditions, therefore emphasizing its importance for hypoxia adaptation.
Collapse
Affiliation(s)
- Elias Neubert
- Carl-Ludwig-Institute of Physiology, University of Leipzig, 04103 Leipzig, Germany; (E.N.); (A.H.)
| | - Beate Rassler
- Carl-Ludwig-Institute of Physiology, University of Leipzig, 04103 Leipzig, Germany; (E.N.); (A.H.)
| | - Annekathrin Hoschke
- Carl-Ludwig-Institute of Physiology, University of Leipzig, 04103 Leipzig, Germany; (E.N.); (A.H.)
| | - Coralie Raffort
- Department of Pediatric Cardiology, Heart Centre, University of Leipzig, 04289 Leipzig, Germany; (C.R.); (A.S.)
| | - Aida Salameh
- Department of Pediatric Cardiology, Heart Centre, University of Leipzig, 04289 Leipzig, Germany; (C.R.); (A.S.)
| |
Collapse
|
7
|
Papay RS, Stauffer SR, Perez DM. A PAM of the α 1A-Adrenergic receptor rescues biomarker, long-term potentiation, and cognitive deficits in Alzheimer's disease mouse models without effects on blood pressure. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2023; 5:100160. [PMID: 37448695 PMCID: PMC10336260 DOI: 10.1016/j.crphar.2023.100160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 05/30/2023] [Accepted: 06/25/2023] [Indexed: 07/15/2023] Open
Abstract
α1-Adrenergic Receptors (ARs) regulate the sympathetic nervous system by the binding of norepinephrine (NE) and epinephrine (Epi) through different subtypes (α1A, α1B, α1D). α1A-AR activation is hypothesized to be memory forming and cognitive enhancing but drug development has been stagnant due to unwanted side effects on blood pressure. We recently reported the pharmacological characterization of the first positive allosteric modulator (PAM) for the α1A-AR with predictive pro-cognitive and memory properties. In this report, we now demonstrate the in vivo characteristics of Compound 3 (Cmpd-3) in two genetically-different Alzheimer's Disease (AD) mouse models. Drug metabolism and pharmacokinetic studies indicate sufficient brain penetrance and rapid uptake into the brain with low to moderate clearance, and a favorable inhibition profile against the major cytochrome p450 enzymes. Oral administration of Cmpd-3 (3-9 mg/kg QD) can fully rescue long-term potentiation defects and AD biomarker profile (amyloid β-40, 42) within 3 months of dosing to levels that were non-significant from WT controls and which outperformed donepezil (1 mg/kg QD). There were also significant effects on paired pulse facilitation and cognitive behavior. Long-term and high-dose in vivo studies with Cmpd-3 revealed no effects on blood pressure. Our results suggest that Cmpd-3 can maintain lasting therapeutic levels and efficacy with disease modifying effects with a once per day dosing regimen in AD mouse models with no observed side effects.
Collapse
Affiliation(s)
- Robert S. Papay
- The Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, The Cleveland Clinic Foundation, 9500 Euclid Ave, Cleveland, OH, 44195, USA
| | - Shaun R. Stauffer
- Center of Therapeutics Discovery, Lerner Research Institute, The Cleveland Clinic Foundation, 9500 Euclid Ave, Cleveland, Ohio, 44195, USA
| | - Dianne M. Perez
- The Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, The Cleveland Clinic Foundation, 9500 Euclid Ave, Cleveland, OH, 44195, USA
| |
Collapse
|
8
|
Perez DM. α 1-Adrenergic Receptors: Insights into Potential Therapeutic Opportunities for COVID-19, Heart Failure, and Alzheimer's Disease. Int J Mol Sci 2023; 24:4188. [PMID: 36835598 PMCID: PMC9963459 DOI: 10.3390/ijms24044188] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/06/2023] [Accepted: 02/08/2023] [Indexed: 02/22/2023] Open
Abstract
α1-Adrenergic receptors (ARs) are members of the G-Protein Coupled Receptor superfamily and with other related receptors (β and α2), they are involved in regulating the sympathetic nervous system through binding and activation by norepinephrine and epinephrine. Traditionally, α1-AR antagonists were first used as anti-hypertensives, as α1-AR activation increases vasoconstriction, but they are not a first-line use at present. The current usage of α1-AR antagonists increases urinary flow in benign prostatic hyperplasia. α1-AR agonists are used in septic shock, but the increased blood pressure response limits use for other conditions. However, with the advent of genetic-based animal models of the subtypes, drug design of highly selective ligands, scientists have discovered potentially newer uses for both agonists and antagonists of the α1-AR. In this review, we highlight newer treatment potential for α1A-AR agonists (heart failure, ischemia, and Alzheimer's disease) and non-selective α1-AR antagonists (COVID-19/SARS, Parkinson's disease, and posttraumatic stress disorder). While the studies reviewed here are still preclinical in cell lines and rodent disease models or have undergone initial clinical trials, potential therapeutics discussed here should not be used for non-approved conditions.
Collapse
Affiliation(s)
- Dianne M Perez
- The Lerner Research Institute, The Cleveland Clinic Foundation, 9500 Euclid Ave, Cleveland, OH 44195, USA
| |
Collapse
|
9
|
Fernandez CJ, Hanna FW, Pacak K, Nazari MA. Catecholamines and blood pressure regulation. ENDOCRINE HYPERTENSION 2023:19-34. [DOI: 10.1016/b978-0-323-96120-2.00010-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
10
|
Bustos-Gómez CI, Gasca-Martínez D, Yáñez-Barrientos E, Hidalgo-Figueroa S, Gonzalez-Rivera ML, Barragan-Galvez JC, Zapata-Morales JR, Isiordia-Espinoza M, Corrales-Escobosa AR, Alonso-Castro AJ. Neuropharmacological Activities of Ceiba aesculifolia (Kunth) Britten & Baker f (Malvaceae). Pharmaceuticals (Basel) 2022; 15:ph15121580. [PMID: 36559031 PMCID: PMC9785833 DOI: 10.3390/ph15121580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 12/11/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Ceiba aesculifolia (Kunth) Britten & Baker f (Malvaceae) is used for the folk treatment of mood disorders. C. aesculifolia bark was extracted in ethanol, and the extract (CAE) was chemically standardized using gas chromatography-mass spectrometry (GC-MS). This study evaluated the effects of CAE (10-100 mg/kg p.o.) on anxiolytic-like activity, sedation, locomotor activity, depression-like activity, and spatial working memory using in vivo rodent models. A possible mechanism for the anxiolytic-like and antidepressant-like actions induced by CAE was assessed using neurotransmission pathway inhibitors. Myristic acid was one of the compounds found in CAE using GC-MS. This study also evaluated the anxiolytic-like activity and the sedative actions of myristic acid and assessed a possible mechanism of action using neurotransmission pathway inhibitors and an in silico analysis. CAE elicited anxiolytic-like activity and antidepressant-like effects (ED50 = 57 mg/kg). CAE (10-100 mg/kg) did not affect locomotor coordination or induce sedation. The anxiolytic-like and antidepressant-like actions of CAE were reverted by prazosin, suggesting a possible participation of the noradrenergic system. The anxiolytic-like activity of myristic acid was reverted by the co-administration of prazosin and partially reverted by ketanserin. The docking study revealed that myristic acid can form favorable interactions within 5-HT2A and α1A-adrenoreceptor binding pockets.
Collapse
Affiliation(s)
- Chrystyan Iván Bustos-Gómez
- Maestría en Ciencias Farmacéuticas, División de Ciencias Naturales y Exactas, Universidad de Guanajuato, Guanajuato 36200, Mexico
| | - Deisy Gasca-Martínez
- Unidad de Análisis Conductual, Instituto de Neurobiología, Campus UNAM-Juriquilla, Juriquilla 76230, Mexico
- Correspondence: (D.G.-M.); (A.J.A.-C.); Tel.: +52-473-732-0006 (ext. 1441) (D.G.-M.); Tel.: +52-(442)-2381036 (ext. 340365) (A.J.A.-C.)
| | - Eunice Yáñez-Barrientos
- Departamento de Química, División de Ciencias Naturales y Exactas, Universidad de Guanajuato, Guanajuato 36200, Mexico
| | - Sergio Hidalgo-Figueroa
- CONACyT-División de Biología Molecular/Instituto Potosino de Investigación Científica y Tecnológica A. C., San Luis Potosí 78216, Mexico
| | - Maria L. Gonzalez-Rivera
- Departamento de Farmacia, División de Ciencias Naturales y Exactas, Universidad de Guanajuato, Guanajuato 36200, Mexico
| | - Juan Carlos Barragan-Galvez
- Departamento de Farmacia, División de Ciencias Naturales y Exactas, Universidad de Guanajuato, Guanajuato 36200, Mexico
| | - Juan Ramón Zapata-Morales
- Departamento de Farmacia, División de Ciencias Naturales y Exactas, Universidad de Guanajuato, Guanajuato 36200, Mexico
| | - Mario Isiordia-Espinoza
- Instituto de Investigación en Ciencias Médicas, Departamento de Clínicas, División de Ciencias Biomédicas, Centro Universitario de los Altos, Universidad de Guadalajara, Tepatitlán de Morelos, Jalisco 47620, Mexico
| | - Alma Rosa Corrales-Escobosa
- CONACyT-División de Biología Molecular/Instituto Potosino de Investigación Científica y Tecnológica A. C., San Luis Potosí 78216, Mexico
| | - Angel Josabad Alonso-Castro
- Instituto de Investigación en Ciencias Médicas, Departamento de Clínicas, División de Ciencias Biomédicas, Centro Universitario de los Altos, Universidad de Guadalajara, Tepatitlán de Morelos, Jalisco 47620, Mexico
- Correspondence: (D.G.-M.); (A.J.A.-C.); Tel.: +52-473-732-0006 (ext. 1441) (D.G.-M.); Tel.: +52-(442)-2381036 (ext. 340365) (A.J.A.-C.)
| |
Collapse
|
11
|
Theory and Applications of the (Cardio) Genomic Fabric Approach to Post-Ischemic and Hypoxia-Induced Heart Failure. J Pers Med 2022; 12:jpm12081246. [PMID: 36013195 PMCID: PMC9410512 DOI: 10.3390/jpm12081246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/21/2022] [Accepted: 07/27/2022] [Indexed: 11/16/2022] Open
Abstract
The genomic fabric paradigm (GFP) characterizes the transcriptome topology by the transcripts’ abundances, the variability of the expression profile, and the inter-coordination of gene expressions in each pathophysiological condition. The expression variability analysis provides an indirect estimate of the cell capability to limit the stochastic fluctuations of the expression levels of key genes, while the expression coordination analysis determines the gene networks in functional pathways. This report illustrates the theoretical bases and the mathematical framework of the GFP with applications to our microarray data from mouse models of post ischemic, and constant and intermittent hypoxia-induced heart failures. GFP analyses revealed the myocardium priorities in keeping the expression of key genes within narrow intervals, determined the statistically significant gene interlinkages, and identified the gene master regulators in the mouse heart left ventricle under normal and ischemic conditions. We quantified the expression regulation, alteration of the expression control, and remodeling of the gene networks caused by the oxygen deprivation and determined the efficacy of the bone marrow mono-nuclear stem cell injections to restore the normal transcriptome. Through the comprehensive assessment of the transcriptome, GFP would pave the way towards the development of personalized gene therapy of cardiac diseases.
Collapse
|
12
|
Deluigi M, Morstein L, Schuster M, Klenk C, Merklinger L, Cridge RR, de Zhang LA, Klipp A, Vacca S, Vaid TM, Mittl PRE, Egloff P, Eberle SA, Zerbe O, Chalmers DK, Scott DJ, Plückthun A. Crystal structure of the α 1B-adrenergic receptor reveals molecular determinants of selective ligand recognition. Nat Commun 2022; 13:382. [PMID: 35046410 PMCID: PMC8770593 DOI: 10.1038/s41467-021-27911-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 12/21/2021] [Indexed: 11/25/2022] Open
Abstract
α-adrenergic receptors (αARs) are G protein-coupled receptors that regulate vital functions of the cardiovascular and nervous systems. The therapeutic potential of αARs, however, is largely unexploited and hampered by the scarcity of subtype-selective ligands. Moreover, several aminergic drugs either show off-target binding to αARs or fail to interact with the desired subtype. Here, we report the crystal structure of human α1BAR bound to the inverse agonist (+)-cyclazosin, enabled by the fusion to a DARPin crystallization chaperone. The α1BAR structure allows the identification of two unique secondary binding pockets. By structural comparison of α1BAR with α2ARs, and by constructing α1BAR-α2CAR chimeras, we identify residues 3.29 and 6.55 as key determinants of ligand selectivity. Our findings provide a basis for discovery of α1BAR-selective ligands and may guide the optimization of aminergic drugs to prevent off-target binding to αARs, or to elicit a selective interaction with the desired subtype. This study reports the X-ray structure of the α1B-adrenergic G protein-coupled receptor bound to an inverse agonist, and unveils key determinants of subtype-selective ligand binding that may help the design of aminergic drugs with fewer side-effects.
Collapse
Affiliation(s)
- Mattia Deluigi
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
| | - Lena Morstein
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
| | - Matthias Schuster
- Department of Chemistry, University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
| | - Christoph Klenk
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
| | - Lisa Merklinger
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland.,Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads, 2800 Kgs, Lyngby, Denmark
| | - Riley R Cridge
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, 30 Royal Parade, Parkville, VIC, 3052, Australia
| | - Lazarus A de Zhang
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, 30 Royal Parade, Parkville, VIC, 3052, Australia.,Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Alexander Klipp
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland.,Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg 1-5/10, CH-8093, Zurich, Switzerland
| | - Santiago Vacca
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
| | - Tasneem M Vaid
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Peer R E Mittl
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
| | - Pascal Egloff
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
| | - Stefanie A Eberle
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland.,Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark
| | - Oliver Zerbe
- Department of Chemistry, University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
| | - David K Chalmers
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
| | - Daniel J Scott
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, 30 Royal Parade, Parkville, VIC, 3052, Australia. .,Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, VIC, 3010, Australia.
| | - Andreas Plückthun
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland.
| |
Collapse
|
13
|
Chen J, Campbell AP, Wakelin LPG, Finch AM. Characterisation of bis(4-aminoquinoline)s as α 1A adrenoceptor allosteric modulators. Eur J Pharmacol 2021; 916:174659. [PMID: 34871559 DOI: 10.1016/j.ejphar.2021.174659] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 11/19/2021] [Accepted: 11/29/2021] [Indexed: 11/30/2022]
Abstract
The development of sub-type selective α1 adrenoceptor ligands has been hampered by the high sequence similarity of the amino acids forming the orthosteric binding pocket of the three α1 adrenoceptor subtypes, along with other biogenic amine receptors. One possible approach to overcome this issue is to target allosteric sites on the α1 adrenoceptors. Previous docking studies suggested that one of the quinoline moieties of a bis(4-aminoquinoline), comprising a 9-carbon methylene linker attached via the amine groups, could interact with residues outside of the orthosteric binding site while, simultaneously, the other quinoline moiety bound within the orthosteric site. We therefore hypothesized that this compound could act in a bitopic manner, displaying both orthosteric and allosteric binding properties. To test this proposition, we investigated the allosteric activity of a series of bis(4-aminoquinoline)s with linker lengths ranging from 2 to 12 methylene units (designated C2-C12). A linear trend of increasing [3H]prazosin dissociation rate with increasing linker length between C7 and C11 was observed, confirming their action as allosteric modulators. These data suggest that the optimal linker length for the bis(4-aminoquinoline)s to occupy the allosteric site of the α1A adrenoceptor is between 7 and 11 methylene units. In addition, the ability of C9 bis(4-aminoquinoline) to modulate the activation of the α1A adrenoceptor by norepinephrine was subsequently examined, showing that C9 acted as a non-competitive antagonist. Our findings indicate that the bis(4-aminoquinolines) are acting as allosteric modulators of orthosteric ligand binding, but not efficacy, in a bitopic manner.
Collapse
Affiliation(s)
- Junli Chen
- Department of Pharmacology, School of Medical Sciences, UNSW Australia, Sydney, NSW, 2052, Australia
| | - Adrian P Campbell
- Department of Pharmacology, School of Medical Sciences, UNSW Australia, Sydney, NSW, 2052, Australia
| | - Laurence P G Wakelin
- Department of Pharmacology, School of Medical Sciences, UNSW Australia, Sydney, NSW, 2052, Australia
| | - Angela M Finch
- Department of Pharmacology, School of Medical Sciences, UNSW Australia, Sydney, NSW, 2052, Australia.
| |
Collapse
|
14
|
Holanda VAD, Oliveira MC, da Silva Junior ED, Gavioli EC. Tamsulosin facilitates depressive-like behaviors in mice: Involvement of endogenous glucocorticoids. Brain Res Bull 2021; 178:29-36. [PMID: 34798218 DOI: 10.1016/j.brainresbull.2021.11.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 11/10/2021] [Accepted: 11/11/2021] [Indexed: 02/04/2023]
Abstract
The benign prostatic hyperplasia (BPH) is the main source of lower urinary tract symptoms. The BPH is a common age-dependent disease and tamsulosin is an α1-adrenoceptor blocker widely prescribed for BPH. Beyond the common adverse effects of tamsulosin, increased diagnosis of dementia after prescription was observed. Importantly, a clinical study suggested that tamsulosin may exert antidepressant effects in BPH patients. Considering the expression of α1-adrenoceptors in the brain, this study aimed to investigate the effects of tamsulosin in the forced swimming and open field tests in mice. For this, tamsulosin (0.001-1 mg/kg) was orally administered subacutely (1, 5 and 23 hr) and acutely (60 min) before tests. Mifepristone (10 mg/kg), a glucocorticoid receptor antagonist, and aminoglutethimide (10 mg/kg), a streoidogenesis inhibitor, were intraperitoneally injected before tamsulosin to investigate the role of the hypothalamic-pituitary-adrenal axis in the mediation of tamsulosin-induced effects. Subacute and acute administrations of tamsulosin increased the immobility time in the first exposition to an inescapable stressful situation. In the re-exposition to the swim task, controls displayed a natural increase in the immobility time, and the treatment with tamsulosin further increased this behavioral parameter. Tamsuslosin did not affect spontaneous locomotion neither in naïve nor in stressed mice. Our findings also showed that mifepristone and aminoglutethimide prevented the tamsulosin-induced increase in the immobility time in the first and second swimming sessions, respectively. In conclusion, tamsulosin may contribute to increased susceptibility to depressive-like behaviors, by facilitating the acquisition of a passive stress-copying strategy. These effects seem to be dependent on endogenous glucocorticoids.
Collapse
Affiliation(s)
- Victor A D Holanda
- Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte, Av. Senador salgado Filho, 3000, Campus Universitário - Lagoa Nova, Natal 59078-900, Brazil
| | - Matheus C Oliveira
- Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte, Av. Senador salgado Filho, 3000, Campus Universitário - Lagoa Nova, Natal 59078-900, Brazil
| | - Edilson D da Silva Junior
- Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte, Av. Senador salgado Filho, 3000, Campus Universitário - Lagoa Nova, Natal 59078-900, Brazil
| | - Elaine C Gavioli
- Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte, Av. Senador salgado Filho, 3000, Campus Universitário - Lagoa Nova, Natal 59078-900, Brazil.
| |
Collapse
|
15
|
The Structural Determinants for α 1-Adrenergic/Serotonin Receptors Activity among Phenylpiperazine-Hydantoin Derivatives. Molecules 2021; 26:molecules26227025. [PMID: 34834117 PMCID: PMC8623851 DOI: 10.3390/molecules26227025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/15/2021] [Accepted: 11/17/2021] [Indexed: 12/26/2022] Open
Abstract
Several studies confirmed the reciprocal interactions between adrenergic and serotoninergic systems and the influence of these phenomena on the pathogenesis of anxiety. Hence, searching for chemical agents with a multifunctional pharmacodynamic profile may bring highly effective therapy for CNS disorders. This study presents a deep structural insight into the hydantoin-arylpiperazine group and their serotonin/α-adrenergic activity. The newly synthesized compounds were tested in the radioligand binding assay and the intrinsic activity was evaluated for the selected derivatives. The computer-aided SAR analysis enabled us to answer questions about the influence of particular structural fragments on selective vs. multifunctional activity. As a result of the performed investigations, there were two leading structures: (a) compound 12 with multifunctional adrenergic-serotonin activity, which is a promising candidate to be an effective anxiolytic agent; (b) compound 14 with high α1A/α1D affinity and selectivity towards α1B, which is recommended due to the elimination of probable cardiotoxic effect. The structural conclusions of this work provide significant support for future lead optimization in order to achieve the desired pharmacodynamic profile in searching for new CNS-modulating agents.
Collapse
|
16
|
Affiliation(s)
- Jean-Luc Balligand
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC) and Cliniques Universitaires Saint-Luc, University of Louvain Medical School, Brussels, Belgium
| | - Lauriane Y M Michel
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC) and Cliniques Universitaires Saint-Luc, University of Louvain Medical School, Brussels, Belgium
| |
Collapse
|
17
|
Perez DM. Targeting Adrenergic Receptors in Metabolic Therapies for Heart Failure. Int J Mol Sci 2021; 22:5783. [PMID: 34071350 PMCID: PMC8198887 DOI: 10.3390/ijms22115783] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 05/20/2021] [Accepted: 05/22/2021] [Indexed: 12/14/2022] Open
Abstract
The heart has a reduced capacity to generate sufficient energy when failing, resulting in an energy-starved condition with diminished functions. Studies have identified numerous changes in metabolic pathways in the failing heart that result in reduced oxidation of both glucose and fatty acid substrates, defects in mitochondrial functions and oxidative phosphorylation, and inefficient substrate utilization for the ATP that is produced. Recent early-phase clinical studies indicate that inhibitors of fatty acid oxidation and antioxidants that target the mitochondria may improve heart function during failure by increasing compensatory glucose oxidation. Adrenergic receptors (α1 and β) are a key sympathetic nervous system regulator that controls cardiac function. β-AR blockers are an established treatment for heart failure and α1A-AR agonists have potential therapeutic benefit. Besides regulating inotropy and chronotropy, α1- and β-adrenergic receptors also regulate metabolic functions in the heart that underlie many cardiac benefits. This review will highlight recent studies that describe how adrenergic receptor-mediated metabolic pathways may be able to restore cardiac energetics to non-failing levels that may offer promising therapeutic strategies.
Collapse
Affiliation(s)
- Dianne M Perez
- The Lerner Research Institute, The Cleveland Clinic Foundation, 9500 Euclid Ave, Cleveland, OH 44195, USA
| |
Collapse
|
18
|
A Study to Decipher the Potential Effects of Butylphthalide against Central Nervous System Diseases Based on Network Pharmacology and Molecular Docking Integration Strategy. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:6694698. [PMID: 34035826 PMCID: PMC8116153 DOI: 10.1155/2021/6694698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 04/05/2021] [Accepted: 04/27/2021] [Indexed: 11/24/2022]
Abstract
Background Butylphthalide (NBP), approved by the China National Medical Products Administration (NMPA) for the treatment of ischemic stroke (IS), showed pleiotropic potentials against central nervous system (CNS) diseases, including neuroprotection and cognitive deficits improvement. However, the effects and corresponding modes of action were not fully explored. This study was designed to investigate the potential of NBP against IS-associated CNS diseases based on network pharmacology (NP) and molecular docking (MD). Methods IS was inputted as the index disease to retrieve the “associated diseases” in DisGeNET. Three-database-based IS genes were obtained and integrated (DisGeNET, Malacards, and OMIM). Then, IS-associated genes were identified by combining these genes. Meanwhile, PubMed references and online databases were applied to identify NBP target genes. The IS-related disease-disease association (DDA) network and NBP-disease regulation network were constructed and analyzed in Cytoscape. In silico MD and references were used to validate the binding affinity of NBP with critical targets and the potential of NBP against certain IS-related CNS disease regulation. Results 175 NBP target genes were obtained, while 312 IS-related disease genes were identified. 36 NBP target genes were predicted to be associated with IS-related CNS diseases, including Alzheimer's disease (AD), epilepsy, major depressive disorder (MDD), amyotrophic lateral sclerosis (ALS), and dementia. Six target genes (i.e., GRIN1, PTGIS, PTGES, ADRA1A, CDK5, and SULT1E1) indicating disease specificity index (DSI) >0.5 showed certain to good degree binding affinity with NBP, ranging from −9.2 to −6.7 kcal/mol. And the binding modes may be mainly related to hydrogen bonds and hydrophobic “bonds.” Further literature validations inferred that these critical NBP targets had a tight association with AD, epilepsy, ALS, and depression. Conclusions Our study proposed a drug-target-disease integrated method to predict the drug repurposing potentials to associated diseases by application of NP and MD, which could be an attractive alternative to facilitate the development of CNS disease therapies. NBP may be promising and showed potentials to be repurposed for treatments for AD, epilepsy, ALS, and depression, and further investigations are warranted to be carefully designed and conducted.
Collapse
|
19
|
Perez DM. Current Developments on the Role of α 1-Adrenergic Receptors in Cognition, Cardioprotection, and Metabolism. Front Cell Dev Biol 2021; 9:652152. [PMID: 34113612 PMCID: PMC8185284 DOI: 10.3389/fcell.2021.652152] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 04/29/2021] [Indexed: 12/13/2022] Open
Abstract
The α1-adrenergic receptors (ARs) are G-protein coupled receptors that bind the endogenous catecholamines, norepinephrine, and epinephrine. They play a key role in the regulation of the sympathetic nervous system along with β and α2-AR family members. While all of the adrenergic receptors bind with similar affinity to the catecholamines, they can regulate different physiologies and pathophysiologies in the body because they couple to different G-proteins and signal transduction pathways, commonly in opposition to one another. While α1-AR subtypes (α1A, α1B, α1C) have long been known to be primary regulators of vascular smooth muscle contraction, blood pressure, and cardiac hypertrophy, their role in neurotransmission, improving cognition, protecting the heart during ischemia and failure, and regulating whole body and organ metabolism are not well known and are more recent developments. These advancements have been made possible through the development of transgenic and knockout mouse models and more selective ligands to advance their research. Here, we will review the recent literature to provide new insights into these physiological functions and possible use as a therapeutic target.
Collapse
Affiliation(s)
- Dianne M Perez
- The Lerner Research Institute, The Cleveland Clinic Foundation, Cleveland, OH, United States
| |
Collapse
|
20
|
Joyce W, Scholman KT, Jensen B, Wang T, Boukens BJ. α 1-adrenergic stimulation increases ventricular action potential duration in the intact mouse heart. Facets (Ott) 2021. [DOI: 10.1139/facets-2020-0081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The role of α1-adrenergic receptors (α-ARs) in the regulation of myocardial function is less well-understood than that of β-ARs. Previous reports in the mouse heart have described that α1-adrenergic stimulation shortens action potential duration in isolated cells or tissues, in contrast to prolongation of the action potential reported in most other mammalian hearts. It has since become appreciated, however, that the mouse heart exhibits marked variation in inotropic response to α1-adrenergic stimulation between ventricles and even individual cardiomyocytes. We investigated the effects of α1-adrenergic stimulation on action potential duration at 80% of repolarization in the right and left ventricles of Langendorff-perfused mouse hearts using optical mapping. In hearts under β-adrenergic blockade (propranolol), phenylephrine or noradrenaline perfusion both increased action potential duration in both ventricles. The increased action potential duration was partially reversed by subsequent perfusion with the α-adrenergic antagonist phentolamine (1 μmol L−1). These data show that α1-receptor stimulation may lead to a prolonging of action potential in the mouse heart and thereby refine our understanding of how action potential duration adjusts during sympathetic stimulation.
Collapse
Affiliation(s)
- William Joyce
- Department of Biology—Zoophysiology, Aarhus University, DK-8000 Aarhus C, Denmark
- Department of Biology, University of Ottawa, 30 Marie Curie, Ottawa, ON K1N 6N5, Canada
| | - Koen T. Scholman
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, 11005 AZ Amsterdam, the Netherlands
| | - Bjarke Jensen
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, 11005 AZ Amsterdam, the Netherlands
| | - Tobias Wang
- Department of Biology—Zoophysiology, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Bastiaan J. Boukens
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, 11005 AZ Amsterdam, the Netherlands
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, 1100 DD Amsterdam, the Netherlands
| |
Collapse
|
21
|
Alluri SR, Kim SW, Volkow ND, Kil KE. PET Radiotracers for CNS-Adrenergic Receptors: Developments and Perspectives. Molecules 2020; 25:molecules25174017. [PMID: 32899124 PMCID: PMC7504810 DOI: 10.3390/molecules25174017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/29/2020] [Accepted: 09/01/2020] [Indexed: 12/30/2022] Open
Abstract
Epinephrine (E) and norepinephrine (NE) play diverse roles in our body’s physiology. In addition to their role in the peripheral nervous system (PNS), E/NE systems including their receptors are critical to the central nervous system (CNS) and to mental health. Various antipsychotics, antidepressants, and psychostimulants exert their influence partially through different subtypes of adrenergic receptors (ARs). Despite the potential of pharmacological applications and long history of research related to E/NE systems, research efforts to identify the roles of ARs in the human brain taking advantage of imaging have been limited by the lack of subtype specific ligands for ARs and brain penetrability issues. This review provides an overview of the development of positron emission tomography (PET) radiotracers for in vivo imaging of AR system in the brain.
Collapse
Affiliation(s)
- Santosh Reddy Alluri
- University of Missouri Research Reactor, University of Missouri, Columbia, MO 65211-5110, USA;
| | - Sung Won Kim
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892-1013, USA;
| | - Nora D. Volkow
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892-1013, USA;
- National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD 20892-1013, USA
- Correspondence: (N.D.V.); (K.-E.K.); Tel.: +1-(301)-443-6480 (N.D.V.); +1-(573)-884-7885 (K.-E.K.)
| | - Kun-Eek Kil
- University of Missouri Research Reactor, University of Missouri, Columbia, MO 65211-5110, USA;
- Department of Veterinary Medicine and Surgery, University of Missouri, Columbia, MO 65211, USA
- Correspondence: (N.D.V.); (K.-E.K.); Tel.: +1-(301)-443-6480 (N.D.V.); +1-(573)-884-7885 (K.-E.K.)
| |
Collapse
|
22
|
Vaid TM, Chalmers DK, Scott DJ, Gooley PR. INPHARMA-Based Determination of Ligand Binding Modes at α 1 -Adrenergic Receptors Explains the Molecular Basis of Subtype Selectivity. Chemistry 2020; 26:11796-11805. [PMID: 32291801 DOI: 10.1002/chem.202000642] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 04/02/2020] [Indexed: 01/06/2023]
Abstract
The structural poses of ligands that bind weakly to protein receptors are challenging to define. In this work we have studied ligand interactions with the adrenoreceptor (AR) subtypes, α1A -AR and α1B -AR, which belong to the G protein-coupled receptor (GPCR) superfamily, by employing the solution-based ligand-observed NMR method interligand NOEs for pharmacophore mapping (INPHARMA). A lack of receptor crystal structures and of subtype-selective drugs has hindered the definition of the physiological roles of each subtype and limited drug development. We determined the binding pose of the weakly binding α1A -AR-selective agonist A-61603 relative to an endogenous agonist, epinephrine, at both α1A -AR and α1B -AR. The NMR experimental data were quantitatively compared, by using SpINPHARMA, to the back-calculated spectra based on ligand poses obtained from all-atom molecular dynamics simulations. The results helped mechanistically explain the selectivity of (R)-A-61603 towards α1A -AR, thus demonstrating an approach for targeting subtype selectivity in ARs.
Collapse
Affiliation(s)
- Tasneem M Vaid
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, 3010, VIC, Australia.,Bio21 Molecular Science & Biotechnology Institute, University of Melbourne, Parkville, 3010, VIC, Australia.,The Florey Institute of Neuroscience & Mental Health, University of Melbourne, Parkville, 3015, VIC, Australia
| | - David K Chalmers
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, 3052, VIC, Australia
| | - Daniel J Scott
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, 3010, VIC, Australia.,The Florey Institute of Neuroscience & Mental Health, University of Melbourne, Parkville, 3015, VIC, Australia
| | - Paul R Gooley
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, 3010, VIC, Australia.,Bio21 Molecular Science & Biotechnology Institute, University of Melbourne, Parkville, 3010, VIC, Australia
| |
Collapse
|
23
|
Papay RS, Perez DM. α 1-Adrenergic receptors increase glucose oxidation under normal and ischemic conditions in adult mouse cardiomyocytes. J Recept Signal Transduct Res 2020; 41:138-144. [PMID: 32757689 DOI: 10.1080/10799893.2020.1799291] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The role of catecholamine receptors in cardiac energy metabolism is unknown. α1-adrenergic receptors (α1-ARs) have been identified to play a role in whole body metabolism but its role in cardiac energy metabolism has not been explored. We used freshly prepared primary adult mouse cardiomyocytes and incubated with either 14C-palmitate or 14C-glucose tracers to measure oxidation rates in the presence or absence of phenylephrine, an α1-AR agonist (with β and α2-AR blockers) under normal cell culture conditions. 14CO2 released was collected over a 10 min period in covered tissue culture plates using a 1 M hyamine hydroxide solution placed in well cups, counted by scintillation and converted into nmoles/hr. We found that phenylephrine stimulated glucose oxidation but not fatty acid oxidation in adult primary cardiomyocytes. α1-AR stimulated glucose oxidation was blocked by the AMPK inhibitor, dorsomorphin dihydrochloride, and the PKC inhibitor, rottlerin. Ischemic conditions were induced by lowering the glucose concentration from 22.5 mM to 1.375 mM. Under ischemic conditions, we found that phenylephrine also increased glucose oxidation. We report a direct role of α1-ARs in regulating glucose oxidation under normal and ischemic conditions that may lead to new therapeutic approaches in treating ischemia.
Collapse
Affiliation(s)
- Robert S Papay
- The Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, The Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Dianne M Perez
- The Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, The Cleveland Clinic Foundation, Cleveland, OH, USA
| |
Collapse
|
24
|
Sohn JH, Lee SH, Kwon YS, Kim JH, Kim Y, Lee JJ. The impact of tamsulosin on cognition in Alzheimer disease with benign prostate hyperplasia: A study using the Hallym Smart Clinical Data Warehouse. Medicine (Baltimore) 2020; 99:e20240. [PMID: 32481389 DOI: 10.1097/md.0000000000020240] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Studies suggest that the use of alpha-blockers increases the risk of dementia in patients with benign prostate hyperplasia (BPH). Due to study limitations, the relationship between the use of alpha-blockers, such as tamsulosin, and the risk of dementia is still unclear. However, alpha1-adrenoreceptors are also present in the brain, so there is potential for adverse effects on cognitive function. Therefore, we investigated possible associations between the use of alpha-blockers and aggravation of cognitive decline in dementia patients using a clinical data analytic solution called the Smart Clinical Data Warehouse (CDW).We retrospectively investigated clinical data using the Smart CDW of Hallym University Medical Center from 2009 to 2019. We enrolled patients with probable Alzheimer disease (AD) who had completed the Mini-Mental State Examination (MMSE) at least twice during follow-up, and who had BPH. We compared the difference in MMSE scores between patients who took tamsulosin for >1000 days and those who did not take any alpha-blocker. We tested the effect of tamsulosin on cognitive decline in patients with AD, using propensity score-matched logistic regression analysis.Eligible cases were included in the tamsulosin (n = 68) or no-medication (n = 153) groups. After propensity score matching, clinical characteristics such as educational attainment and vascular risk factors were similar in the tamsulosin and no-medication groups. The MMSE scores did not differ significantly between the tamsulosin and no-medication groups (P = .470).The results suggest that tamsulosin for BPH is not associated with worsening of the cognitive decline in patients with AD.
Collapse
Affiliation(s)
| | | | - Young-Suk Kwon
- Department of Anesthesiology and Pain Medicine, Chuncheon Sacred Heart Hospital, Hallym University College of Medicine
- Institute of New Frontier Research, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Jong-Ho Kim
- Department of Anesthesiology and Pain Medicine, Chuncheon Sacred Heart Hospital, Hallym University College of Medicine
- Institute of New Frontier Research, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Youngmi Kim
- Institute of New Frontier Research, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Jae Jun Lee
- Department of Anesthesiology and Pain Medicine, Chuncheon Sacred Heart Hospital, Hallym University College of Medicine
- Institute of New Frontier Research, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| |
Collapse
|
25
|
Wu FJ, Williams LM, Abdul-Ridha A, Gunatilaka A, Vaid TM, Kocan M, Whitehead AR, Griffin MDW, Bathgate RAD, Scott DJ, Gooley PR. Probing the correlation between ligand efficacy and conformational diversity at the α 1A-adrenoreceptor reveals allosteric coupling of its microswitches. J Biol Chem 2020; 295:7404-7417. [PMID: 32303636 PMCID: PMC7247315 DOI: 10.1074/jbc.ra120.012842] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 04/14/2020] [Indexed: 12/11/2022] Open
Abstract
G protein-coupled receptors (GPCRs) use a series of conserved microswitches to transmit signals across the cell membrane via an allosteric network encompassing the ligand-binding site and the G protein-binding site. Crystal structures of GPCRs provide snapshots of their inactive and active states, but poorly describe the conformational dynamics of the allosteric network that underlies GPCR activation. Here, we analyzed the correlation between ligand binding and receptor conformation of the α1A-adrenoreceptor, a GPCR that stimulates smooth muscle contraction in response to binding noradrenaline. NMR of [13CϵH3]methionine-labeled α1A-adrenoreceptor variants, each exhibiting differing signaling capacities, revealed how different classes of ligands modulate the conformational equilibria of this receptor. [13CϵH3]Methionine residues near the microswitches exhibited distinct states that correlated with ligand efficacies, supporting a conformational selection mechanism. We propose that allosteric coupling among the microswitches controls the conformation of the α1A-adrenoreceptor and underlies the mechanism of ligand modulation of GPCR signaling in cells.
Collapse
Affiliation(s)
- Feng-Jie Wu
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville 3052, VIC, Australia; Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville 3052, VIC, Australia; The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3052, VIC, Australia
| | - Lisa M Williams
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3052, VIC, Australia
| | - Alaa Abdul-Ridha
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3052, VIC, Australia
| | - Avanka Gunatilaka
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3052, VIC, Australia
| | - Tasneem M Vaid
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville 3052, VIC, Australia; Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville 3052, VIC, Australia; The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3052, VIC, Australia
| | - Martina Kocan
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3052, VIC, Australia
| | - Alice R Whitehead
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3052, VIC, Australia
| | - Michael D W Griffin
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville 3052, VIC, Australia; Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville 3052, VIC, Australia
| | - Ross A D Bathgate
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville 3052, VIC, Australia; The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3052, VIC, Australia
| | - Daniel J Scott
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville 3052, VIC, Australia; The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3052, VIC, Australia.
| | - Paul R Gooley
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville 3052, VIC, Australia; Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville 3052, VIC, Australia.
| |
Collapse
|
26
|
Janezic EM, Lauer SML, Williams RG, Chungyoun M, Lee KS, Navaluna E, Lau HT, Ong SE, Hague C. N-glycosylation of α 1D-adrenergic receptor N-terminal domain is required for correct trafficking, function, and biogenesis. Sci Rep 2020; 10:7209. [PMID: 32350295 PMCID: PMC7190626 DOI: 10.1038/s41598-020-64102-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 04/09/2020] [Indexed: 01/21/2023] Open
Abstract
G protein-coupled receptor (GPCR) biogenesis, trafficking, and function are regulated by post-translational modifications, including N-glycosylation of asparagine residues. α1D-adrenergic receptors (α1D-ARs) - key regulators of central and autonomic nervous system function - contain two putative N-glycosylation sites within the large N-terminal domain at N65 and N82. However, determining the glycosylation state of this receptor has proven challenging. Towards understanding the role of these putative glycosylation sites, site-directed mutagenesis and lectin affinity purification identified N65 and N82 as bona fide acceptors for N-glycans. Surprisingly, we also report that simultaneously mutating N65 and N82 causes early termination of α1D-AR between transmembrane domain 2 and 3. Label-free dynamic mass redistribution and cell surface trafficking assays revealed that single and double glycosylation deficient mutants display limited function with impaired plasma membrane expression. Confocal microscopy imaging analysis and SNAP-tag sucrose density fractionation assays revealed the dual glycosylation mutant α1D-AR is widely distributed throughout the cytosol and nucleus. Based on these novel findings, we propose α1D-AR transmembrane domain 2 acts as an ER localization signal during active protein biogenesis, and that α1D-AR N-terminal glycosylation is required for complete translation of nascent, functional receptor.
Collapse
Affiliation(s)
- Eric M Janezic
- Department of Pharmacology, School of Medicine, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98185, USA
| | - Sophia My-Linh Lauer
- Department of Pharmacology, School of Medicine, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98185, USA
| | - Robert George Williams
- Department of Pharmacology, School of Medicine, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98185, USA
| | - Michael Chungyoun
- Department of Pharmacology, School of Medicine, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98185, USA
| | - Kyung-Soon Lee
- Department of Pharmacology, School of Medicine, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98185, USA
| | - Edelmar Navaluna
- Department of Pharmacology, School of Medicine, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98185, USA
| | - Ho-Tak Lau
- Department of Pharmacology, School of Medicine, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98185, USA
| | - Shao-En Ong
- Department of Pharmacology, School of Medicine, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98185, USA
| | - Chris Hague
- Department of Pharmacology, School of Medicine, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98185, USA.
| |
Collapse
|
27
|
Shorter DI, Zhang X, Domingo CB, Nielsen EM, Kosten TR, Nielsen DA. Doxazosin treatment in cocaine use disorder: pharmacogenetic response based on an alpha-1 adrenoreceptor subtype D genetic variant. THE AMERICAN JOURNAL OF DRUG AND ALCOHOL ABUSE 2020; 46:184-193. [PMID: 31914324 DOI: 10.1080/00952990.2019.1674864] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Background: The α1 antagonist doxazosin reduces cocaine use in individuals with cocaine use disorder (CUD) through a functional polymorphism of the α1 adrenoreceptor. The regulatory role of the α1 adrenoreceptor subtype D (ADRA1D) gene polymorphism in CUD is uncharacterized.Objectives: To study how the genetic variant of ADRA1D gene (T1848A, rs2236554) may affect the treatment efficacy of doxazosin in reducing cocaine use.Methods: This 12-week pilot trial included 76 participants with CUD with ADRA1D (T1848A, rs2236554) AA (N = 40) or AT/TT genotype (N = 36). Participants were randomized to doxazosin (8 mg/day, N = 47) or placebo (N = 29), and followed with thrice weekly urine toxicology and once weekly cognitive behavioral psychotherapy.Results: The AA and the AT/TT groups had comparable baseline rates of cocaine positive urines at weeks 1-2 (~ 76%). In the placebo group, an increase of cocaine positive urines in the AT/TT group was found as compared to the AA group (24% vs. 9%). In the doxazosin group, a greater decrease in cocaine positive urines was found in the AT/TT group relative to the AA group. The difference between the doxazosin and placebo groups in cocaine negative urines became evident at weeks 5-6 and peaked at weeks 9-10 (~35% difference). The AT/TT group demonstrated a significant medication and time by medication effect (p < .001), whereas the AA group did not.Conclusion: The T-allele carriers showed a greater reduction of cocaine use after treatment with doxazosin in participants with the ADRA1D gene polymorphism (T1848A), suggesting that this SNP may serve as a pharmacogenetic marker in pharmacotherapy of CUD.
Collapse
Affiliation(s)
- Daryl I Shorter
- Mental Health Care Line, Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas, USA.,Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, Texas, USA
| | - Xuefeng Zhang
- Mental Health Care Line, Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas, USA.,Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, Texas, USA
| | - Coreen B Domingo
- Mental Health Care Line, Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas, USA.,Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, Texas, USA
| | - Ellen M Nielsen
- Mental Health Care Line, Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas, USA.,Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, Texas, USA
| | - Thomas R Kosten
- Mental Health Care Line, Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas, USA.,Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, Texas, USA
| | - David A Nielsen
- Mental Health Care Line, Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas, USA.,Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
28
|
Yao Y, Liu Y, Zeng Z, Zhao Y, Li T, Chen R, Zhang R. Identification of Target Genes of Antiarrhythmic Traditional Chinese Medicine Wenxin Keli. Cardiovasc Ther 2020; 2020:3480276. [PMID: 32565909 PMCID: PMC7284932 DOI: 10.1155/2020/3480276] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 11/16/2019] [Accepted: 01/11/2020] [Indexed: 01/15/2023] Open
Abstract
Wenxin Keli (WXKL) is a traditional Chinese medicine drug approved for the treatment of cardiovascular diseases. This study aimed to identify WXKL-targeting genes involved in antiarrhythmic efficacy of WXKL. The Traditional Chinese Medicine Systems Pharmacology (TCMSP) technology platform was used to screen active compounds of WXKL and WXKL-targeting arrhythmia-related genes. A pig model of myocardial ischemia (MI) was established by balloon-expanding the endothelium of the left coronary artery. Pigs were divided into the model group and WXKL group (n = 6). MI, QT interval, heart rate, and arrhythmia were recorded, and the mRNA expression of target genes in myocardial tissues was detected by PCR. Eleven active ingredients of WXKL and eight WXKL-targeting arrhythmia-related genes were screened. Five pathways were enriched, and an "ingredient-gene-path" network was constructed. WXKL markedly decreased the incidence of arrhythmia in the MI pig model (P < 0.05). The QT interval was significantly shortened, and the heart rate was slowed down in the WXKL group compared with the model group (P < 0.05). In addition, the expression of sodium channel protein type 5 subunit alpha (SCN5A) and beta-2 adrenergic receptor (ADRB2) was downregulated, while muscarinic acetylcholine receptor M2 (CHRM2) was upregulated in the WXKL group (P < 0.05). In conclusion, WXKL may shorten the QT interval and slow down the heart rate by downregulating SCN5A and ADRB2 and upregulating CHRM2 during MI. These findings provide novel insight into molecular mechanisms of WXKL in reducing the incidence of ventricular arrhythmia.
Collapse
MESH Headings
- Action Potentials/drug effects
- Action Potentials/genetics
- Animals
- Anti-Arrhythmia Agents/pharmacology
- Arrhythmias, Cardiac/genetics
- Arrhythmias, Cardiac/metabolism
- Arrhythmias, Cardiac/physiopathology
- Arrhythmias, Cardiac/prevention & control
- Disease Models, Animal
- Drugs, Chinese Herbal/pharmacology
- Gene Expression Regulation
- Gene Regulatory Networks
- Heart Rate/drug effects
- Heart Rate/genetics
- Male
- Medicine, Chinese Traditional
- Myocardial Ischemia/drug therapy
- Myocardial Ischemia/genetics
- Myocardial Ischemia/metabolism
- Myocardial Ischemia/physiopathology
- NAV1.5 Voltage-Gated Sodium Channel/genetics
- NAV1.5 Voltage-Gated Sodium Channel/metabolism
- Protein Interaction Maps
- Receptor, Muscarinic M2/genetics
- Receptor, Muscarinic M2/metabolism
- Receptors, Adrenergic, beta-2/genetics
- Receptors, Adrenergic, beta-2/metabolism
- Swine
- Swine, Miniature
- Time Factors
Collapse
Affiliation(s)
- Yusi Yao
- Department of Cardiovascular Diseases, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong 510080, China
| | - Yuhong Liu
- Department of Cardiovascular Diseases, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong 510080, China
| | - Zhihuan Zeng
- Department of Cardiovascular Diseases, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong 510080, China
| | - Yanqun Zhao
- Department of Cardiovascular Diseases, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong 510080, China
| | - Tudi Li
- Department of Cardiovascular Diseases, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong 510080, China
| | - Rong Chen
- Department of Cardiovascular Diseases, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong 510080, China
| | - Rendan Zhang
- Department of Cardiovascular Diseases, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong 510080, China
| |
Collapse
|
29
|
Akinaga J, García‐Sáinz JA, S. Pupo A. Updates in the function and regulation of α 1 -adrenoceptors. Br J Pharmacol 2019; 176:2343-2357. [PMID: 30740663 PMCID: PMC6592863 DOI: 10.1111/bph.14617] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 12/19/2018] [Accepted: 01/21/2019] [Indexed: 12/16/2022] Open
Abstract
α1 -Adrenoceptors are seven transmembrane domain GPCRs involved in numerous physiological functions controlled by the endogenous catecholamines, noradrenaline and adrenaline, and targeted by drugs useful in therapeutics. Three separate genes, whose products are named α1A -, α1B -, and α1D - adrenoceptors, encode these receptors. Although the existence of multiple α1 -adrenoceptors has been acknowledged for almost 25 years, the specific functions regulated by each subtype are still largely unknown. Despite the limited comprehension, the identification of a single class of subtype-selective ligands for the α1A - adrenoceptors, the so-called α-blockers for prostate dysfunction, has led to major improvement in therapeutics, demonstrating the need for continued efforts in the field. This review article surveys the tissue distribution of the three α1 -adrenoceptor subtypes in the cardiovascular system, genitourinary system, and CNS, highlighting the functions already identified as mediated by the predominant activation of specific subtypes. In addition, this review covers the recent advances in the understanding of the molecular mechanisms involved in the regulation of each of the α1 -adrenoceptor subtypes by phosphorylation and interaction with proteins involved in their desensitization and internalization. LINKED ARTICLES: This article is part of a themed section on Adrenoceptors-New Roles for Old Players. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.14/issuetoc.
Collapse
Affiliation(s)
- Juliana Akinaga
- Department of PharmacologyInstituto de Biociências, UNESPBotucatuBrazil
| | - J. Adolfo García‐Sáinz
- Instituto de Fisiología CelularUniversidad Nacional Autónoma de MéxicoCiudad de MéxicoMexico
| | - André S. Pupo
- Department of PharmacologyInstituto de Biociências, UNESPBotucatuBrazil
| |
Collapse
|
30
|
Williams LM, He X, Vaid TM, Abdul‐Ridha A, Whitehead AR, Gooley PR, Bathgate RA, Williams SJ, Scott DJ. Diazepam is not a direct allosteric modulator of α 1-adrenoceptors, but modulates receptor signaling by inhibiting phosphodiesterase-4. Pharmacol Res Perspect 2019; 7:e00455. [PMID: 30619611 PMCID: PMC6306559 DOI: 10.1002/prp2.455] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 11/30/2018] [Accepted: 12/04/2018] [Indexed: 12/19/2022] Open
Abstract
α1A- and α1B-adrenoceptors (ARs) are G protein-coupled receptors (GPCRs) that are activated by adrenaline and noradrenaline to modulate smooth muscle contraction in the periphery, and neuronal outputs in the central nervous system (CNS). α1A- and α1B-AR are clinically targeted with antagonists for hypertension and benign prostatic hyperplasia and are emerging CNS targets for treating neurodegenerative diseases. The benzodiazepines midazolam, diazepam, and lorazepam are proposed to be positive allosteric modulators (PAMs) of α1-ARs. Here, using thermostabilized, purified, α1A- and α1B-ARs, we sought to identify the benzodiazepine binding site and modulatory mechanism to inform the design of selective PAMs. However, using a combination of biophysical approaches no evidence was found for direct binding of several benzodiazepines to purified, stabilized α1A- and α1B-ARs. Similarly, in cell-based assays expressing unmodified α1A- and α1B-ARs, benzodiazepine treatment had no effect on fluorescent ligand binding, agonist-stimulated Ca2+ release, or G protein activation. In contrast, several benzodiazepines positively modulated phenylephrine stimulation of a cAMP response element pathway by α1A- and α1B-ARs; however, this was shown to be caused by off-target inhibition of phosphodiesterases, known targets of diazepam. This study highlights how purified, stabilized GPCRs are useful for validating allosteric ligand binding and that care needs to be taken before assigning new targets to benzodiazepines.
Collapse
Affiliation(s)
- Lisa M. Williams
- The Florey Institute of Neuroscience and Mental HealthUniversity of MelbourneParkvilleVicAustralia
| | - Xiaoji He
- The Florey Institute of Neuroscience and Mental HealthUniversity of MelbourneParkvilleVicAustralia
- School of Chemistry and Bio21 Molecular Science and Biotechnology InstituteUniversity of MelbourneParkvilleVicAustralia
| | - Tasneem M. Vaid
- The Florey Institute of Neuroscience and Mental HealthUniversity of MelbourneParkvilleVicAustralia
- Department of Biochemistry and Molecular BiologyUniversity of MelbourneParkvilleVicAustralia
- The Bio21 Molecular Science and Biotechnology InstituteUniversity of MelbourneParkvilleVicAustralia
| | - Alaa Abdul‐Ridha
- The Florey Institute of Neuroscience and Mental HealthUniversity of MelbourneParkvilleVicAustralia
| | - Alice R. Whitehead
- The Florey Institute of Neuroscience and Mental HealthUniversity of MelbourneParkvilleVicAustralia
| | - Paul R. Gooley
- Department of Biochemistry and Molecular BiologyUniversity of MelbourneParkvilleVicAustralia
- The Bio21 Molecular Science and Biotechnology InstituteUniversity of MelbourneParkvilleVicAustralia
| | - Ross A.D. Bathgate
- The Florey Institute of Neuroscience and Mental HealthUniversity of MelbourneParkvilleVicAustralia
- Department of Biochemistry and Molecular BiologyUniversity of MelbourneParkvilleVicAustralia
| | - Spencer J. Williams
- School of Chemistry and Bio21 Molecular Science and Biotechnology InstituteUniversity of MelbourneParkvilleVicAustralia
- The Bio21 Molecular Science and Biotechnology InstituteUniversity of MelbourneParkvilleVicAustralia
| | - Daniel J. Scott
- The Florey Institute of Neuroscience and Mental HealthUniversity of MelbourneParkvilleVicAustralia
- Department of Biochemistry and Molecular BiologyUniversity of MelbourneParkvilleVicAustralia
| |
Collapse
|
31
|
Abstract
PURPOSE OF REVIEW Lower urinary tract symptoms (LUTS) result from age-related changes in detrusor function and prostatic growth that are driven by alterations in the ratio of circulating androgens and estrogens. Alpha-adrenergic receptor blockers are commonly used to treat LUTS because they influence urethral tone and intra-urethral pressure. Molecular cloning studies have identified three α1-adrenergic receptor subtypes (α1A, α1B, and α1D). The α1A subtype is predominant in the human prostate but is also present in many parts of the brain that direct cognitive function. Tamsulosin is the most widely used α1-adrenergic receptor antagonist with 12.6 million prescriptions filled in 2010 alone. When compared to the other common types of α1-adrenergic receptor antagonists (i.e., terazosin, doxazosin, and alfuzosin), tamsulosin is 10- to 38-fold more selective for the α1A versus the α1B subtype. RECENT FINDINGS Duan et al. have recently shown that men taking tamsulosin have a higher risk of developing dementia when compared to men taking other α-adrenergic antagonists or no α-adrenergic antagonists at all (HR 1.17; 95% CI 1.14-1.21). Based upon this retrospective analysis, we believe that tamsulosin, because of its unique affinity for α1A-adrenergic receptors, may increase the risk of developing dementia when used for an extended period of time. If these findings are confirmed, they carry significant public health implications for an aging society.
Collapse
Affiliation(s)
- Jason K Frankel
- Department of Surgery, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT, 06030-8073, USA
| | - Yinghui Duan
- Department of Community Medicine and Health Care, University of Connecticut Health Center, Farmington, CT, USA
| | - Peter C Albertsen
- Department of Surgery, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT, 06030-8073, USA.
| |
Collapse
|
32
|
Ghasemi M, Mehranfard N. Mechanisms underlying anticonvulsant and proconvulsant actions of norepinephrine. Neuropharmacology 2018; 137:297-308. [DOI: 10.1016/j.neuropharm.2018.05.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 05/09/2018] [Accepted: 05/10/2018] [Indexed: 01/02/2023]
|
33
|
Yong KJ, Vaid TM, Shilling PJ, Wu FJ, Williams LM, Deluigi M, Plückthun A, Bathgate RAD, Gooley PR, Scott DJ. Determinants of Ligand Subtype-Selectivity at α 1A-Adrenoceptor Revealed Using Saturation Transfer Difference (STD) NMR. ACS Chem Biol 2018. [PMID: 29537256 DOI: 10.1021/acschembio.8b00191] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
α1A- and α1B-adrenoceptors (α1A-AR and α1B-AR) are closely related G protein-coupled receptors (GPCRs) that modulate the cardiovascular and nervous systems in response to binding epinephrine and norepinephrine. The GPCR gene superfamily is made up of numerous subfamilies that, like α1A-AR and α1B-AR, are activated by the same endogenous agonists but may modulate different physiological processes. A major challenge in GPCR research and drug discovery is determining how compounds interact with receptors at the molecular level, especially to assist in the optimization of drug leads. Nuclear magnetic resonance spectroscopy (NMR) can provide great insight into ligand-binding epitopes, modes, and kinetics. Ideally, ligand-based NMR methods require purified, well-behaved protein samples. The instability of GPCRs upon purification in detergents, however, makes the application of NMR to study ligand binding challenging. Here, stabilized α1A-AR and α1B-AR variants were engineered using Cellular High-throughput Encapsulation, Solubilization, and Screening (CHESS), allowing the analysis of ligand binding with Saturation Transfer Difference NMR (STD NMR). STD NMR was used to map the binding epitopes of epinephrine and A-61603 to both receptors, revealing the molecular determinants for the selectivity of A-61603 for α1A-AR over α1B-AR. The use of stabilized GPCRs for ligand-observed NMR experiments will lead to a deeper understanding of binding processes and assist structure-based drug design.
Collapse
Affiliation(s)
- Kelvin J. Yong
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, 30 Royal Parade, Parkville, Victoria 3052, Australia
- The Department of Biochemistry and Molecular Biology, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Tasneem M. Vaid
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, 30 Royal Parade, Parkville, Victoria 3052, Australia
- The Department of Biochemistry and Molecular Biology, The University of Melbourne, Parkville, Victoria 3010, Australia
- The Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Patrick J. Shilling
- The Department of Biochemistry and Molecular Biology, The University of Melbourne, Parkville, Victoria 3010, Australia
- The Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Feng-Jie Wu
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, 30 Royal Parade, Parkville, Victoria 3052, Australia
- The Department of Biochemistry and Molecular Biology, The University of Melbourne, Parkville, Victoria 3010, Australia
- The Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Lisa M. Williams
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, 30 Royal Parade, Parkville, Victoria 3052, Australia
| | - Mattia Deluigi
- Department of Biochemistry, University of Zurich, 8057 Zurich, Switzerland
| | - Andreas Plückthun
- Department of Biochemistry, University of Zurich, 8057 Zurich, Switzerland
| | - Ross A. D. Bathgate
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, 30 Royal Parade, Parkville, Victoria 3052, Australia
- The Department of Biochemistry and Molecular Biology, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Paul R. Gooley
- The Department of Biochemistry and Molecular Biology, The University of Melbourne, Parkville, Victoria 3010, Australia
- The Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Daniel J. Scott
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, 30 Royal Parade, Parkville, Victoria 3052, Australia
- The Department of Biochemistry and Molecular Biology, The University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
34
|
Duan Y, Grady JJ, Albertsen PC, Helen Wu Z. Tamsulosin and the risk of dementia in older men with benign prostatic hyperplasia. Pharmacoepidemiol Drug Saf 2018; 27:340-348. [DOI: 10.1002/pds.4361] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 10/23/2017] [Accepted: 10/25/2017] [Indexed: 12/22/2022]
Affiliation(s)
- Yinghui Duan
- Department of Community Medicine and Health Care; University of Connecticut Health Center; Farmington CT USA
- Connecticut Institute for Clinical and Translational Science (CICATS), Farmington; CT USA
| | - James J. Grady
- Department of Community Medicine and Health Care; University of Connecticut Health Center; Farmington CT USA
- Connecticut Institute for Clinical and Translational Science (CICATS), Farmington; CT USA
| | - Peter C. Albertsen
- Department of Surgery (Urology); University of Connecticut Health Center; Farmington CT USA
| | - Z. Helen Wu
- Connecticut Institute for Clinical and Translational Science (CICATS), Farmington; CT USA
- Department of Psychiatry; University of Connecticut Health Center; Farmington CT USA
| |
Collapse
|
35
|
Maletic V, Eramo A, Gwin K, Offord SJ, Duffy RA. The Role of Norepinephrine and Its α-Adrenergic Receptors in the Pathophysiology and Treatment of Major Depressive Disorder and Schizophrenia: A Systematic Review. Front Psychiatry 2017; 8:42. [PMID: 28367128 PMCID: PMC5355451 DOI: 10.3389/fpsyt.2017.00042] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 03/01/2017] [Indexed: 12/11/2022] Open
Abstract
Norepinephrine (NE) is recognized as having a key role in the pathophysiology of major depressive disorder (MDD) and schizophrenia, although its distinct actions via α-adrenergic receptors (α-ARs) are not well defined. We performed a systematic review examining the roles of NE and α-ARs in MDD and schizophrenia. PubMed and ProQuest database searches were performed to identify English language papers published between 2008 and 2015. In total, 2,427 publications (PubMed, n = 669; ProQuest, n = 1,758) were identified. Duplicates, articles deemed not relevant, case studies, reviews, meta-analyses, preclinical reports, or articles on non-target indications were excluded. To limit the review to the most recent data representative of the literature, the review further focused on publications from 2010 to 2015, which were screened independently by all authors. A total of 16 research reports were identified: six clinical trial reports, six genetic studies, two biomarker studies, and two receptor studies. Overall, the studies provided indirect evidence that α-AR activity may play an important role in aberrant regulation of cognition, arousal, and valence systems associated with MDD and schizophrenia. Characterization of the NE pathway in patients may provide clinicians with information for more personalized therapy of these heterogeneous diseases. Current clinical studies do not provide direct evidence to support the role of NE α-ARs in the pathophysiology of MDD and schizophrenia and in the treatment response of patients with these diseases, in particular with relation to specific valence systems. Clinical studies that attempt to define associations between specific receptor binding profiles of psychotropics and particular clinical outcomes are needed.
Collapse
Affiliation(s)
- Vladimir Maletic
- Department of Neuropsychiatry and Behavioral Science, University of South Carolina , Columbia, SC , USA
| | - Anna Eramo
- Medical Affairs - Psychiatry, Lundbeck LLC , Deerfield, IL , USA
| | - Keva Gwin
- Medical Affairs - Psychiatry, Lundbeck LLC , Deerfield, IL , USA
| | - Steve J Offord
- Medical Affairs, Otsuka Pharmaceutical Development and Commercialization, Inc. , Princeton, NJ , USA
| | - Ruth A Duffy
- Medical Affairs, Otsuka Pharmaceutical Development and Commercialization, Inc. , Princeton, NJ , USA
| |
Collapse
|
36
|
Campbell AP, Wakelin LPG, Denny WA, Finch AM. Homobivalent Conjugation Increases the Allosteric Effect of 9-aminoacridine at the α1-Adrenergic Receptors. Mol Pharmacol 2016; 91:135-144. [PMID: 27903755 DOI: 10.1124/mol.116.105874] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Accepted: 11/28/2016] [Indexed: 12/14/2022] Open
Abstract
The α1-adrenergic receptors are targets for a number of cardiovascular and central nervous system conditions, but the current drugs for these receptors lack specificity to be of optimal clinical value. Allosteric modulators offer an alternative mechanism of action to traditional α1-adrenergic ligands, yet there is little information describing this drug class at the α1-adrenergic receptors. We have identified a series of 9-aminoacridine compounds that demonstrate allosteric modulation of the α1A- and α1B-adrenergic receptors. The 9-aminoacridines increase the rate of [3H]prazosin dissociation from the α1A- and α1B-adrenergic receptors and noncompetitively inhibit receptor activation by the endogenous agonist norepinephrine. The structurally similar compound, tacrine, which is a known allosteric modulator of the muscarinic receptors, is also shown to be a modulator of the α1-adrenergic receptors, which suggests a general lack of selectivity for allosteric binding sites across aminergic G protein-coupled receptor. Conjugation of two 9-aminoacridine pharmacophores, using linkers of varying length, increases the potency and efficacy of the allosteric effects of this ligand, likely through optimization of bitopic engagement of the allosteric and orthosteric binding sites of the receptor. Such a bivalent approach may provide a mechanism for fine tuning the efficacy of allosteric compounds in future drug design efforts.
Collapse
Affiliation(s)
- Adrian P Campbell
- Department of Pharmacology, School of Medical Sciences, UNSW Australia, Kensington, Australia (A.P.C., L.P.G.W., A.M.F.); Auckland Cancer Society Research Centre, Faculty of Medicine and Health Science, University of Auckland, Auckland, New Zealand (W.A.D.)
| | - Laurence P G Wakelin
- Department of Pharmacology, School of Medical Sciences, UNSW Australia, Kensington, Australia (A.P.C., L.P.G.W., A.M.F.); Auckland Cancer Society Research Centre, Faculty of Medicine and Health Science, University of Auckland, Auckland, New Zealand (W.A.D.)
| | - William A Denny
- Department of Pharmacology, School of Medical Sciences, UNSW Australia, Kensington, Australia (A.P.C., L.P.G.W., A.M.F.); Auckland Cancer Society Research Centre, Faculty of Medicine and Health Science, University of Auckland, Auckland, New Zealand (W.A.D.)
| | - Angela M Finch
- Department of Pharmacology, School of Medical Sciences, UNSW Australia, Kensington, Australia (A.P.C., L.P.G.W., A.M.F.); Auckland Cancer Society Research Centre, Faculty of Medicine and Health Science, University of Auckland, Auckland, New Zealand (W.A.D.)
| |
Collapse
|
37
|
Paparone S, Severini C, Ciotti MT, D'Agata V, Calissano P, Cavallaro S. Transcriptional landscapes at the intersection of neuronal apoptosis and substance P-induced survival: exploring pathways and drug targets. Cell Death Discov 2016; 2:16050. [PMID: 27551538 PMCID: PMC4979452 DOI: 10.1038/cddiscovery.2016.50] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 05/20/2016] [Accepted: 05/25/2016] [Indexed: 12/29/2022] Open
Abstract
A change in the delicate equilibrium between apoptosis and survival regulates the neurons fate during the development of nervous system and its homeostasis in adulthood. Signaling pathways promoting or protecting from apoptosis are activated by multiple signals, including those elicited by neurotrophic factors, and depend upon specific transcriptional programs. To decipher the rescue program induced by substance P (SP) in cerebellar granule neurons, we analyzed their whole-genome expression profiles after induction of apoptosis and treatment with SP. Transcriptional pathways associated with the survival effect of SP included genes encoding for proteins that may act as pharmacological targets. Inhibition of one of these, the Myc pro-oncogene by treatment with 10058-F4, reverted in a dose-dependent manner the rescue effect of SP. In addition to elucidate the transcriptional mechanisms at the intersection of neuronal apoptosis and survival, our systems biology-based perspective paves the way towards an innovative pharmacology based on targets downstream of neurotrophic factor receptors.
Collapse
Affiliation(s)
- S Paparone
- Institute of Neurological Sciences, Italian National Research Council , Via Paolo Gaifami, 18, Catania 95125, Italy
| | - C Severini
- Institute of Cell Biology and Neurobiology, Italian National Research Council, Via del Fosso di Fiorano 64, Roma 00143, Italy; European Brain Research Institute, Via del Fosso di Fiorano 64, Roma 00143, Italy
| | - M T Ciotti
- Institute of Cell Biology and Neurobiology, Italian National Research Council , Via del Fosso di Fiorano 64, Roma 00143, Italy
| | - V D'Agata
- Department of Biomedical and Biotechnological Sciences, Section of Human Anatomy and Histology, University of Catania , Catania 95125, Italy
| | - P Calissano
- European Brain Research Institute , Via del Fosso di Fiorano 64, Roma 00143, Italy
| | - S Cavallaro
- Institute of Neurological Sciences, Italian National Research Council , Via Paolo Gaifami, 18, Catania 95125, Italy
| |
Collapse
|
38
|
Shi T, Papay RS, Perez DM. The role of α 1-adrenergic receptors in regulating metabolism: increased glucose tolerance, leptin secretion and lipid oxidation. J Recept Signal Transduct Res 2016; 37:124-132. [PMID: 27277698 DOI: 10.1080/10799893.2016.1193522] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The role of α1-adrenergic receptors (α1-ARs) and their subtypes in metabolism is not well known. Most previous studies were performed before the advent of transgenic mouse models and utilized transformed cell lines and poorly selective antagonists. We have now studied the metabolic regulation of the α1A- and α1B-AR subtypes in vivo using knock-out (KO) and transgenic mice that express a constitutively active mutant (CAM) form of the receptor, assessing subtype-selective functions. CAM mice increased glucose tolerance while KO mice display impaired glucose tolerance. CAM mice increased while KO decreased glucose uptake into white fat tissue and skeletal muscle with the CAM α1A-AR showing selective glucose uptake into the heart. Using indirect calorimetry, both CAM mice demonstrated increased whole body fatty acid oxidation, while KO mice preferentially oxidized carbohydrate. CAM α1A-AR mice displayed significantly decreased fasting plasma triglycerides and glucose levels while α1A-AR KO displayed increased levels of triglycerides and glucose. Both CAM mice displayed increased plasma levels of leptin while KO mice decreased leptin levels. Most metabolic effects were more efficacious with the α1A-AR subtype. Our results suggest that stimulation of α1-ARs results in a favorable metabolic profile of increased glucose tolerance, cardiac glucose uptake, leptin secretion and increased whole body lipid metabolism that may contribute to its previously recognized cardioprotective and neuroprotective benefits.
Collapse
Affiliation(s)
- Ting Shi
- a Department of Molecular Cardiology , Lerner Research Institute, Cleveland Clinic Foundation , Cleveland , OH , USA
| | - Robert S Papay
- a Department of Molecular Cardiology , Lerner Research Institute, Cleveland Clinic Foundation , Cleveland , OH , USA
| | - Dianne M Perez
- a Department of Molecular Cardiology , Lerner Research Institute, Cleveland Clinic Foundation , Cleveland , OH , USA
| |
Collapse
|
39
|
Shi T, Papay RS, Perez DM. α1A-Adrenergic receptor prevents cardiac ischemic damage through PKCδ/GLUT1/4-mediated glucose uptake. J Recept Signal Transduct Res 2015; 36:261-70. [PMID: 26832303 DOI: 10.3109/10799893.2015.1091475] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
While α(1)-adrenergic receptors (ARs) have been previously shown to limit ischemic cardiac damage, the mechanisms remain unclear. Most previous studies utilized low oxygen conditions in addition to ischemic buffers with glucose deficiencies, but we discovered profound differences if the two conditions are separated. We assessed both mouse neonatal and adult myocytes and HL-1 cells in a series of assays assessing ischemic damage under hypoxic or low glucose conditions. We found that α(1)-AR stimulation protected against increased lactate dehydrogenase release or Annexin V(+) apoptosis under conditions that were due to low glucose concentration not to hypoxia. The α(1)-AR antagonist prazosin or nonselective protein kinase C (PKC) inhibitors blocked the protective effect. α(1)-AR stimulation increased (3)H-deoxyglucose uptake that was blocked with either an inhibitor to glucose transporter 1 or 4 (GLUT1 or GLUT4) or small interfering RNA (siRNA) against PKCδ. GLUT1/4 inhibition also blocked α(1)-AR-mediated protection from apoptosis. The PKC inhibitor rottlerin or siRNA against PKCδ blocked α(1)-AR stimulated GLUT1 or GLUT4 plasma membrane translocation. α(1)-AR stimulation increased plasma membrane concentration of either GLUT1 or GLUT4 in a time-dependent fashion. Transgenic mice overexpressing the α(1A)-AR but not α(1B)-AR mice displayed increased glucose uptake and increased GLUT1 and GLUT4 plasma membrane translocation in the adult heart while α(1A)-AR but not α(1B)-AR knockout mice displayed lowered glucose uptake and GLUT translocation. Our results suggest that α(1)-AR activation is anti-apoptotic and protective during cardiac ischemia due to glucose deprivation and not hypoxia by enhancing glucose uptake into the heart via PKCδ-mediated GLUT translocation that may be specific to the α(1A)-AR subtype.
Collapse
Affiliation(s)
- Ting Shi
- a Department of Molecular Cardiology , Lerner Research Institute, Cleveland Clinic Foundation , Cleveland , OH , USA
| | - Robert S Papay
- a Department of Molecular Cardiology , Lerner Research Institute, Cleveland Clinic Foundation , Cleveland , OH , USA
| | - Dianne M Perez
- a Department of Molecular Cardiology , Lerner Research Institute, Cleveland Clinic Foundation , Cleveland , OH , USA
| |
Collapse
|
40
|
Copik AJ, Baldys A, Nguyen K, Sahdeo S, Ho H, Kosaka A, Dietrich PJ, Fitch B, Raymond JR, Ford APDW, Button D, Milla ME. Isoproterenol acts as a biased agonist of the alpha-1A-adrenoceptor that selectively activates the MAPK/ERK pathway. PLoS One 2015; 10:e0115701. [PMID: 25606852 PMCID: PMC4301629 DOI: 10.1371/journal.pone.0115701] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2014] [Accepted: 11/26/2014] [Indexed: 11/24/2022] Open
Abstract
The α1A-AR is thought to couple predominantly to the Gαq/PLC pathway and lead to phosphoinositide hydrolysis and calcium mobilization, although certain agonists acting at this receptor have been reported to trigger activation of arachidonic acid formation and MAPK pathways. For several G protein-coupled receptors (GPCRs) agonists can manifest a bias for activation of particular effector signaling output, i.e. not all agonists of a given GPCR generate responses through utilization of the same signaling cascade(s). Previous work with Gαq coupling-defective variants of α1A-AR, as well as a combination of Ca2+ channel blockers, uncovered cross-talk between α1A-AR and β2-AR that leads to potentiation of a Gαq-independent signaling cascade in response to α1A-AR activation. We hypothesized that molecules exist that act as biased agonists to selectively activate this pathway. In this report, isoproterenol (Iso), typically viewed as β-AR-selective agonist, was examined with respect to activation of α1A-AR. α1A-AR selective antagonists were used to specifically block Iso evoked signaling in different cellular backgrounds and confirm its action at α1A-AR. Iso induced signaling at α1A-AR was further interrogated by probing steps along the Gαq /PLC, Gαs and MAPK/ERK pathways. In HEK-293/EBNA cells transiently transduced with α1A-AR, and CHO_α1A-AR stable cells, Iso evoked low potency ERK activity as well as Ca2+ mobilization that could be blocked by α1A-AR selective antagonists. The kinetics of Iso induced Ca2+ transients differed from typical Gαq- mediated Ca2+ mobilization, lacking both the fast IP3R mediated response and the sustained phase of Ca2+ re-entry. Moreover, no inositol phosphate (IP) accumulation could be detected in either cell line after stimulation with Iso, but activation was accompanied by receptor internalization. Data are presented that indicate that Iso represents a novel type of α1A-AR partial agonist with signaling bias toward MAPK/ERK signaling cascade that is likely independent of coupling to Gαq.
Collapse
Affiliation(s)
- Alicja. J. Copik
- Biochemical Pharmacology, Inflammation Discovery, Roche Palo Alto LLC, 3401 Hillview Drive, Palo Alto, CA 94304, United States of America
| | - Aleksander Baldys
- Nephrology Division, Department of Medicine, Medical University of South Carolina, and Medical and Research Services, Ralph H Johnson Veterans Affairs Medical Center, Charleston, South Carolina 29425, United States of America
| | - Khanh Nguyen
- Discovery Technologies, Roche Palo Alto LLC, 3401 Hillview Drive, Palo Alto, CA 94304, United States of America
| | - Sunil Sahdeo
- Biochemical Pharmacology, Inflammation Discovery, Roche Palo Alto LLC, 3401 Hillview Drive, Palo Alto, CA 94304, United States of America
| | - Hoangdung Ho
- Discovery Technologies, Roche Palo Alto LLC, 3401 Hillview Drive, Palo Alto, CA 94304, United States of America
| | - Alan Kosaka
- Discovery Technologies, Roche Palo Alto LLC, 3401 Hillview Drive, Palo Alto, CA 94304, United States of America
| | - Paul J. Dietrich
- Discovery Technologies, Roche Palo Alto LLC, 3401 Hillview Drive, Palo Alto, CA 94304, United States of America
| | - Bill Fitch
- Discovery Technologies, Roche Palo Alto LLC, 3401 Hillview Drive, Palo Alto, CA 94304, United States of America
| | - John R. Raymond
- Nephrology Division, Department of Medicine, Medical University of South Carolina, and Medical and Research Services, Ralph H Johnson Veterans Affairs Medical Center, Charleston, South Carolina 29425, United States of America
| | - Anthony P. D. W. Ford
- Biochemical Pharmacology, Inflammation Discovery, Roche Palo Alto LLC, 3401 Hillview Drive, Palo Alto, CA 94304, United States of America
| | - Donald Button
- Biochemical Pharmacology, Inflammation Discovery, Roche Palo Alto LLC, 3401 Hillview Drive, Palo Alto, CA 94304, United States of America
| | - Marcos E. Milla
- Biochemical Pharmacology, Inflammation Discovery, Roche Palo Alto LLC, 3401 Hillview Drive, Palo Alto, CA 94304, United States of America
- * E-mail:
| |
Collapse
|
41
|
Merriman JD, Aouizerat BE, Cataldo JK, Dunn LB, Kober K, Langford DJ, West C, Cooper BA, Paul SM, Miaskowski C. Associations between catecholaminergic, GABAergic, and serotonergic genes and self-reported attentional function in oncology patients and their family caregivers. Eur J Oncol Nurs 2014; 19:251-9. [PMID: 25524657 DOI: 10.1016/j.ejon.2014.11.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2014] [Accepted: 11/19/2014] [Indexed: 11/28/2022]
Abstract
PURPOSE OF THE RESEARCH Evaluate for associations between variations in genes involved in catecholaminergic, gamma-aminobutyric acid (GABA)-ergic, and serotonergic mechanisms of neurotransmission and attentional function latent classes. PATIENTS AND METHODS This descriptive, longitudinal study was conducted at two radiation therapy departments. The sample included three latent classes of individuals with distinct trajectories of self-reported attentional function during radiation therapy, who were previously identified using growth mixture modeling among 167 oncology patients and 85 of their family caregivers. Multivariable models were used to evaluate for genotypic associations of neurotransmission genes with attentional function latent class membership, after controlling for covariates. RESULTS Variations in catecholaminergic (i.e., ADRA1D rs4815675, SLC6A3 rs37022), GABAergic (i.e., SLC6A1 rs2697138), and serotonergic (i.e., HTR2A rs2296972, rs9534496) neurotransmission genes were significant predictors of latent class membership in multivariable models. CONCLUSIONS Findings suggest that variations in genes that encode for three distinct but related neurotransmission systems are involved in alterations in attentional function. Knowledge of both phenotypic and genetic markers associated with alterations in attentional function can be used by clinicians to identify patients and family caregivers who are at higher risk for this symptom. Increased understanding of the genetic markers associated with alterations in attentional function may provide insights into the underlying mechanisms for this significant clinical problem.
Collapse
Affiliation(s)
- John D Merriman
- School of Nursing, University of California, San Francisco, 2 Koret Way, Box 0610, San Francisco, CA 94143-0610, USA.
| | - Bradley E Aouizerat
- School of Nursing, University of California, San Francisco, 2 Koret Way, Box 0610, San Francisco, CA 94143-0610, USA; Institute for Human Genetics, University of California, San Francisco, 513 Parnassus Avenue, Box 0794, San Francisco, CA 94143-0794, USA.
| | - Janine K Cataldo
- School of Nursing, University of California, San Francisco, 2 Koret Way, Box 0610, San Francisco, CA 94143-0610, USA.
| | - Laura B Dunn
- School of Medicine, University of California, San Francisco, 513 Parnassus Avenue, Box 0410, San Francisco, CA 94143-0410, USA.
| | - Kord Kober
- School of Nursing, University of California, San Francisco, 2 Koret Way, Box 0610, San Francisco, CA 94143-0610, USA.
| | - Dale J Langford
- School of Nursing, University of California, San Francisco, 2 Koret Way, Box 0610, San Francisco, CA 94143-0610, USA.
| | - Claudia West
- School of Nursing, University of California, San Francisco, 2 Koret Way, Box 0610, San Francisco, CA 94143-0610, USA.
| | - Bruce A Cooper
- School of Nursing, University of California, San Francisco, 2 Koret Way, Box 0610, San Francisco, CA 94143-0610, USA.
| | - Steven M Paul
- School of Nursing, University of California, San Francisco, 2 Koret Way, Box 0610, San Francisco, CA 94143-0610, USA.
| | - Christine Miaskowski
- School of Nursing, University of California, San Francisco, 2 Koret Way, Box 0610, San Francisco, CA 94143-0610, USA.
| |
Collapse
|
42
|
Drummond PD. Neuronal changes resulting in up-regulation of alpha-1 adrenoceptors after peripheral nerve injury. Neural Regen Res 2014; 9:1337-40. [PMID: 25221588 PMCID: PMC4160862 DOI: 10.4103/1673-5374.137583] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/14/2014] [Indexed: 12/13/2022] Open
Abstract
Under normal conditions, the sympathetic neurotransmitter noradrenaline inhibits the production and release of pro-inflammatory cytokines. However, after peripheral nerve and tissue injury, pro-inflammatory cytokines appear to induce the expression of the alpha1A-adrenoceptor subtype on immune cells and perhaps also on other cells in the injured tissue. In turn, noradrenaline may act on up-regulated alpha1-adrenoceptors to increase the production of the pro-inflammatory cytokine interleukin-6. In addition, the release of inflammatory mediators and nerve growth factor from keratinocytes and other cells may augment the expression of alpha1-adrenoceptors on peripheral nerve fibers. Consequently, nociceptive afferents acquire an abnormal excitability to adrenergic agents, and inflammatory processes build. These mechanisms could contribute to the development of sympathetically maintained pain in conditions such as post-herpetic neuralgia, cutaneous neuromas, amputation stump pain and complex regional pain syndrome.
Collapse
Affiliation(s)
- Peter D Drummond
- Centre for Research on Chronic Pain and Inflammatory Diseases, Murdoch University, Perth, Western Australia, Australia
| |
Collapse
|
43
|
Collette KM, Zhou XD, Amoth HM, Lyons MJ, Papay RS, Sens DA, Perez DM, Doze VA. Long-term α1B-adrenergic receptor activation shortens lifespan, while α1A-adrenergic receptor stimulation prolongs lifespan in association with decreased cancer incidence. AGE (DORDRECHT, NETHERLANDS) 2014; 36:9675. [PMID: 24994537 PMCID: PMC4150908 DOI: 10.1007/s11357-014-9675-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Accepted: 06/18/2014] [Indexed: 06/03/2023]
Abstract
The α1-adrenergic receptor (α1AR) subtypes, α1AAR and α1BAR, have differential effects in the heart and central nervous system. Long-term stimulation of the α1AAR subtype prolongs lifespan and provides cardio- and neuro-protective effects. We examined the lifespan of constitutively active mutant (CAM)-α1BAR mice and the incidence of cancer in mice expressing the CAM form of either the α1AAR (CAM-α1AAR mice) or α1BAR. CAM-α1BAR mice have a significantly shortened lifespan when compared with wild-type (WT) animals; however, the effect was sex dependent. Female CAM-α1BAR mice lived significantly shorter lives, while the median lifespan of male CAM-α1BAR mice was not different when compared with that of WT animals. There was no difference in the incidence of cancer in either sex of CAM-α1BAR mice. The incidence of cancer was significantly decreased in CAM-α1AAR mice when compared with that in WT, and no sex-dependent effects were observed. Further study is warranted on cancer incidence after activation of each α1AR subtype and the effect of sex on lifespan following activation of the α1BAR. The implications of a decrease in cancer incidence following long-term α1AAR stimulation could lead to improved treatments for cancer.
Collapse
Affiliation(s)
- Katie M. Collette
- />Department of Basic Sciences, School of Medicine and Health Sciences, University of North Dakota, 501 N. Columbia Rd., Grand Forks, ND 58202 USA
| | - Xu Dong Zhou
- />Department of Pathology, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202 USA
| | - Haley M. Amoth
- />Department of Basic Sciences, School of Medicine and Health Sciences, University of North Dakota, 501 N. Columbia Rd., Grand Forks, ND 58202 USA
| | - Mariaha J. Lyons
- />Department of Basic Sciences, School of Medicine and Health Sciences, University of North Dakota, 501 N. Columbia Rd., Grand Forks, ND 58202 USA
| | - Robert S. Papay
- />Department of Molecular Cardiology, The Lerner Research Institute, The Cleveland Clinic Foundation, Cleveland, OH 44195 USA
| | - Donald A. Sens
- />Department of Pathology, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202 USA
| | - Dianne M. Perez
- />Department of Molecular Cardiology, The Lerner Research Institute, The Cleveland Clinic Foundation, Cleveland, OH 44195 USA
| | - Van A. Doze
- />Department of Basic Sciences, School of Medicine and Health Sciences, University of North Dakota, 501 N. Columbia Rd., Grand Forks, ND 58202 USA
| |
Collapse
|
44
|
Alpha1a-adrenoceptor genetic variant induces cardiomyoblast-to-fibroblast-like cell transition via distinct signaling pathways. Cell Signal 2014; 26:1985-97. [PMID: 24835978 DOI: 10.1016/j.cellsig.2014.05.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Revised: 05/08/2014] [Accepted: 05/09/2014] [Indexed: 12/21/2022]
Abstract
The role of naturally occurring human α1a-Adrenergic Receptor (α1aAR) genetic variants associated with cardiovascular disorders is poorly understood. Here, we present the novel findings that expression of human α1aAR-247R (247R) genetic variant in cardiomyoblasts leads to transition of cardiomyoblasts into a fibroblast-like phenotype, evidenced by morphology and distinct de novo expression of characteristic genes. These fibroblast-like cells exhibit constitutive, high proliferative capacity and agonist-induced hypertrophy compared with cells prior to transition. We demonstrate that constitutive, synergistic activation of EGFR, Src and ERK kinases is the potential molecular mechanism of this transition. We also demonstrate that 247R triggers two distinct EGFR transactivation-dependent signaling pathways: 1) constitutive Gq-independent β-arrestin-1/Src/MMP/EGFR/ERK-dependent hyperproliferation and 2) agonist-induced Gq- and EGFR/STAT-dependent hypertrophy. Interestingly, in cardiomyoblasts agonist-independent hyperproliferation is MMP-dependent, but in fibroblast-like cells it is MMP-independent, suggesting that expression of α1aAR genetic variant in cardiomyocytes may trigger extracellular matrix remodeling. Thus, these novel findings demonstrate that EGFR transactivation by α1aAR-247R leads to hyperproliferation, hypertrophy and alterations in cardiomyoblasts, suggesting that these unique genetically-mediated alterations in signaling pathways and cellular function may lead to myocardial fibrosis. Such extracellular matrix remodeling may contribute to the genesis of arrhythmias in certain types of heart failure.
Collapse
|
45
|
Sakata K, Duke SM. Lack of BDNF expression through promoter IV disturbs expression of monoamine genes in the frontal cortex and hippocampus. Neuroscience 2013; 260:265-75. [PMID: 24345476 DOI: 10.1016/j.neuroscience.2013.12.013] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 11/29/2013] [Accepted: 12/06/2013] [Indexed: 02/07/2023]
Abstract
Brain-derived neurotrophic factor (BDNF) is implicated in the pathophysiology of psychiatric conditions including major depression and schizophrenia. Mice lacking activity-driven BDNF expression through promoter IV (knock-in promoter IV: KIV) exhibit depression-like behavior, inflexible learning, and impaired response inhibition. Monoamine systems (serotonin, dopamine, and noradrenaline) are suggested to be involved in depression and schizophrenia since many of the current antidepressants and antipsychotics increase the brain levels of monoamines and/or act on monoamine receptors. To elucidate the impact of activity-driven BDNF on the monoamine systems, we examined mRNA levels for 30 monoamine-related genes, including receptors, transporters, and synthesizing enzymes, in KIV and control wild-type mice by using quantitative reverse-transcription polymerase chain reaction (qRT-PCR). mRNA levels were measured in the frontal cortex and hippocampus, which are regions related to depression and schizophrenia and where promoter IV is active. The frontal cortex of KIV mice showed reduced levels of mRNA expression for serotonin receptors 1b, 2a, and 5b (5HTR1b, 5HTR2a, 5HTR5b), dopamine D2 receptors (DRD2), and adrenergic receptors alpha 1a and 1d (AdRα1a and AdRα1b), but increased levels for serotonin synthesizing enzyme, tryptophan hydroxylase (TPH), and dopamine D4 receptor (DRD4) when compared to control wild-type mice. The hippocampus of KIV mice showed decreased levels of 5HTR5b. Our results provide causal evidence that lack of promoter IV-driven BDNF disturbs expression of monoaminergic genes in the frontal cortex and hippocampus. These disturbed expression changes in the monoamine systems may mediate the depression- and schizophrenia-like behavior of KIV mice. Our results also suggest that antidepressant and antipsychotic treatments may actually interfere with and normalize the disturbed monoamine systems caused by reduced activity-dependent BDNF, while the treatment responses to these drugs may differ in the subject with reduced BDNF levels caused by stress and lack of neuronal activity.
Collapse
Affiliation(s)
- K Sakata
- Department of Pharmacology, University of Tennessee Health Science Center, Memphis, TN, USA.
| | - S M Duke
- Department of Pharmacology, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
46
|
Heng BC, Aubel D, Fussenegger M. An overview of the diverse roles of G-protein coupled receptors (GPCRs) in the pathophysiology of various human diseases. Biotechnol Adv 2013; 31:1676-94. [DOI: 10.1016/j.biotechadv.2013.08.017] [Citation(s) in RCA: 125] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Revised: 08/19/2013] [Accepted: 08/19/2013] [Indexed: 12/23/2022]
|
47
|
Papay RS, Shi T, Piascik MT, Naga Prasad SV, Perez DM. α₁A-adrenergic receptors regulate cardiac hypertrophy in vivo through interleukin-6 secretion. Mol Pharmacol 2013; 83:939-48. [PMID: 23404509 DOI: 10.1124/mol.112.084483] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
The role of α₁-adrenergic receptors (ARs) in the regulation of cardiac hypertrophy is still unclear, because transgenic mice demonstrated hypertrophy or the lack of it despite high receptor overexpression. To further address the role of the α₁-ARs in cardiac hypertrophy, we analyzed unique transgenic mice that overexpress constitutively active mutation (CAM) α₁A-ARs or CAM α₁B-ARs under the regulation of large fragments of their native promoters. These constitutively active receptors are expressed in all tissues that endogenously express their wild-type counterparts as opposed to only myocyte-targeted transgenic mice. In this study, we discovered that CAM α₁A-AR mice in vivo have cardiac hypertrophy independent of changes in blood pressure, corroborating earlier studies, but in contrast to myocyte-targeted α₁A-AR mice. We also found cardiac hypertrophy in CAM α₁B-AR mice, in agreement with previous studies, but hypertrophy only developed in older mice. We also discovered unique α₁-AR-mediated hypertrophic signaling that was AR subtype-specific with CAM α₁A-AR mice secreting atrial naturietic factor and interleukin-6 (IL-6), whereas CAM α₁B-AR mice expressed activated nuclear factor-κB (NF-κB). These particular hypertrophic signals were blocked when the other AR subtype was coactivated. We also discovered that crossbreeding the two CAM models (double CAM α₁A/B-AR) inhibited the development of hypertrophy and was reversible with single receptor activation, suggesting that coactivation of the receptors can lead to novel antagonistic signal transduction. This was confirmed by demonstrating antagonistic signals that were even lower than normal controls in the double CAM α₁A/B-AR mice for p38, NF-κB, and the IL-6/glycoprotein 130/signal transducer and activator of transcription 3 pathway. Because α₁A/B double knockout mice fail to develop hypertrophy in response to IL-6, our results suggest that IL-6 is a major mediator of α₁A-AR cardiac hypertrophy.
Collapse
Affiliation(s)
- Robert S Papay
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio 44195, USA
| | | | | | | | | |
Collapse
|
48
|
Shi T, Moravec CS, Perez DM. Novel proteins associated with human dilated cardiomyopathy: selective reduction in α(1A)-adrenergic receptors and increased desensitization proteins. J Recept Signal Transduct Res 2013; 33:96-106. [PMID: 23384050 DOI: 10.3109/10799893.2013.764897] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Abstract Therapeutics to treat human heart failure (HF) and the identification of proteins associated with HF are still limited. We analyzed α(1)-adrenergic receptor (AR) subtypes in human HF and performed proteomic analysis on more uniform samples to identify novel proteins associated with human HF. Six failing hearts with end-stage dilated cardiomyopathy (DCM) and four non-failing heart controls were subjected to proteomic analysis. Out of 48 identified proteins, 26 proteins were redundant between samples. Ten of these 26 proteins were previously reported to be associated with HF. Of the newly identified proteins, we found several muscle proteins and mitochondrial/electron transport proteins, while novel were functionally similar to previous reports. However, we also found novel proteins involved in functional classes such as β-oxidation and G-protein coupled receptor signaling and desensitization not previously associated with HF. We also performed radioligand-binding studies on the heart samples and not only confirmed a large loss of β(1)-ARs in end-stage DCM, but also found a selective decrease in the α(1A)-AR subtype not previously reported. We have identified new proteins and functional categories associated with end-stage DCM. We also report that similar to the previously characterized loss of β(1)-AR in HF, there is also a concomitant loss of α(1A)-ARs, which are considered cardioprotective proteins.
Collapse
Affiliation(s)
- Ting Shi
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland, OH, USA
| | | | | |
Collapse
|
49
|
Akinaga J, Lima V, Kiguti LRDA, Hebeler-Barbosa F, Alcántara-Hernández R, García-Sáinz JA, Pupo AS. Differential phosphorylation, desensitization, and internalization of α1A-adrenoceptors activated by norepinephrine and oxymetazoline. Mol Pharmacol 2013; 83:870-81. [PMID: 23364786 DOI: 10.1124/mol.112.082313] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Loss of response on repetitive drug exposure (i.e., tachyphylaxis) is a particular problem for the vasoconstrictor effects of medications containing oxymetazoline (OXY), an α1-adrenoceptor (AR) agonist of the imidazoline class. One cause of tachyphylaxis is receptor desensitization, usually accompanied by phosphorylation and internalization. It is well established that α1A-ARs are less phosphorylated, desensitized, and internalized on exposure to the phenethylamines norepinephrine (NE), epinephrine, or phenylephrine (PE) than are the α1B and α1D subtypes. However, here we show in human embryonic kidney-293 cells that the low-efficacy agonist OXY induces G protein-coupled receptor kinase 2-dependent α1A-AR phosphorylation, followed by rapid desensitization and internalization (∼40% internalization after 5 minutes of stimulation), whereas phosphorylation of α1A-ARs exposed to NE depends to a large extent on protein kinase C activity and is not followed by desensitization, and the receptors undergo delayed internalization (∼35% after 60 minutes of stimulation). Native α1A-ARs from rat tail artery and vas deferens are also desensitized by OXY, but not by NE or PE, indicating that this property of OXY is not limited to recombinant receptors expressed in cell systems. The results of the present study are clearly indicative of agonist-directed α1A-AR regulation. OXY shows functional selectivity relative to NE and PE at α1A-ARs, leading to significant receptor desensitization and internalization, which is important in view of the therapeutic vasoconstrictor effects of this drug and the varied biologic process regulated by α1A-ARs.
Collapse
Affiliation(s)
- Juliana Akinaga
- Department of Pharmacology, Instituto de Biociências, Universidade Estadual Paulista (UNESP), Botucatu, São Paulo, Brazil
| | | | | | | | | | | | | |
Collapse
|
50
|
Odnoshivkina UG, Petrov AM, Zefirov AL. Mechanisms of delayed inotropic response of mouse atria to activation of β(2)-adrenoreceptors. DOKLADY BIOLOGICAL SCIENCES : PROCEEDINGS OF THE ACADEMY OF SCIENCES OF THE USSR, BIOLOGICAL SCIENCES SECTIONS 2012; 446:290-2. [PMID: 23129275 DOI: 10.1134/s0012496612050134] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2012] [Indexed: 11/23/2022]
|