1
|
Bucher ML, Dicent J, Duarte Hospital C, Miller GW. Neurotoxicology of dopamine: Victim or assailant? Neurotoxicology 2024; 103:175-188. [PMID: 38857676 DOI: 10.1016/j.neuro.2024.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 06/02/2024] [Accepted: 06/03/2024] [Indexed: 06/12/2024]
Abstract
Since the identification of dopamine as a neurotransmitter in the mid-20th century, investigators have examined the regulation of dopamine homeostasis at a basic biological level and in human disorders. Genetic animal models that manipulate the expression of proteins involved in dopamine homeostasis have provided key insight into the consequences of dysregulated dopamine. As a result, we have come to understand the potential of dopamine to act as an endogenous neurotoxin through the generation of reactive oxygen species and reactive metabolites that can damage cellular macromolecules. Endogenous factors, such as genetic variation and subcellular processes, and exogenous factors, such as environmental exposures, have been identified as contributors to the dysregulation of dopamine homeostasis. Given the variety of dysregulating factors that impact dopamine homeostasis and the potential for dopamine itself to contribute to further cellular dysfunction, dopamine can be viewed as both the victim and an assailant of neurotoxicity. Parkinson's disease has emerged as the exemplar case study of dopamine dysregulation due to the genetic and environmental factors known to contribute to disease risk, and due to the evidence of dysregulated dopamine as a pathologic and pathogenic feature of the disease. This review, inspired by the talk, "Dopamine in Durham: location, location, location" presented by Dr. Miller for the Jacob Hooisma Memorial Lecture at the International Neurotoxicology Association meeting in 2023, offers a primer on dopamine toxicity covering endogenous and exogenous factors that disrupt dopamine homeostasis and the actions of dopamine as an endogenous neurotoxin.
Collapse
Affiliation(s)
- Meghan L Bucher
- Department of Environmental Health Sciences, Mailman School of Public Health at Columbia University, New York, NY 10032, USA
| | - Jocelyn Dicent
- Department of Environmental Health Sciences, Mailman School of Public Health at Columbia University, New York, NY 10032, USA
| | - Carolina Duarte Hospital
- Department of Environmental Health Sciences, Mailman School of Public Health at Columbia University, New York, NY 10032, USA
| | - Gary W Miller
- Department of Environmental Health Sciences, Mailman School of Public Health at Columbia University, New York, NY 10032, USA; Department of Molecular Pharmacology and Therapeutics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
2
|
He Z, Yin BK, Wang K, Zhao B, Chen Y, Li ZC, Chen J. The alpha2-adrenergic receptor agonist clonidine protects against cerebral ischemia/reperfusion induced neuronal apoptosis in rats. Metab Brain Dis 2024; 39:741-752. [PMID: 38833094 DOI: 10.1007/s11011-024-01354-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 05/06/2024] [Indexed: 06/06/2024]
Abstract
Apoptosis is the crucial pathological mechanism following cerebral ischemic injury. Our previous studies demonstrated that clonidine, one agonist of alpha2-adrenergic receptor (α2-AR), could attenuate cerebral ischemic injury in a rat model of middle cerebral artery occlusion/reperfusion (MCAO/R). However, it's unclear whether clonidine exerts neuroprotective effects by regulating neuronal apoptosis. In this study, we elucidated whether clonidine can exert anti-apoptotic effects in cerebral ischemic injury, and further explored the possible mechanisms. Neurological deficit score was measured to evaluate the neurological function. TTC staining was used for the measurement of brain infarct size. Hematoxylin-Eosin (HE) staining was applied to examine the cell morphology. TUNEL and DAPI fluorescent staining methods were used to analyze the cell apoptosis in brain tissue. Fluorescence quantitative real-time PCR was performed to assess the gene expression of Caspase-3 and P53. Western blotting assay was applied to detect the protein expression of Caspase-3 and P53. The results showed that clonidine improved neurological function, reduced brain infarct size, alleviated neuronal damage, and reduced the ratio of cell apoptosis in the brain with MCAO/R injury. moreover, clonidine down-regulated the gene and protein expression of Caspase-3 and P53 which were over-expressed after MCAO/R injury. Whereas, yohimbine (one selective α2-AR antagonist) mitigated the anti-apoptosis effects of clonidine, accompanied by reversed gene and protein expression changes. The results indicated that clonidine attenuated cerebral MCAO/R injury via suppressing neuronal apoptosis, which may be mediated, at least in part, by activating α2-AR.
Collapse
Affiliation(s)
- Zhi He
- Department of Pharmacology, College of Medicine, Jiaxing University, 314001, Jiaxing, People's Republic of China.
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, 443002, Yichang, People's Republic of China.
- College of Basic Medical Sciences, China Three Gorges University, 443002, Yichang, People's Republic of China.
| | - Bo-Kai Yin
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, 443002, Yichang, People's Republic of China
- Yichang Yiling Hospital, 443000, Yichang, People's Republic of China
- Zhongnan Hospital of Wuhan University, 430071, Wuhan, People's Republic of China
| | - Ke Wang
- Department of Pharmacology, College of Medicine, Jiaxing University, 314001, Jiaxing, People's Republic of China
| | - Bo Zhao
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, 443002, Yichang, People's Republic of China
- College of Basic Medical Sciences, China Three Gorges University, 443002, Yichang, People's Republic of China
| | - Yue Chen
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, 443002, Yichang, People's Republic of China
- College of Basic Medical Sciences, China Three Gorges University, 443002, Yichang, People's Republic of China
| | - Zi-Cheng Li
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, 443002, Yichang, People's Republic of China.
- College of Basic Medical Sciences, China Three Gorges University, 443002, Yichang, People's Republic of China.
| | - Jing Chen
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, 443002, Yichang, People's Republic of China.
- College of Medicine and Health Sciences, China Three Gorges University, No.8 Daxue Road, 443002, Yichang, People's Republic of China.
| |
Collapse
|
3
|
Maaliki D, Jaffa AA, Nasser S, Sahebkar A, Eid AH. Adrenoceptor Desensitization: Current Understanding of Mechanisms. Pharmacol Rev 2024; 76:358-387. [PMID: 38697858 DOI: 10.1124/pharmrev.123.000831] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 01/15/2024] [Accepted: 01/18/2024] [Indexed: 05/05/2024] Open
Abstract
G-protein coupled receptors (GPCRs) transduce a wide range of extracellular signals. They are key players in the majority of biologic functions including vision, olfaction, chemotaxis, and immunity. However, as essential as most of them are to body function and homeostasis, overactivation of GPCRs has been implicated in many pathologic diseases such as cancer, asthma, and heart failure (HF). Therefore, an important feature of G protein signaling systems is the ability to control GPCR responsiveness, and one key process to control overstimulation involves initiating receptor desensitization. A number of steps are appreciated in the desensitization process, including cell surface receptor phosphorylation, internalization, and downregulation. Rapid or short-term desensitization occurs within minutes and involves receptor phosphorylation via the action of intracellular protein kinases, the binding of β-arrestins, and the consequent uncoupling of GPCRs from their cognate heterotrimeric G proteins. On the other hand, long-term desensitization occurs over hours to days and involves receptor downregulation or a decrease in cell surface receptor protein level. Of the proteins involved in this biologic phenomenon, β-arrestins play a particularly significant role in both short- and long-term desensitization mechanisms. In addition, β-arrestins are involved in the phenomenon of biased agonism, where the biased ligand preferentially activates one of several downstream signaling pathways, leading to altered cellular responses. In this context, this review discusses the different patterns of desensitization of the α 1-, α 2- and the β adrenoceptors and highlights the role of β-arrestins in regulating physiologic responsiveness through desensitization and biased agonism. SIGNIFICANCE STATEMENT: A sophisticated network of proteins orchestrates the molecular regulation of GPCR activity. Adrenoceptors are GPCRs that play vast roles in many physiological processes. Without tightly controlled desensitization of these receptors, homeostatic imbalance may ensue, thus precipitating various diseases. Here, we critically appraise the mechanisms implicated in adrenoceptor desensitization. A better understanding of these mechanisms helps identify new druggable targets within the GPCR desensitization machinery and opens exciting therapeutic fronts in the treatment of several pathologies.
Collapse
Affiliation(s)
- Dina Maaliki
- Department of Pharmacology and Toxicology, American University of Beirut, Beirut, Lebanon (D.M.); School of Medicine, University of South Carolina, Columbia, South Carolina (A.A.J.); Keele University, Staffordshire, United Kingdom (S.N.); Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran (A.S.); Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran (A.S.); and Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar (A.H.E.)
| | - Aneese A Jaffa
- Department of Pharmacology and Toxicology, American University of Beirut, Beirut, Lebanon (D.M.); School of Medicine, University of South Carolina, Columbia, South Carolina (A.A.J.); Keele University, Staffordshire, United Kingdom (S.N.); Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran (A.S.); Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran (A.S.); and Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar (A.H.E.)
| | - Suzanne Nasser
- Department of Pharmacology and Toxicology, American University of Beirut, Beirut, Lebanon (D.M.); School of Medicine, University of South Carolina, Columbia, South Carolina (A.A.J.); Keele University, Staffordshire, United Kingdom (S.N.); Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran (A.S.); Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran (A.S.); and Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar (A.H.E.)
| | - Amirhossein Sahebkar
- Department of Pharmacology and Toxicology, American University of Beirut, Beirut, Lebanon (D.M.); School of Medicine, University of South Carolina, Columbia, South Carolina (A.A.J.); Keele University, Staffordshire, United Kingdom (S.N.); Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran (A.S.); Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran (A.S.); and Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar (A.H.E.)
| | - Ali H Eid
- Department of Pharmacology and Toxicology, American University of Beirut, Beirut, Lebanon (D.M.); School of Medicine, University of South Carolina, Columbia, South Carolina (A.A.J.); Keele University, Staffordshire, United Kingdom (S.N.); Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran (A.S.); Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran (A.S.); and Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar (A.H.E.)
| |
Collapse
|
4
|
Callaway CW, Flickinger KL, Weissman A, Guyette FX, DeMaio R, Jonsson A, Wu V, Monteleone JL, Prescott P, Birabaharan J, Buysse DJ, Empey PE, Nolin TD, West RE. Alpha-2-adrenergic agonists reduce resting energy expenditure in humans during external cooling. Temperature (Austin) 2024; 11:280-298. [PMID: 39193049 PMCID: PMC11346546 DOI: 10.1080/23328940.2024.2339781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 04/02/2024] [Accepted: 04/02/2024] [Indexed: 08/29/2024] Open
Abstract
Intravenous alpha-2-adrenergic receptor agonists reduce energy expenditure and lower the temperature when shivering begins in humans, allowing a decrease in core body temperature. Because there are few data about similar effects from oral drugs, we tested whether single oral doses of the sedative dexmedetomidine (1 µg/kg sublingual or 4 µg/kg swallowed) or the muscle relaxant tizanidine (8 mg or 16 mg), combined with surface cooling, reduce energy expenditure and core body temperature in humans. A total of 26 healthy participants completed 41 one-day laboratory studies measuring core body temperature using an ingested telemetry capsule and measuring energy expenditure using indirect calorimetry for up to 6 hours after drug ingestion. Dexmedetomidine induced a median 13% - 19% peak reduction and tizanidine induced a median 15% - 22% peak reduction in energy expenditure relative to baseline. Core body temperature decreased a median of 0.5°C - 0.6°C and 0.5°C - 0.7°C respectively. Decreases in temperature occurred after peak reductions in energy expenditure. Energy expenditure increased with a decrease in core temperature in control participants but did not occur after 4 µg/kg dexmedetomidine or 16 mg tizanidine. Plasma levels of dexmedetomidine but not tizanidine were related to mean temperature change. Decreases in heart rate, blood pressure, respiratory rate, cardiac stroke volume index, and cardiac index were associated with the change in metabolic rate after higher drug doses. We conclude that both oral dexmedetomidine and oral tizanidine reduce energy expenditure and allow decrease in core temperature in humans.
Collapse
Affiliation(s)
- Clifton W. Callaway
- Department of Emergency Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Katharyn L. Flickinger
- Department of Emergency Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Alexandra Weissman
- Department of Emergency Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Francis X. Guyette
- Department of Emergency Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Ryann DeMaio
- Department of Emergency Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Andrea Jonsson
- Department of Emergency Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Victor Wu
- Department of Emergency Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jenna L. Monteleone
- Department of Emergency Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Peter Prescott
- Department of Emergency Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jonathan Birabaharan
- Center for Clinical Pharmaceutical Sciences, Department of Pharmacy and Therapeutics, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, USA
| | - Daniel J. Buysse
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Philip E. Empey
- Center for Clinical Pharmaceutical Sciences, Department of Pharmacy and Therapeutics, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, USA
| | - Thomas D. Nolin
- Center for Clinical Pharmaceutical Sciences, Department of Pharmacy and Therapeutics, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, USA
| | - Raymond E. West
- Center for Clinical Pharmaceutical Sciences, Department of Pharmacy and Therapeutics, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, USA
| |
Collapse
|
5
|
Naderi S, Tamaddonfard E, Nafisi S, Soltanalinejad-Taghiabad F. Effect of thymoquinone on acetic acid-induced visceral nociception in rats: role of central cannabinoid and α 2-adrenergic receptors. VETERINARY RESEARCH FORUM : AN INTERNATIONAL QUARTERLY JOURNAL 2024; 15:131-138. [PMID: 38770373 PMCID: PMC11102585 DOI: 10.30466/vrf.2023.2005364.3922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 10/16/2023] [Indexed: 05/22/2024]
Abstract
Thymoquinone (TQ) is the main biologically active substance of Nigella sativa (black seeds). It has anti-cancer, anti-inflammatory, anti-diabetic, anti-oxidative and anti-nociceptive properties. This study was aimed to explore the effect of TQ on acetic acid-induced visceral nociception. The central mechanisms of the effect of TQ were investigated using cannabinergic (AM251) and α2-adrenergic (yohimbine [Yoh]) antagonists. The lateral ventricle of the brain was cannulated for intracerebroventricular (ICV) injections. Visceral nociception was induced by intra-peritoneal (IP) injection of acetic acid (1.00% in a volume of 1.00 mL). Measuring the latency time to the first writhing appearance and counting the number of writhing in 5-min intervals for a period of 60 min were performed. Locomotor activity was determined using an open-field test. Oral administration (PO) of 2.50 and 10.00 mg kg-1 TQ increased the latency time to the first writhing appearance and decreased the number of writhing. The AM251 (5.00 µg per rat; ICV) and Yoh (5.00 µg per rat; ICV) partially prevented TQ (10.00 mg kg-1; PO)-induced anti-nociception. Locomotor activity was not altered by these treatments. The results of the present study showed that TQ had the ability to reduce visceral nociception caused by IP injection of acetic acid. The central mechanisms of this action of TQ might be partially mediated by cannabinergic and α2-adrenegic receptors.
Collapse
Affiliation(s)
- Somayyeh Naderi
- PhD Candidate, Department of Basic Sciences, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran;
| | - Esmaeal Tamaddonfard
- Department of Basic Sciences, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran.
| | - Saeid Nafisi
- Department of Basic Sciences, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran.
| | | |
Collapse
|
6
|
Ruan Y, Buonfiglio F, Gericke A. Adrenoceptors in the Eye - Physiological and Pathophysiological Relevance. Handb Exp Pharmacol 2024; 285:453-505. [PMID: 38082203 DOI: 10.1007/164_2023_702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
The autonomic nervous system plays a crucial role in the innervation of the eye. Consequently, it comes as no surprise that catecholamines and their corresponding receptors have been extensively studied and characterized in numerous ocular structures, including the cornea, conjunctiva, lacrimal gland, trabecular meshwork, uvea, and retina. These investigations have unveiled substantial clinical implications, particularly in the context of treating glaucoma, a progressive neurodegenerative disorder responsible for irreversible vision loss on a global scale. The primary therapeutic approaches for glaucoma frequently involve the modulation of α1-, α2-, and β-adrenoceptors, making them pivotal targets. In this chapter, we offer a comprehensive overview of the expression, distribution, and functional roles of adrenoceptors within various components of the eye and its associated structures. Additionally, we delve into the pivotal role of adrenoceptors in the pathophysiology of glaucoma. Furthermore, we provide a concise historical perspective on adrenoceptor research, examine the distinct contributions of individual adrenoceptor subtypes to the treatment of various ocular conditions, and propose potential future avenues of exploration in this field.
Collapse
Affiliation(s)
- Yue Ruan
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Francesco Buonfiglio
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Adrian Gericke
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany.
| |
Collapse
|
7
|
Brooke AK, Murrow DP, Caldwell KCN, Witt CE, Ross AE. Measuring neuron-regulated immune cell physiology via the alpha-2 adrenergic receptor in an ex vivo murine spleen model. Cell Mol Life Sci 2023; 80:354. [PMID: 37945921 PMCID: PMC11071927 DOI: 10.1007/s00018-023-05012-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/27/2023] [Accepted: 10/23/2023] [Indexed: 11/12/2023]
Abstract
The communication between the nervous and immune systems plays a crucial role in regulating immune cell function and inflammatory responses. Sympathetic neurons, which innervate the spleen, have been implicated in modulating immune cell activity. The neurotransmitter norepinephrine (NE), released by sympathetic neurons, influences immune cell responses by binding to adrenergic receptors on their surface. The alpha-2 adrenergic receptor (α2AR), expressed predominantly on sympathetic neurons, has received attention due to its autoreceptor function and ability to modulate NE release. In this study, we used fast-scan cyclic voltammetry (FSCV) to provide the first subsecond measurements of NE released in the white pulp region of the spleen and validated it with yohimbine, a known antagonist of α2AR. For further application of FSCV in neuroimmunology, we investigated the extent to which subsecond NE from sympathetic neurons is important for immune cell physiology and cytokine production, focusing on tumor necrosis factor-alpha (TNF-α), interleukin-10 (IL-10), and interleukin-6 (IL-6). Our findings provide insights into the regulatory mechanisms underlying sympathetic-immune interactions and show the significance of using FSCV, a traditional neurochemistry technique, to study these neuroimmune mechanisms.
Collapse
Affiliation(s)
- Alexandra K Brooke
- Department of Chemistry, University of Cincinnati, 312 College Dr. 404 Crosley Tower, Cincinnati, OH, 45221-0172, USA
| | - Daniel P Murrow
- Department of Chemistry, University of Cincinnati, 312 College Dr. 404 Crosley Tower, Cincinnati, OH, 45221-0172, USA
| | - Kaejaren C N Caldwell
- Department of Chemistry, University of Cincinnati, 312 College Dr. 404 Crosley Tower, Cincinnati, OH, 45221-0172, USA
| | - Colby E Witt
- Department of Chemistry, University of Cincinnati, 312 College Dr. 404 Crosley Tower, Cincinnati, OH, 45221-0172, USA
| | - Ashley E Ross
- Department of Chemistry, University of Cincinnati, 312 College Dr. 404 Crosley Tower, Cincinnati, OH, 45221-0172, USA.
| |
Collapse
|
8
|
Kielbinski M, Bernacka J, Zajda K, Wawrzczak-Bargieła A, Maćkowiak M, Przewlocki R, Solecki W. Acute stress modulates noradrenergic signaling in the ventral tegmental area-amygdalar circuit. J Neurochem 2023; 164:598-612. [PMID: 36161462 DOI: 10.1111/jnc.15698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 09/09/2022] [Accepted: 09/22/2022] [Indexed: 11/29/2022]
Abstract
Noradrenergic neurotransmission is a critical mediator of stress responses. In turn, exposure to stress induces noradrenergic system adaptations, some of which are implicated in the etiology of stress-related disorders. Adrenergic receptors (ARs) in the ventral tegmental area (VTA) have been demonstrated to regulate phasic dopamine (DA) release in the forebrain, necessary for behavioral responses to conditional cues. However, the impact of stress on noradrenergic modulation of the VTA has not been previously explored. We demonstrate that ARs in the VTA regulate dopaminergic activity in the VTA-BLA (basolateral amygdala) circuit, a key system for processing stress-related stimuli; and that such control is altered by acute stress. We utilized fast-scan cyclic voltammetry to assess the effects of intra-VTA microinfusion of α1 -AR and α2 -AR antagonists (terazosin and RX-821002, respectively), on electrically evoked phasic DA release in the BLA in stress-naïve and stressed (unavoidable electric shocks - UES) anesthetized male Sprague-Dawley rats. In addition, we used western blotting to explore UES-induced alterations in AR protein level in the VTA. Intra-VTA terazosin or RX-821002 dose-dependently attenuated DA release in the BLA. Interestingly, UES decreased the effects of intra-VTA α2 -AR blockade on DA release (24 h but not 7 days after stress), while the effects of terazosin were unchanged. Despite changes in α2 -AR physiological function in the VTA, UES did not alter α2 -AR protein levels in either intracellular or membrane fractions. These findings demonstrate that NA-ergic modulation of the VTA-BLA circuit undergoes significant alterations in response to acute stress, with α2 -AR signaling indicated as a key target.
Collapse
Affiliation(s)
- Michal Kielbinski
- Department of Neurobiology and Neuropsychology, Jagiellonian University, Institute of Applied Psychology, Krakow, Poland
| | - Joanna Bernacka
- Department of Neurobiology and Neuropsychology, Jagiellonian University, Institute of Applied Psychology, Krakow, Poland.,Department of Pharmacology, Laboratory of Pharmacology and Brain Biostructure, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Katarzyna Zajda
- Department of Neurobiology and Neuropsychology, Jagiellonian University, Institute of Applied Psychology, Krakow, Poland
| | - Agnieszka Wawrzczak-Bargieła
- Department of Pharmacology, Laboratory of Pharmacology and Brain Biostructure, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Marzena Maćkowiak
- Department of Pharmacology, Laboratory of Pharmacology and Brain Biostructure, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Ryszard Przewlocki
- Department of Molecular Neuropharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Wojciech Solecki
- Department of Neurobiology and Neuropsychology, Jagiellonian University, Institute of Applied Psychology, Krakow, Poland
| |
Collapse
|
9
|
Sedative Properties of Dexmedetomidine Are Mediated Independently from Native Thalamic Hyperpolarization-Activated Cyclic Nucleotide-Gated Channel Function at Clinically Relevant Concentrations. Int J Mol Sci 2022; 24:ijms24010519. [PMID: 36613961 PMCID: PMC9820684 DOI: 10.3390/ijms24010519] [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: 11/24/2022] [Revised: 12/19/2022] [Accepted: 12/20/2022] [Indexed: 12/31/2022] Open
Abstract
Dexmedetomidine is a selective α2-adrenoceptor agonist and appears to disinhibit endogenous sleep-promoting pathways, as well as to attenuate noradrenergic excitation. Recent evidence suggests that dexmedetomidine might also directly inhibit hyperpolarization-activated cyclic-nucleotide gated (HCN) channels. We analyzed the effects of dexmedetomidine on native HCN channel function in thalamocortical relay neurons of the ventrobasal complex of the thalamus from mice, performing whole-cell patch-clamp recordings. Over a clinically relevant range of concentrations (1-10 µM), the effects of dexmedetomidine were modest. At a concentration of 10 µM, dexmedetomidine significantly reduced maximal Ih amplitude (relative reduction: 0.86 [0.78-0.91], n = 10, and p = 0.021), yet changes to the half-maximal activation potential V1/2 occurred exclusively in the presence of the very high concentration of 100 µM (-4,7 [-7.5--4.0] mV, n = 10, and p = 0.009). Coincidentally, only the very high concentration of 100 µM induced a significant deceleration of the fast component of the HCN activation time course (τfast: +135.1 [+64.7-+151.3] ms, n = 10, and p = 0.002). With the exception of significantly increasing the membrane input resistance (starting at 10 µM), dexmedetomidine did not affect biophysical membrane properties and HCN channel-mediated parameters of neuronal excitability. Hence, the sedative qualities of dexmedetomidine and its effect on the thalamocortical network are not decisively shaped by direct inhibition of HCN channel function.
Collapse
|
10
|
Munawar N, Nader J, Khadadah NH, Al Madhoun A, Al-Ali W, Varghese LA, Masocha W, Al-Mulla F, Bitar MS. Guanfacine Normalizes the Overexpression of Presynaptic α-2A Adrenoceptor Signaling and Ameliorates Neuropathic Pain in a Chronic Animal Model of Type 1 Diabetes. Pharmaceutics 2022; 14:pharmaceutics14102146. [PMID: 36297581 PMCID: PMC9609777 DOI: 10.3390/pharmaceutics14102146] [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: 06/23/2022] [Revised: 10/01/2022] [Accepted: 10/03/2022] [Indexed: 11/25/2022] Open
Abstract
Background: Diabetes is associated with several complications, including neuropathic pain, which is difficult to manage with currently available drugs. Descending noradrenergic neurons possess antinociceptive activity; however, their involvement in diabetic neuropathic pain remains to be explored. Methods: To infer the regulatory role of this system, we examined as a function of diabetes, the expression and localization of alpha-2A adrenoceptors (α2-AR) in the dorsal root ganglia and key regions of the central nervous system, including pons and lumbar segment of the spinal cord using qRT-PCR, Western blotting, and immunofluorescence-based techniques. Results: The data revealed that presynaptic synaptosomal-associated protein-25 labeled α2-AR in the central and peripheral nervous system of streptozotocin diabetic rats was upregulated both at the mRNA and protein levels. Interestingly, the levels of postsynaptic density protein-95 labeled postsynaptic neuronal α2-AR remained unaltered as a function of diabetes. These biochemical abnormalities in the noradrenergic system of diabetic animals were associated with increased pain sensitivity as typified by the presence of thermal hyperalgesia and cold/mechanical allodynia. The pain-related behaviors were assessed using Hargreaves apparatus, cold-plate and dynamic plantar aesthesiometer. Chronically administered guanfacine, a selective α2-AR agonist, to diabetic animals downregulated the upregulation of neuronal presynaptic α2-AR and ameliorated the hyperalgesia and the cold/mechanical allodynia in these animals. Conclusion: Together, these findings demonstrate that guanfacine may function as a potent analgesic and highlight α2-AR, a key component of the descending neuronal autoinhibitory pathway, as a potential therapeutic target in the treatment of diabetic neuropathic pain.
Collapse
Affiliation(s)
- Neha Munawar
- Department of Pharmacology and Toxicology, Faculty of Medicine, Kuwait University, Al-Jabriya 046302, Kuwait
| | - Joelle Nader
- Department of Mathematics and Natural Sciences, American University of Kuwait, Salmiya 20002, Kuwait
| | - Najat H. Khadadah
- Department of Pharmacology and Toxicology, Faculty of Medicine, Kuwait University, Al-Jabriya 046302, Kuwait
| | - Ashraf Al Madhoun
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman 15400, Kuwait
- Department of Animal and Imaging Core Facilities, Dasman Diabetes Institute, Dasman 15400, Kuwait
| | - Waleed Al-Ali
- Department of Pathology, Faculty of Medicine, Kuwait University, Al-Jabriya 046302, Kuwait
| | - Linu A. Varghese
- Department of Pharmacology and Toxicology, Faculty of Medicine, Kuwait University, Al-Jabriya 046302, Kuwait
| | - Willias Masocha
- Department of Pharmacology and Therapeutics, College of Pharmacy, Kuwait University, Al-Jabriya 046302, Kuwait
| | - Fahd Al-Mulla
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman 15400, Kuwait
| | - Milad S. Bitar
- Department of Pharmacology and Toxicology, Faculty of Medicine, Kuwait University, Al-Jabriya 046302, Kuwait
- Correspondence:
| |
Collapse
|
11
|
Proudman RGW, Akinaga J, Baker JG. The signaling and selectivity of α-adrenoceptor agonists for the human α2A, α2B and α2C-adrenoceptors and comparison with human α1 and β-adrenoceptors. Pharmacol Res Perspect 2022; 10:e01003. [PMID: 36101495 PMCID: PMC9471048 DOI: 10.1002/prp2.1003] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/05/2022] [Accepted: 08/15/2022] [Indexed: 11/11/2022] Open
Abstract
α2-adrenoceptors, (α2A, α2B and α2C-subtypes), are Gi-coupled receptors. Central activation of brain α2A and α2C-adrenoceptors is the main site for α2-agonist mediated clinical responses in hypertension, ADHD, muscle spasm and ITU management of sedation, reduction in opiate requirements, nausea and delirium. However, despite having the same Gi-potency in functional assays, some α2-agonists also stimulate Gs-responses whilst others do not. This was investigated. Agonist responses to 49 different α-agonists were studied (CRE-gene transcription, cAMP, ERK1/2-phosphorylation and binding affinity) in CHO cells stably expressing the human α2A, α2B or α2C-adrenoceptor, enabling ligand intrinsic efficacy to be determined (binding KD /Gi-IC50 ). Ligands with high intrinsic efficacy (e.g., brimonidine and moxonidine at α2A) stimulated biphasic (Gi-Gs) concentration responses, however for ligands with low intrinsic efficacy (e.g., naphazoline), responses were monophasic (Gi-only). ERK1/2-phosphorylation responses appeared to be Gi-mediated. For Gs-mediated responses to be observed, both a system with high receptor reserve and high agonist intrinsic efficacy were required. From the Gi-mediated efficacy ratio, the degree of Gs-coupling could be predicted. The clinical relevance and precise receptor conformational changes that occur, given the structural diversity of compounds with high intrinsic efficacy, remains to be determined. Comparison with α1 and β1/β2-adrenoceptors demonstrated subclass affinity selectivity for some compounds (e.g., α2:dexmedetomidine, α1:A61603) whilst e.g., oxymetazoline had high affinity for both α2A and α1A-subtypes, compared to all others. Some compounds had subclass selectivity due to selective intrinsic efficacy (e.g., α2:brimonidine, α1:methoxamine/etilefrine). A detailed knowledge of these agonist characteristics is vital for improving computer-based deep-learning and drug design.
Collapse
Affiliation(s)
- Richard G. W. Proudman
- Cell Signalling Research Group, Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, C Floor Medical School, Queen's Medical CentreUniversity of NottinghamNottinghamUK
| | - Juliana Akinaga
- Cell Signalling Research Group, Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, C Floor Medical School, Queen's Medical CentreUniversity of NottinghamNottinghamUK
| | - Jillian G. Baker
- Cell Signalling Research Group, Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, C Floor Medical School, Queen's Medical CentreUniversity of NottinghamNottinghamUK
| |
Collapse
|
12
|
Joanna B, Michal K, Agnieszka WB, Katarzyna Z, Marzena M, Ryszard P, Wojciech S. Alpha-2A but not 2B/C noradrenergic receptors in ventral tegmental area regulate phasic dopamine release in nucleus accumbens core. Neuropharmacology 2022; 220:109258. [PMID: 36116534 DOI: 10.1016/j.neuropharm.2022.109258] [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: 04/19/2022] [Revised: 09/09/2022] [Accepted: 09/11/2022] [Indexed: 11/20/2022]
Abstract
Adrenergic receptors (AR) in the ventral tegmental area (VTA) modulate local neuronal activity and, as a consequence, dopamine (DA) release in the mesolimbic forebrain. Such modulation has functional significance: intra-VTA blockade of α1-AR attenuates behavioral responses to salient environmental stimuli in rat models of drug seeking and conditioned fear as well as phasic DA release in the nucleus accumbens (NAc). In contrast, α2-AR in the VTA has been suggested to act primarily as autoreceptors, limiting local noradrenergic input. The regulation of noradrenaline efflux by α2-AR could be of clinical interest, as α2-AR agonists are proposed as promising pharmacological tools in the treatment of PTSD and substance use disorder. Thus, the aim of our study was to determine the subtype-specificity of α2-ARs in the VTA capable of modulating phasic DA release. We used fast scan cyclic voltammetry (FSCV) in anaesthetized male rats to measure DA release in the NAc after combined electrical stimulation and infusion of selected α2-AR antagonists into the VTA. Intra-VTA microinfusion of idazoxan - a non-subtype-specific α2-AR antagonist, as well as BRL-44408 - a selective α2A-AR antagonist, attenuated electrically-evoked DA in the NAc. In contrast, local administration of JP-1302 or imiloxan (α2B- and α2C-AR antagonists, respectively) had no effect. The effect of BRL-44408 on DA release was attenuated by intra-VTA DA D2 antagonist (raclopride) pre-administration. Finally, we confirmed the presence of α2A-AR protein in the VTA using western blotting. In conclusion, these data specify α2A-, but not α2B- or α2C-AR as the receptor subtype controlling NA release in the VTA.
Collapse
Affiliation(s)
- Bernacka Joanna
- Jagiellonian University, Institute of Applied Psychology, Department of Neurobiology and Neuropsychology, Łojasiewicza Str. 4, 30-348, Krakow, Poland; Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Pharmacology, Laboratory of Pharmacology and Brain Biostructure, Smętna Str. 12, 31-343, Krakow, Poland
| | - Kielbinski Michal
- Jagiellonian University, Institute of Applied Psychology, Department of Neurobiology and Neuropsychology, Łojasiewicza Str. 4, 30-348, Krakow, Poland
| | - Wawrzczak-Bargieła Agnieszka
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Pharmacology, Laboratory of Pharmacology and Brain Biostructure, Smętna Str. 12, 31-343, Krakow, Poland
| | - Zajda Katarzyna
- Jagiellonian University, Institute of Applied Psychology, Department of Neurobiology and Neuropsychology, Łojasiewicza Str. 4, 30-348, Krakow, Poland
| | - Maćkowiak Marzena
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Pharmacology, Laboratory of Pharmacology and Brain Biostructure, Smętna Str. 12, 31-343, Krakow, Poland
| | - Przewlocki Ryszard
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Molecular Neuropharmacology, Smętna Str. 12, 31-343, Krakow, Poland
| | - Solecki Wojciech
- Jagiellonian University, Institute of Applied Psychology, Department of Neurobiology and Neuropsychology, Łojasiewicza Str. 4, 30-348, Krakow, Poland.
| |
Collapse
|
13
|
Alluri S, Eisenberg SM, Grisanti LA, Tanner M, Volkow ND, Kim SW, Kil KE. Preclinical evaluation of new C-11 labeled benzo-1,4-dioxane PET radiotracers for brain α2C adrenergic receptors. Eur J Med Chem 2022; 243:114764. [DOI: 10.1016/j.ejmech.2022.114764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 09/04/2022] [Accepted: 09/07/2022] [Indexed: 11/24/2022]
|
14
|
Rayff da Silva P, Diniz Nunes Pazos N, Karla Silva do Nascimento Gonzaga T, Cabral de Andrade J, Brito Monteiro Á, Caroline Ribeiro Portela A, Fernandes Oliveira Pires H, Dos Santos Maia M, Vilar da Fonsêca D, T Scotti M, Maria Barbosa Filho J, Pergentino de Sousa D, Francisco Bezerra Felipe C, Nóbrega de Almeida R, Scotti L. Anxiolytic and antidepressant-like effects of monoterpene tetrahydrolinalool and in silico approach of new potential targets. Curr Top Med Chem 2022; 22:1530-1552. [PMID: 35524664 DOI: 10.2174/1568026622666220505104726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 02/03/2022] [Accepted: 02/16/2022] [Indexed: 11/22/2022]
Abstract
INTRODUCTION- The drugs currently available for treatment of anxiety and depression act through modulation of the neurotransmission systems involved in the neurobiology of the disorder, yet they of-ten present side effects, which can impair patient adherence to treatment. METHOD- This, has driven the search for new molecules with anxiolytic and antidepressant potential. Aromatic plants are rich in essential oils, and their chemical constituents, such as monoterpenes, are be-ing studied for these disorders. This study aims to evaluate the anxiolytic and antidepressant-like poten-tial of the monoterpene tetrahydrolinalool in in vivo animal models, and review pharmacological targets with validation through molecular docking. Male Swiss mice (Mus musculus) were treated with THL (37.5-600 mg kg-1 p.o.) and submitted to the elevated plus maze, open field, rota rod, and forced swim tests. In the elevated plus-maze, THL at doses of 37.5 and 75 mg kg-1 induced a significant increase in the percentage of entries (72.7 and 64.3% respectively), and lengths of stay (80.3 and 76.8% respective-ly) in the open arms tests. RESULT- These doses did not compromise locomotor activity or motor coordination in the animals. In the open field, rota rod tests, and the forced swimming model, treatment with THL significantly reduced immobility times at doses of 150, 300, and 600 mg kg-1, and by respective percentages of 69.3, 60.9 and 68.7%. CONCLUSION- In molecular docking assay, which investigated potential targets, THL presented sat-isfactory energy values for: nNOs, SGC, IL-6, 5-HT1A, NMDAr, and D1. These demonstrate the po-tential of THL (a derivative of natural origin) in in vivo and in silico models, making it a drug candidate.
Collapse
Affiliation(s)
- Pablo Rayff da Silva
- Psychopharmacology Laboratory, Institute of Drugs and Medicines Research, Federal University of Paraíba, 58051-085, Via Ipê Amarelo, S/N, João Pessoa, Paraíba, Brazil
| | - Natalia Diniz Nunes Pazos
- Psychopharmacology Laboratory, Institute of Drugs and Medicines Research, Federal University of Paraíba, 58051-085, Via Ipê Amarelo, S/N, João Pessoa, Paraíba, Brazil
| | | | - Jéssica Cabral de Andrade
- Psychopharmacology Laboratory, Institute of Drugs and Medicines Research, Federal University of Paraíba, 58051-085, Via Ipê Amarelo, S/N, João Pessoa, Paraíba, Brazil
| | - Álefe Brito Monteiro
- Psychopharmacology Laboratory, Institute of Drugs and Medicines Research, Federal University of Paraíba, 58051-085, Via Ipê Amarelo, S/N, João Pessoa, Paraíba, Brazil
| | - Anne Caroline Ribeiro Portela
- Psychopharmacology Laboratory, Institute of Drugs and Medicines Research, Federal University of Paraíba, 58051-085, Via Ipê Amarelo, S/N, João Pessoa, Paraíba, Brazil
| | - Hugo Fernandes Oliveira Pires
- Psychopharmacology Laboratory, Institute of Drugs and Medicines Research, Federal University of Paraíba, 58051-085, Via Ipê Amarelo, S/N, João Pessoa, Paraíba, Brazil
| | - Mayara Dos Santos Maia
- Cheminformatics Laboratory, Institute of Drugs and Medicines Research, Federal University of Paraíba, 58051-900, Via Ipê Amarelo, S/N, João Pessoa, Paraíba, Brazil
| | - Diogo Vilar da Fonsêca
- Collegiate of Medicine, Federal University of São Francisco Valley, 48607-190, Rua Aurora, S/N, Bahia, Brazil
| | - Marcus T Scotti
- Cheminformatics Laboratory, Institute of Drugs and Medicines Research, Federal University of Paraíba, 58051-900, Via Ipê Amarelo, S/N, João Pessoa, Paraíba, Brazil
| | - José Maria Barbosa Filho
- Pharmaceutical Chemistry Laboratory, Department of Pharmaceutical Sciences, Federal University of Paraíba, 58051-900, Via Ipê Amarelo, S/N, João Pessoa, Brazil
| | - Damião Pergentino de Sousa
- Pharmaceutical Chemistry Laboratory, Department of Pharmaceutical Sciences, Federal University of Paraíba, 58051-900, Via Ipê Amarelo, S/N, João Pessoa, Brazil
| | - Cícero Francisco Bezerra Felipe
- Psychopharmacology Laboratory, Institute of Drugs and Medicines Research, Federal University of Paraíba, 58051-085, Via Ipê Amarelo, S/N, João Pessoa, Paraíba, Brazil
| | - Reinaldo Nóbrega de Almeida
- Psychopharmacology Laboratory, Institute of Drugs and Medicines Research, Federal University of Paraíba, 58051-085, Via Ipê Amarelo, S/N, João Pessoa, Paraíba, Brazil
| | - Luciana Scotti
- Cheminformatics Laboratory, Institute of Drugs and Medicines Research, Federal University of Paraíba, 58051-900, Via Ipê Amarelo, S/N, João Pessoa, Paraíba, Brazil
| |
Collapse
|
15
|
Yim YY, McDonald WH, Betke KM, Kaya A, Hyde K, Erreger K, Gilsbach R, Hein L, Hamm HE. Specificities of Gβγ subunits for the SNARE complex before and after stimulation of α 2a-adrenergic receptors. Sci Signal 2021; 14:eabc4970. [PMID: 34932372 DOI: 10.1126/scisignal.abc4970] [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] [Indexed: 11/02/2022]
Abstract
Ligand binding to G protein–coupled receptors (GPCRs), such as the α2a-adrenergic receptor (α2aAR), results in the activation of heterotrimeric G proteins, which consist of functionally distinct Gα subunits and Gβγ dimers. α2aAR-dependent inhibition of synaptic transmission regulates functions such as spontaneous locomotor activity, anesthetic sparing, and working memory enhancement and requires the soluble NSF attachment protein receptor (SNARE) complex, a Gβγ effector. To understand how the Gβγ-SNARE complex underlies the α2aAR-dependent inhibition of synaptic transmission, we examined the specificity of Gβγ subunits for the SNARE complex in adrenergic neurons, in which auto-α2aARs respond to epinephrine released from these neurons, and nonadrenergic neurons, in which hetero-α2aARs respond to epinephrine released from other neurons. We performed a quantitative, targeted multiple reaction monitoring proteomic analysis of Gβ and Gγ subunits bound to the SNARE complex in synaptosomes from mouse brains. In the absence of stimulation of auto-α2aARs, Gβ1 and Gγ3 interacted with the SNARE complex. However, Gβ1, Gβ2, and Gγ3 were found in the complex when auto-α2aARs were activated by epinephrine. Further understanding of the specific usage of distinct Gβγ subunits in vivo may provide insights into the homeostatic regulation of synaptic transmission and the mechanisms of dysfunction that occur in neurological diseases.
Collapse
Affiliation(s)
- Yun Young Yim
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| | - W Hayes McDonald
- Department of Biochemistry and Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN 37232, USA
| | - Katherine M Betke
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| | - Ali Kaya
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| | - Karren Hyde
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| | - Kevin Erreger
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| | - Ralf Gilsbach
- Fachbereich Medizin, Institute for Cardiovascular Physiology, Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Lutz Hein
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Heidi E Hamm
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| |
Collapse
|
16
|
Fernandes DC, Martins BP, Silva GPD, Fonseca END, Santos SVM, Velozo LSM, Gayer CRM, Sabino KCDC, Coelho MGP. Echinodorus macrophyllus fraction with a high level of flavonoid inhibits peripheral and central mechanisms of nociception. J Tradit Complement Med 2021; 12:123-130. [PMID: 35528477 PMCID: PMC9072821 DOI: 10.1016/j.jtcme.2021.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 08/26/2020] [Accepted: 07/05/2021] [Indexed: 11/26/2022] Open
|
17
|
Cao LL, Marshall JM, Fabritz L, Brain KL. Resting cardiac sympathetic firing frequencies suppress terminal norepinephrine transporter uptake. Auton Neurosci 2021; 232:102794. [PMID: 33714751 DOI: 10.1016/j.autneu.2021.102794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 02/23/2021] [Accepted: 02/25/2021] [Indexed: 10/22/2022]
Abstract
The prejunctional norepinephrine transporter (NET) is responsible for the clearance of released norepinephrine (NE) back into the sympathetic nerve terminal. NET regulation must be tightly controlled as variations could have important implications for neurotransmission. Thus far, the effects of sympathetic neuronal activity on NET function have been unclear. Here, we optically monitor single-terminal cardiac NET activity ex vivo in response to a broad range of sympathetic postganglionic action potential (AP) firing frequencies. Isolated murine left atrial appendages were loaded with a fluorescent NET substrate [Neurotransmitter Transporter Uptake Assay (NTUA)] and imaged with confocal microscopy. Sympathetic APs were induced with electrical field stimulation at 0.2-10 Hz (0.1-0.2 ms pulse width). Exogenous NE was applied during the NTUA uptake- and washout phases to investigate substrate competition and displacement, respectively, on transport. Single-terminal NET reuptake rate was rapidly suppressed in a frequency-dependent manner with an inhibitory EF50 of 0.9 Hz. At 2 Hz, the effect was reversed by the α2-adrenoceptor antagonist yohimbine (1 μM) (p < 0.01) with no further effect imposed by the muscarinic receptor antagonist atropine (1 μM). Additionally, high exogenous NE concentrations abolished NET reuptake (1 μM NE; p < 0.0001) and displaced terminal specific NTUA during washout (1-100 μM NE; p < 0.0001). We have also identified α2-adrenoceptor-induced suppression of NET reuptake rate during resting stimulation frequencies, which could oppose the effect of autoinhibition-mediated suppression of exocytosis and thus amplify the effects of sympathetic drive on cardiac function.
Collapse
Affiliation(s)
- Lily L Cao
- School of Biomedical Science, Institute of Clinical Science, College of Medical and Dental Sciences, University of Birmingham, B15 2TT, UK.
| | - Janice M Marshall
- School of Biomedical Science, Institute of Clinical Science, College of Medical and Dental Sciences, University of Birmingham, B15 2TT, UK.
| | - Larissa Fabritz
- Institute of Cardiovascular Science, College of Medical and Dental Sciences, University of Birmingham, B15 2TT, UK; Department of Cardiology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK.
| | - Keith L Brain
- School of Biomedical Science, Institute of Clinical Science, College of Medical and Dental Sciences, University of Birmingham, B15 2TT, UK.
| |
Collapse
|
18
|
The Role of Adrenoceptors in the Retina. Cells 2020; 9:cells9122594. [PMID: 33287335 PMCID: PMC7761662 DOI: 10.3390/cells9122594] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 11/29/2020] [Accepted: 12/01/2020] [Indexed: 01/16/2023] Open
Abstract
The retina is a part of the central nervous system, a thin multilayer with neuronal lamination, responsible for detecting, preprocessing, and sending visual information to the brain. Many retinal diseases are characterized by hemodynamic perturbations and neurodegeneration leading to vision loss and reduced quality of life. Since catecholamines and respective bindings sites have been characterized in the retina, we systematically reviewed the literature with regard to retinal expression, distribution and function of alpha1 (α1)-, alpha2 (α2)-, and beta (β)-adrenoceptors (ARs). Moreover, we discuss the role of the individual adrenoceptors as targets for the treatment of retinal diseases.
Collapse
|
19
|
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: 0.8] [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
|
20
|
Saikia S, Bordoloi M, Sarmah R. Established and In-trial GPCR Families in Clinical Trials: A Review for Target Selection. Curr Drug Targets 2020; 20:522-539. [PMID: 30394207 DOI: 10.2174/1389450120666181105152439] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 08/28/2018] [Accepted: 10/22/2018] [Indexed: 12/14/2022]
Abstract
The largest family of drug targets in clinical trials constitute of GPCRs (G-protein coupled receptors) which accounts for about 34% of FDA (Food and Drug Administration) approved drugs acting on 108 unique GPCRs. Factors such as readily identifiable conserved motif in structures, 127 orphan GPCRs despite various de-orphaning techniques, directed functional antibodies for validation as drug targets, etc. has widened their therapeutic windows. The availability of 44 crystal structures of unique receptors, unexplored non-olfactory GPCRs (encoded by 50% of the human genome) and 205 ligand receptor complexes now present a strong foundation for structure-based drug discovery and design. The growing impact of polypharmacology for complex diseases like schizophrenia, cancer etc. warrants the need for novel targets and considering the undiscriminating and selectivity of GPCRs, they can fulfill this purpose. Again, natural genetic variations within the human genome sometimes delude the therapeutic expectations of some drugs, resulting in medication response differences and ADRs (adverse drug reactions). Around ~30 billion US dollars are dumped annually for poor accounting of ADRs in the US alone. To curb such undesirable reactions, the knowledge of established and currently in clinical trials GPCRs families can offer huge understanding towards the drug designing prospects including "off-target" effects reducing economical resource and time. The druggability of GPCR protein families and critical roles played by them in complex diseases are explained. Class A, class B1, class C and class F are generally established family and GPCRs in phase I (19%), phase II(29%), phase III(52%) studies are also reviewed. From the phase I studies, frizzled receptors accounted for the highest in trial targets, neuropeptides in phase II and melanocortin in phase III studies. Also, the bioapplications for nanoparticles along with future prospects for both nanomedicine and GPCR drug industry are discussed. Further, the use of computational techniques and methods employed for different target validations are also reviewed along with their future potential for the GPCR based drug discovery.
Collapse
Affiliation(s)
- Surovi Saikia
- Natural Products Chemistry Group, CSIR North East Institute of Science & Technology, Jorhat-785006, Assam, India
| | - Manobjyoti Bordoloi
- Natural Products Chemistry Group, CSIR North East Institute of Science & Technology, Jorhat-785006, Assam, India
| | - Rajeev Sarmah
- Allied Health Sciences, Assam Down Town University, Panikhaiti, Guwahati 781026, Assam, India
| |
Collapse
|
21
|
Krzyczmonik A, Keller T, López-Picón FR, Forsback S, Kirjavainen AK, Takkinen JS, Wasilewska A, Scheinin M, Haaparanta-Solin M, Sączewski F, Solin O. Radiosynthesis and Preclinical Evaluation of an α 2A-Adrenoceptor Tracer Candidate, 6-[ 18F]Fluoro-marsanidine. Mol Imaging Biol 2020; 21:879-887. [PMID: 30710261 DOI: 10.1007/s11307-019-01317-6] [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] [Indexed: 10/27/2022]
Abstract
PURPOSE The α2-adrenoceptors mediate many effects of norepinephrine and epinephrine, and participate in the regulation of neuronal, endocrine, cardiovascular, vegetative, and metabolic functions. Of the three receptor subtypes, only α2A and α2C are found in the brain in significant amounts. Subtype-selective positron emission tomography (PET) imaging of α2-adrenoceptors has been limited to the α2C subtype. Here, we report the synthesis of 6-[18F]fluoro-marsanidine, a subtype-selective PET tracer candidate for α2A-adrenoceptors, and its preclinical evaluation in rats and mice. PROCEDURES 6-[18F]Fluoro-marsanidine was synthesized using electrophilic F-18 fluorination with [18F]Selectfluor bis(triflate). The tracer was evaluated in Sprague Dawley rats and in α2A-knockout (KO) and wild-type (WT) mice for subtype selectivity. In vivo PET imaging and ex vivo brain autoradiography were performed to determine the tracer distribution in the brain. The specificity of the tracer for the target was determined by pretreatment with the subtype-non-selective α2-agonist medetomidine. The peripheral biodistribution and extent of metabolism of 6-[18F]fluoro-marsanidine were also analyzed. RESULTS 6-[18F]Fluoro-marsanidine was synthesized with [18F]Selectfluor bis(triflate) in a radiochemical yield of 6.4 ± 1.7 %. The molar activity was 3.1 to 26.6 GBq/μmol, and the radiochemical purity was > 99 %. In vivo studies in mice revealed lower uptake in the brains of α2A-KO mice compared to WT mice. The results for selectivity were confirmed by ex vivo brain autoradiography. Blocking studies revealed reduced uptake in α2A-adrenoceptor-rich brain regions in pretreated animals, demonstrating the specificity of the tracer. Metabolite analyses revealed very rapid metabolism of 6-[18F]fluoro-marsanidine with blood-brain barrier-permeable metabolites in both rats and mice. CONCLUSION 6-[18F]Fluoro-marsanidine was synthesized and evaluated as a PET tracer candidate for brain α2A-adrenoceptors. However, rapid metabolism, extensive presence of labeled metabolites in the brain, and high non-specific uptake in mouse and rat brain make 6-[18F]fluoro-marsanidine unsuitable for α2A-adrenoceptor targeting in rodents in vivo.
Collapse
Affiliation(s)
- Anna Krzyczmonik
- Radiopharmaceutical Chemistry Laboratory, Turku PET Centre, University of Turku, Turku, Finland
| | - Thomas Keller
- Radiopharmaceutical Chemistry Laboratory, Turku PET Centre, University of Turku, Turku, Finland
| | - Francisco R López-Picón
- PET Preclinical Imaging Laboratory, Turku PET Centre, University of Turku, Turku, Finland.,MediCity Research Laboratory, University of Turku, Turku, Finland
| | - Sarita Forsback
- Radiopharmaceutical Chemistry Laboratory, Turku PET Centre, University of Turku, Turku, Finland.,Department of Chemistry, University of Turku, Turku, Finland
| | - Anna K Kirjavainen
- Radiopharmaceutical Chemistry Laboratory, Turku PET Centre, University of Turku, Turku, Finland
| | - Jatta S Takkinen
- PET Preclinical Imaging Laboratory, Turku PET Centre, University of Turku, Turku, Finland.,MediCity Research Laboratory, University of Turku, Turku, Finland
| | - Aleksandra Wasilewska
- Department of Chemical Technology of Drugs, Faculty of Pharmacy, Medical University of Gdańsk, Gdańsk, Poland
| | - Mika Scheinin
- Institute of Biomedicine, University of Turku, and Unit of Clinical Pharmacology, Turku University Hospital, Turku, Finland
| | - Merja Haaparanta-Solin
- PET Preclinical Imaging Laboratory, Turku PET Centre, University of Turku, Turku, Finland.,MediCity Research Laboratory, University of Turku, Turku, Finland
| | - Franciszek Sączewski
- Department of Chemical Technology of Drugs, Faculty of Pharmacy, Medical University of Gdańsk, Gdańsk, Poland
| | - Olof Solin
- Radiopharmaceutical Chemistry Laboratory, Turku PET Centre, University of Turku, Turku, Finland. .,Department of Chemistry, University of Turku, Turku, Finland. .,Accelerator Laboratory, Turku PET Centre, Åbo Akademi University, Kiinamyllynkatu 4-8, FI-20520, Turku, Finland.
| |
Collapse
|
22
|
Wu Z, Wang T, Li L, Hui Y, Zhang Q, Yuan H. Activation and blockade of α 2-adrenoceptors in the prelimbic cortex regulate anxiety-like behaviors in hemiparkinsonian rats. Biochem Biophys Res Commun 2019; 519:697-704. [PMID: 31542234 DOI: 10.1016/j.bbrc.2019.09.038] [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] [Received: 08/19/2019] [Accepted: 09/11/2019] [Indexed: 11/28/2022]
Abstract
At present, whether α2-adrenoceptors in the prelimbic cortex (PrL) are involved in Parkinson's disease-related anxiety is unclear. We examined the effects of PrL α2-adrenoceptors on anxiety-like behaviors in rats with unilateral 6-hydroxydopamine lesions of the medial forebrain bundle. Compared to the sham operation, the lesion induced anxiety-like responses as measured by the open field test and elevated plus-maze test. Intra-PrL injection of the α2-adrenoceptor agonist clonidine (1.25, 2.5 or 5 μg/rat) produced anxiolytic effects in sham-operated and lesioned rats. Furthermore, intra-PrL injection of the α2-adrenoceptor antagonist idazoxan (1, 2 or 4 μg/rat) induced anxiogenic effects in two groups of rats. The effective doses produced by clonidine and idazoxan in lesioned rats were higher than those in sham-operated rats. Neurochemical results showed that intra-PrL injection of clonidine (5 μg/rat) or idazoxan (4 μg/rat) decreased or increased dopamine (DA) and noradrenaline (NA) and serotonin (5-HT) levels in the medial prefrontal cortex (mPFC) and amygdala in sham-operated and lesioned rats, respectively. These results suggest that α2-adrenoceptors in the PrL are involved in the regulation of anxiety-like behaviors, which is attributable to changes in DA, NA and 5-HT levels in the mPFC and amygdala after activation and blockade of α2-adrenoceptors.
Collapse
Affiliation(s)
- Zhongheng Wu
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Tao Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Libo Li
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Yanping Hui
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Qiaojun Zhang
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Haifeng Yuan
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
23
|
Effect of Stimulation of α 2-Adrenergic Receptors on Action Potential of Working Cardiomyocytes in Rat Atrium. Bull Exp Biol Med 2019; 167:603-605. [PMID: 31606802 DOI: 10.1007/s10517-019-04579-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Indexed: 10/25/2022]
Abstract
The study examined the effect of clonidine hydrochloride (10-9-10-5 М) on electrical activity of working cardiomyocytes in rat right atrium. Stimulation of α2-adrenergic receptor with clonidine changed electrical activity of the heart. All tested concentrations of the agonist lengthened the action potential and decreased the firing rate of cardiomyocytes in a dose-dependent manner. The maximum effects of clonidine were observed at concentration of 10-5 М.
Collapse
|
24
|
Xue BB, Chen BH, Tang YN, Weng CW, Lin LN. Dexmedetomidine protects against lung injury induced by limb ischemia-reperfusion via the TLR4/MyD88/NF-κB pathway. Kaohsiung J Med Sci 2019; 35:672-678. [PMID: 31373750 DOI: 10.1002/kjm2.12115] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 07/10/2019] [Indexed: 12/15/2022] Open
Abstract
Dexmedetomidine (DEX) can protect the lung from ischemia-reperfusion (I/R) injury, but the underlying mechanisms are not fully understood. The aims of this study were to determine whether DEX attenuates lung injury following lower extremity I/R and to investigate the related toll-like receptor 4 (TLR4) signaling pathway. Twenty-eight SD rats were divided into four groups (n = 7): Sham, I/R, I/R + DEX (25 μg/kg prior to ischemia), and I/R + DEX + Atip (250 μg/kg atipamezole before DEX treatment). Lower extremity I/R was induced by left femoral artery clamping for 3 hours and followed by 2 hours reperfusion. Quantitative alveolar damage and the wet/dry (W/D) ratio were calculated. Interleukin (IL)-1, IL-6, and tumor necrosis factor (TNF)-α in the bronchoalveolar lavage fluid (BALF) and serum and myeloperoxidase (MPO) in the lung were measured. The TLR4 and MyD88 mRNA expression levels were measured by RT-PCR, nuclear factor (NF)-κB, and phosphorylated NF-κB by western blot, respectively. Quantitative alveolar damage, W/D ratio, MPO, BALF and serum IL-1, IL-6, and TNF-α, and TLR4, MyD88, NF-κB, and p-NF-κB expression significantly increased in the I/R group relative to the Sham group. DEX preconditioning significantly reduced lung edema, and histological injury relative to the I/R group. Serum and BALF IL-1, IL-6, and TNF-α levels, MPO activity and TLR4, MyD88, NF-κB, and p-NF-κB expression were also significantly reduced in the I/R + DEX group compared with the I/R group. Atipamezole partially reversed all the aforementioned effects. DEX preconditioning protects the lungs against lower extremity I/R injury via α2-adrenoceptor-dependent and α2-adrenoceptor-independent mechanisms. It also suppresses the TLR4 pathway and reduces inflammation.
Collapse
Affiliation(s)
- Bin-Bin Xue
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Bai-Hui Chen
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ya-Ning Tang
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Cheng-Wei Weng
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Li-Na Lin
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
25
|
Spay C, Albares M, Lio G, Thobois S, Broussolle E, Lau B, Ballanger B, Boulinguez P. Clonidine modulates the activity of the subthalamic-supplementary motor loop: evidence from a pharmacological study combining deep brain stimulation and electroencephalography recordings in Parkinsonian patients. J Neurochem 2019; 146:333-347. [PMID: 29675956 DOI: 10.1111/jnc.14447] [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: 01/18/2018] [Revised: 03/15/2018] [Accepted: 04/04/2018] [Indexed: 12/24/2022]
Abstract
Clonidine is an anti-hypertensive medication which acts as an alpha-adrenergic receptor agonist. As the noradrenergic system is likely to support cognitive functions including attention and executive control, other clinical uses of clonidine have recently gained popularity for the treatment of neuropsychiatric disorders like attention-deficit hyperactivity disorder or Tourette syndrome, but the mechanism of action is still unclear. Here, we test the hypothesis that the noradrenergic system regulates the activity of subthalamo-motor cortical loops, and that this influence can be modulated by clonidine. We used pharmacological manipulation of clonidine in a placebo-controlled study in combination with subthalamic nucleus-deep brain stimulation (STN-DBS) in 16 Parkinson's disease patients performing a reaction time task requiring to refrain from reacting (proactive inhibition). We recorded electroencephalographical activity of the whole cortex, and applied spectral analyses directly at the source level after advanced blind source separation. We found only one cortical source localized to the supplementary motor area (SMA) that supported an interaction of pharmacological and subthalamic stimulation. Under placebo, STN-DBS reduced proactive alpha power in the SMA, a marker of local inhibitory activity. This effect was associated with the speeding-up of movement initiation. Clonidine substantially increased proactive alpha power from the SMA source, and canceled out the benefits of STN-DBS on movement initiation. These results provide the first direct neural evidence in humans that the tonic inhibitory activity of the subthalamocortical loops underlying the control of movement initiation is coupled to the noradrenergic system, and that this activity can be targeted by pharmacological agents acting on alpha-adrenergic receptors.
Collapse
Affiliation(s)
- Charlotte Spay
- Université de Lyon, Lyon, France.,Université Lyon 1, Villeurbanne, France.,INSERM, U 1028, Lyon Neuroscience Research Center, Lyon, France.,CNRS, UMR 5292, Lyon Neuroscience Research Center, Lyon, France
| | - Marion Albares
- Université de Lyon, Lyon, France.,Université Lyon 1, Villeurbanne, France.,CNRS, UMR 5229, Institut des Sciences Cognitives Marc Jeannerod, Bron, France.,Sorbonne Universités, UPMC Université Pierre et Marie Curie Paris 06, UMR 7225, Paris, France.,INSERM UMR 1127, Institut du cerveau et de la moelle épinière, ICM, Paris, France.,CNRS, UMR 7225, Institut du cerveau et de la moelle épinière, ICM, Paris, France
| | - Guillaume Lio
- Université de Lyon, Lyon, France.,Université Lyon 1, Villeurbanne, France.,CNRS, UMR 5229, Institut des Sciences Cognitives Marc Jeannerod, Bron, France
| | - Stephane Thobois
- Université de Lyon, Lyon, France.,Université Lyon 1, Villeurbanne, France.,CNRS, UMR 5229, Institut des Sciences Cognitives Marc Jeannerod, Bron, France.,Hospices civils de Lyon, hôpital neurologique Pierre Wertheimer, Bron, France
| | - Emmanuel Broussolle
- Université de Lyon, Lyon, France.,Université Lyon 1, Villeurbanne, France.,CNRS, UMR 5229, Institut des Sciences Cognitives Marc Jeannerod, Bron, France.,Hospices civils de Lyon, hôpital neurologique Pierre Wertheimer, Bron, France
| | - Brian Lau
- Sorbonne Universités, UPMC Université Pierre et Marie Curie Paris 06, UMR 7225, Paris, France.,INSERM UMR 1127, Institut du cerveau et de la moelle épinière, ICM, Paris, France.,CNRS, UMR 7225, Institut du cerveau et de la moelle épinière, ICM, Paris, France
| | - Benedicte Ballanger
- Université de Lyon, Lyon, France.,Université Lyon 1, Villeurbanne, France.,INSERM, U 1028, Lyon Neuroscience Research Center, Lyon, France.,CNRS, UMR 5292, Lyon Neuroscience Research Center, Lyon, France
| | - Philippe Boulinguez
- Université de Lyon, Lyon, France.,Université Lyon 1, Villeurbanne, France.,INSERM, U 1028, Lyon Neuroscience Research Center, Lyon, France.,CNRS, UMR 5292, Lyon Neuroscience Research Center, Lyon, France
| |
Collapse
|
26
|
Bahari Z, Meftahi GH. Spinal α 2 -adrenoceptors and neuropathic pain modulation; therapeutic target. Br J Pharmacol 2019; 176:2366-2381. [PMID: 30657594 DOI: 10.1111/bph.14580] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 11/23/2018] [Accepted: 12/04/2018] [Indexed: 12/22/2022] Open
Abstract
Neuropathic pain can arise from disease or damage to the nervous system. The most common symptoms of neuropathic pain include spontaneous pain, allodynia, and hyperalgesia. There is still limited knowledge about the factors that initiate and maintain neuropathic pain. However, ample evidence has proved the antinociceptive role of spinal α-adrenoceptors following nerve injury. It is well-documented that noradrenergic descending pathways from supraspinal loci exert an inhibitory influence on the spinal cord nociceptive neurons, mostly through the activation of spinal α2 -adrenoceptors. This, in turn, suppresses transmission of pain input and the hyperexcitability of spinal dorsal horn neurons. There is considerable evidence demonstrating that spinal application of α2 -adrenoceptor agonists leads to analgesic effects in animal models of neuropathic pain. Today, despite the recent rapid development of neuroscience and drug discovery, effective drugs with clear basic mechanisms have remained a mystery. Here, we give an overview of the cellular mechanisms through which brainstem adrenergic descending inhibitory processing can alter spinal pain transmission to the higher centres, and how these pathways change in neuropathic pain conditions focusing on the role of spinal α2 -adrenoceptors in the spinal dorsal horn. We then suggest that α2 -adrenoceptor agonist may be useful to treat neuropathic pain. 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)
- Zahra Bahari
- Neuroscience Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran.,Department of Physiology and Medical Physics, Faculty of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
27
|
Carr BJ, Mihara K, Ramachandran R, Saifeddine M, Nathanson NM, Stell WK, Hollenberg MD. Myopia-Inhibiting Concentrations of Muscarinic Receptor Antagonists Block Activation of Alpha2A-Adrenoceptors In Vitro. Invest Ophthalmol Vis Sci 2019; 59:2778-2791. [PMID: 29860464 DOI: 10.1167/iovs.17-22562] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Myopia is a refractive disorder that degrades vision. It can be treated with atropine, a muscarinic acetylcholine receptor (mAChR) antagonist, but the mechanism is unknown. Atropine may block α-adrenoceptors at concentrations ≥0.1 mM, and another potent myopia-inhibiting ligand, mamba toxin-3 (MT3), binds equally well to human mAChR M4 and α1A- and α2A-adrenoceptors. We hypothesized that mAChR antagonists could inhibit myopia via α2A-adrenoceptors, rather than mAChR M4. Methods Human mAChR M4 (M4), chicken mAChR M4 (cM4), or human α2A-adrenergic receptor (hADRA2A) clones were cotransfected with CRE/promoter-luciferase (CRE-Luc; agonist-induced luminescence) and Renilla luciferase (RLuc; normalizing control) into human cells. Inhibition of normalized agonist-induced luminescence by antagonists (ATR: atropine; MT3; HIM: himbacine; PRZ: pirenzepine; TRP: tropicamide; OXY: oxyphenonium; QNB: 3-quinuclidinyl benzilate; DIC: dicyclomine; MEP: mepenzolate) was measured using the Dual-Glo Luciferase Assay System. Results Relative inhibitory potencies of mAChR antagonists at mAChR M4/cM4, from most to least potent, were QNB > OXY ≥ ATR > MEP > HIM > DIC > PRZ > TRP. MT3 was 56× less potent at cM4 than at M4. Relative potencies of mAChR antagonists at hADRA2A, from most to least potent, were MT3 > HIM > ATR > OXY > PRZ > TRP > QNB > MEP; DIC did not antagonize. Conclusions Muscarinic antagonists block hADRA2A signaling at concentrations comparable to those used to inhibit chick myopia (≥0.1 mM) in vivo. Relative potencies at hADRA2A, but not M4/cM4, correlate with reported abilities to inhibit chick form-deprivation myopia. mAChR antagonists might inhibit myopia via α2-adrenoceptors, instead of through the mAChR M4/cM4 receptor subtype.
Collapse
Affiliation(s)
- Brittany J Carr
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Inflammation Research Network-Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Department of Neuroscience, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Koichiro Mihara
- Inflammation Research Network-Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Rithwik Ramachandran
- Inflammation Research Network-Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Department of Physiology and Pharmacology, Western University, London, Ontario, Canada
| | - Mahmoud Saifeddine
- Inflammation Research Network-Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Neil M Nathanson
- Department of Pharmacology, University of Washington, Seattle, Washington, United States
| | - William K Stell
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Morley D Hollenberg
- Inflammation Research Network-Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
28
|
Zurawski Z, Thompson Gray AD, Brady LJ, Page B, Church E, Harris NA, Dohn MR, Yim YY, Hyde K, Mortlock DP, Jones CK, Winder DG, Alford S, Hamm HE. Disabling the Gβγ-SNARE interaction disrupts GPCR-mediated presynaptic inhibition, leading to physiological and behavioral phenotypes. Sci Signal 2019; 12:12/569/eaat8595. [PMID: 30783011 DOI: 10.1126/scisignal.aat8595] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
G protein-coupled receptors (GPCRs) that couple to Gi/o proteins modulate neurotransmission presynaptically by inhibiting exocytosis. Release of Gβγ subunits from activated G proteins decreases the activity of voltage-gated Ca2+ channels (VGCCs), decreasing excitability. A less understood Gβγ-mediated mechanism downstream of Ca2+ entry is the binding of Gβγ to SNARE complexes, which facilitate the fusion of vesicles with the cell plasma membrane in exocytosis. Here, we generated mice expressing a form of the SNARE protein SNAP25 with premature truncation of the C terminus and that were therefore partially deficient in this interaction. SNAP25Δ3 homozygote mice exhibited normal presynaptic inhibition by GABAB receptors, which inhibit VGCCs, but defective presynaptic inhibition by receptors that work directly on the SNARE complex, such as 5-hydroxytryptamine (serotonin) 5-HT1b receptors and adrenergic α2a receptors. Simultaneously stimulating receptors that act through both mechanisms showed synergistic inhibitory effects. SNAP25Δ3 homozygote mice had various behavioral phenotypes, including increased stress-induced hyperthermia, defective spatial learning, impaired gait, and supraspinal nociception. These data suggest that the inhibition of exocytosis by Gi/o-coupled GPCRs through the Gβγ-SNARE interaction is a crucial component of numerous physiological and behavioral processes.
Collapse
Affiliation(s)
- Zack Zurawski
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.,Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | | | - Lillian J Brady
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| | - Brian Page
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Emily Church
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Nicholas A Harris
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | - Michael R Dohn
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| | - Yun Young Yim
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| | - Karren Hyde
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| | - Douglas P Mortlock
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | - Carrie K Jones
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Danny G Winder
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | - Simon Alford
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Heidi E Hamm
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.
| |
Collapse
|
29
|
The in vivo specificity of synaptic Gβ and Gγ subunits to the α 2a adrenergic receptor at CNS synapses. Sci Rep 2019; 9:1718. [PMID: 30737458 PMCID: PMC6368627 DOI: 10.1038/s41598-018-37222-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 11/26/2018] [Indexed: 11/21/2022] Open
Abstract
G proteins are major transducers of signals from G-protein coupled receptors (GPCRs). They are made up of α, β, and γ subunits, with 16 Gα, 5 Gβ and 12 Gγ subunits. Though much is known about the specificity of Gα subunits, the specificity of Gβγs activated by a given GPCR and that activate each effector in vivo is not known. Here, we examined the in vivo Gβγ specificity of presynaptic α2a-adrenergic receptors (α2aARs) in both adrenergic (auto-α2aARs) and non-adrenergic neurons (hetero-α2aARs) for the first time. With a quantitative MRM proteomic analysis of neuronal Gβ and Gγ subunits, and co-immunoprecipitation of tagged α2aARs from mouse models including transgenic FLAG-α2aARs and knock-in HA-α2aARs, we investigated the in vivo specificity of Gβ and Gγ subunits to auto-α2aARs and hetero-α2aARs activated with epinephrine to understand the role of Gβγ specificity in diverse physiological functions such as anesthetic sparing, and working memory enhancement. We detected Gβ2, Gγ2, Gγ3, and Gγ4 with activated auto α2aARs, whereas we found Gβ4 and Gγ12 preferentially interacted with activated hetero-α2aARs. Further understanding of in vivo Gβγ specificity to various GPCRs offers new insights into the multiplicity of genes for Gβ and Gγ, and the mechanisms underlying GPCR signaling through Gβγ subunits.
Collapse
|
30
|
Szabadi E. Functional Organization of the Sympathetic Pathways Controlling the Pupil: Light-Inhibited and Light-Stimulated Pathways. Front Neurol 2018; 9:1069. [PMID: 30619035 PMCID: PMC6305320 DOI: 10.3389/fneur.2018.01069] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 11/23/2018] [Indexed: 11/13/2022] Open
Abstract
Pupil dilation is mediated by a sympathetic output acting in opposition to parasympathetically mediated pupil constriction. While light stimulates the parasympathetic output, giving rise to the light reflex, it can both inhibit and stimulate the sympathetic output. Light-inhibited sympathetic pathways originate in retina-receptive neurones of the pretectum and the suprachiasmatic nucleus (SCN): by attenuating sympathetic activity, they allow unimpeded operation of the light reflex. Light stimulates the noradrenergic and serotonergic pathways. The hub of the noradrenergic pathway is the locus coeruleus (LC) containing both excitatory sympathetic premotor neurones (SympPN) projecting to preganglionic neurones in the spinal cord, and inhibitory parasympathetic premotor neurones (ParaPN) projecting to preganglionic neurones in the Edinger-Westphal nucleus (EWN). SympPN receive inputs from the SCN via the dorsomedial hypothalamus, orexinergic neurones of the latero-posterior hypothalamus, wake- and sleep-promoting neurones of the hypothalamus and brain stem, nociceptive collaterals of the spinothalamic tract, whereas ParaPN receive inputs from the amygdala, sleep/arousal network, nociceptive spinothalamic collaterals. The activity of LC neurones is regulated by inhibitory α2-adrenoceptors. There is a species difference in the function of the preautonomic LC. In diurnal animals, the α2-adrenoceptor agonist clonidine stimulates mainly autoreceptors on SymPN, causing miosis, whereas in nocturnal animals it stimulates postsynaptic α2-arenoceptors in the EWN, causing mydriasis. Noxious stimulation activates SympPN in diurnal animals and ParaPN in nocturnal animals, leading to pupil dilation via sympathoexcitation and parasympathetic inhibition, respectively. These differences may be attributed to increased activity of excitatory LC neurones due to stimulation by light in diurnal animals. This may also underlie the wake-promoting effect of light in diurnal animals, in contrast to its sleep-promoting effect in nocturnal species. The hub of the serotonergic pathway is the dorsal raphe nucleus that is light-sensitive, both directly and indirectly (via an orexinergic input). The light-stimulated pathways mediate a latent mydriatic effect of light on the pupil that can be unmasked by drugs that either inhibit or stimulate SympPN in these pathways. The noradrenergic pathway has widespread connections to neural networks controlling a variety of functions, such as sleep/arousal, pain, and fear/anxiety. Many physiological and psychological variables modulate pupil function via this pathway.
Collapse
Affiliation(s)
- Elemer Szabadi
- Developmental Psychiatry, Queen's Medical Centre, University of Nottingham, Nottingham, United Kingdom
| |
Collapse
|
31
|
Oh DJ, Chen JL, Vajaranant TS, Dikopf MS. Brimonidine tartrate for the treatment of glaucoma. Expert Opin Pharmacother 2018; 20:115-122. [DOI: 10.1080/14656566.2018.1544241] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Daniel J. Oh
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, IL, USA
| | - Judy L. Chen
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, IL, USA
| | - Thasarat S. Vajaranant
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, IL, USA
| | - Mark S. Dikopf
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
32
|
Maaliki D, Issa K, Al Shehabi T, El-Yazbi A, Eid AH. The role of α2-adrenergic receptors in hypertensive preeclampsia: A hypothesis. Microcirculation 2018; 26:e12511. [PMID: 30383326 DOI: 10.1111/micc.12511] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 10/18/2018] [Accepted: 10/29/2018] [Indexed: 02/06/2023]
Abstract
Preeclampsia, a major disorder of human pregnancy, manifests as persistent hypertension and proteinuria presenting after 20 weeks of pregnancy. Multiple systemic symptoms might be associated with preeclampsia including thrombocytopenia, liver impairment, pulmonary edema, and cerebral disturbances. However, vascular dysfunction remains the core pathological driver of preeclampsia. Defective placental implantation followed by dysfunctional placental spiral artery development promotes a hypoxic environment. Massive endothelial dysfunction characterized by reduced vasodilation, augmented vasoconstriction, and increased vascular permeability and inflammation ensues. Interestingly, the same signaling and inflammatory pathways implicated in preeclampsia appear to be shared with other vascular disorders involving alteration of α2 -AR function. The role of α2 -ARs in the regulation of microcirculatory function has long been recognized, thus raising the question of whether they are involved in the pathogenesis of vascular dysfunction in preeclampsia. Here, we review possible interplay between signaling and inflammatory pathways common to preeclampsia and α2 -AR function/regulation. We speculate on the potential contribution of these receptors to the observed phenotype and the potential role for their pharmacological modulators as therapeutic interventions with preeclampsia.
Collapse
Affiliation(s)
- Dina Maaliki
- Department of Pharmacology and Toxicology, American University of Beirut, Beirut, Lebanon
| | - Khodr Issa
- Department of Pharmacology and Toxicology, American University of Beirut, Beirut, Lebanon
| | - Tuqa Al Shehabi
- Department of Biological and Environmental Sciences, Qatar University, Doha, Qatar
| | - Ahmed El-Yazbi
- Department of Pharmacology and Toxicology, American University of Beirut, Beirut, Lebanon.,Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Ali H Eid
- Department of Pharmacology and Toxicology, American University of Beirut, Beirut, Lebanon.,Department of Biological and Environmental Sciences, Qatar University, Doha, Qatar.,Department of Biomedical Sciences, Qatar University, Doha, Qatar
| |
Collapse
|
33
|
Human Placenta Expresses α 2-Adrenergic Receptors and May Be Implicated in Pathogenesis of Preeclampsia and Fetal Growth Restriction. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:2774-2785. [PMID: 30273604 DOI: 10.1016/j.ajpath.2018.08.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 08/07/2018] [Accepted: 08/23/2018] [Indexed: 02/02/2023]
Abstract
α2-Adrenergic receptors (α2ARs) are G-protein-coupled receptors involved in catecholamine signaling by extracellular regulated protein kinase 1 and 2 (ERK1/2) pathways. We examined placental expression and function of α2AR subtypes in women with severe preeclampsia (sPE) with and without intrauterine growth restriction (IUGR). Placental biopsies were analyzed from 52 women with i) sPE (n = 8); ii) sPE + IUGR (n = 9); iii) idiopathic IUGR (n = 8); iv) idiopathic preterm birth (n = 16); and v) healthy term controls (n = 11). Expression of α2AR subtypes (α2A, α2B, α2C) and phospho-ERK1/2 (receptor activation marker) was investigated by immunohistochemistry and/or quantitative real-time RT-PCR. The effects of α2CAR knockdown on syncytialization (syncytin-1 and -2) and β-human chorionic gonadotropin secretion were examined in BeWo cells stimulated with forskolin. The effects of α2AR agonist UK 14,304 and specific α2CAR antagonist were tested, using a trophoblast migration assay. All three α2ARs were expressed and functionally active in human placenta with site-specific localization. Highest α2BAR and α2CAR mRNA expression was identified in sPE + IUGR. α2CAR knockdown increased expression of syncytin-1 and -2 but decreased secretion of β-human chorionic gonadotropin. UK 14,304 impaired trophoblast migration. The observed α2AR expression pattern suggests different function for each subtype. α2CAR modulates trophoblast syncytialization and migration and may carry pathogenic role in sPE + IUGR.
Collapse
|
34
|
Effect of α 2-Adrenoceptor Stimulation on Functional Parameters of Langendorff-Isolated Rat Heart. Bull Exp Biol Med 2018; 165:593-596. [PMID: 30242585 DOI: 10.1007/s10517-018-4220-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Indexed: 10/28/2022]
Abstract
We studied the effect of α2-adrenoreceptor agonist clonidine hydrochloride in concentrations of 10-9-10-6 M on inotropy, chronotropy, and coronary flow in Langendorff-isolated heart of adult rats. It was found that α2-adrenoreceptor agonist changed all studied parameters. Left ventricular myocardium contraction force decreased after application of all tested concentrations, the maximum effect was observed at a concentration of 10-6 M. Stimulation of α2-adrenergic receptors in concentrations of 10-8, 10-7, and 10-6 M produced a two-phase effect (initial increase and a subsequent decrease) on the coronary flow. Clonidine hydrochloride in the maximum concentration (10-6 M) caused a decrease in HR in one group and an increase in the other.
Collapse
|
35
|
Martins GM, Teixeira MBCG, Silva MV, Neofiti-Papi B, Miranda-Rodrigues M, Brum PC, Gouveia CHA. Global Disruption of α2A Adrenoceptor Barely Affects Bone Tissue but Minimizes the Detrimental Effects of Thyrotoxicosis on Cortical Bone. Front Endocrinol (Lausanne) 2018; 9:486. [PMID: 30233491 PMCID: PMC6127616 DOI: 10.3389/fendo.2018.00486] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Accepted: 08/06/2018] [Indexed: 01/02/2023] Open
Abstract
Evidence shows that sympathetic nervous system (SNS) activation inhibits bone formation and activates bone resorption leading to bone loss. Because thyroid hormone (TH) interacts with the SNS to control several physiological processes, we raised the hypothesis that this interaction also controls bone remodeling. We have previously shown that mice with double-gene inactivation of α2A- and -adrenoceptors (α2A/2C-AR-/-) present high bone mass (HBM) phenotype and resistance to thyrotoxicosis-induced osteopenia, which supports a TH-SNS interaction to control bone mass and suggests that it involves α2-AR signaling. Accordingly, we detected expression of α2A-AR, α2B-AR and α2C-AR in the skeleton, and that triiodothyronine (T3) modulates α2C-AR mRNA expression in the bone. Later, we found that mice with single-gene inactivation of α2C-AR (α2C-AR-/-) present low bone mass in the femur and HBM in the vertebra, but that both skeletal sites are resistant to TH-induce osteopenia, showing that the SNS actions occur in a skeletal site-dependent manner, and that thyrotoxicosis depends on α2C-AR signaling to promote bone loss. To further dissect the specific roles of α2-AR subtypes, in this study, we evaluated the skeletal phenotype of mice with single-gene inactivation of α2A-AR (α2A-AR-/-), and the effect of daily treatment with a supraphysiological dose of T3, for 4 or 12 weeks, on bone microarchitecture and bone resistance to fracture. Micro-computed tomographic (μCT) analysis revealed normal trabecular and cortical bone structure in the femur and vertebra of euthyroid α2A-AR-/- mice. Thyrotoxicosis was more detrimental to femoral trabecular bone in α2A-AR-/- than in WT mice, whereas this bone compartment had been previously shown to present resistance to thyrotoxicosis in α2C-AR-/- mice. Altogether these findings reveal that TH excess depends on α2C-AR signaling to negatively affect femoral trabecular bone. In contrast, thyrotoxicosis was more deleterious to femoral and vertebral cortical bone in WT than in α2A-AR-/- mice, suggesting that α2A-AR signaling contributes to TH actions on cortical bone. These findings further support a TH-SNS interaction to control bone physiology, and suggest that α2A-AR and α2C-AR signaling pathways have key roles in the mechanisms through which thyrotoxicosis promotes its detrimental effects on bone remodeling, structure and resistance to fracture.
Collapse
Affiliation(s)
- Gisele M. Martins
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Department of Morphology, Federal University of Espírito Santo, Vitória, Brazil
| | | | - Marcos V. Silva
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Department of Morphology, Federal University of Sergipe, Aracaju, Brazil
| | - Bianca Neofiti-Papi
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Manuela Miranda-Rodrigues
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- University of Western Ontario, London, ON, Canada
| | - Patricia C. Brum
- School of Physical Education and Sport, University of São Paulo, São Paulo, Brazil
| | - Cecilia H. A. Gouveia
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- School of Medicine, University of São Paulo, São Paulo, Brazil
- *Correspondence: Cecilia H. A. Gouveia
| |
Collapse
|
36
|
Spadari RC, Cavadas C, de Carvalho AETS, Ortolani D, de Moura AL, Vassalo PF. Role of Beta-adrenergic Receptors and Sirtuin Signaling in the Heart During Aging, Heart Failure, and Adaptation to Stress. Cell Mol Neurobiol 2018; 38:109-120. [PMID: 29063982 DOI: 10.1007/s10571-017-0557-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Accepted: 10/06/2017] [Indexed: 01/03/2023]
Abstract
In the heart, catecholamine effects occur by activation of beta-adrenergic receptors (β-ARs), mainly the beta 1 (β1-AR) and beta 2 (β2-AR) subtypes, both of which couple to the Gs protein that activates the adenylyl cyclase signaling pathway. The β2-ARs can also couple to the Gi protein that counterbalances the effect of the Gs protein on cyclic adenosine monophosphate production and activates the phosphatidylinositol 3-kinase (PI3K)-Akt signaling pathway. In several cardiovascular disorders, including heart failure, as well as in aging and in animal models of environmental stress, a reduction in the β1/β2-AR ratio and activation of the β2-AR-Gi-PI3K-Akt signaling pathway have been observed. Recent studies have shown that sirtuins modulate certain organic processes, including the cellular stress response, through activation of the PI3K-Akt signaling pathway and of downstream molecules such as p53, Akt, HIF1-α, and nuclear factor-kappa B. In the heart, SIRT1, SIRT3, and β2-ARs are crucial to the regulation of the cardiomyocyte energy metabolism, oxidative stress, reactive oxygen species production, and autophagy. SIRT1 and the β2-AR-Gi complex also control signaling pathways of cell survival and death. Here, we review the role played by β2-ARs and sirtuins during aging, heart failure, and adaptation to stress, focusing on the putative interplay between the two. That relationship, if proven, merits further investigation in the context of cardiac function and dysfunction.
Collapse
Affiliation(s)
- Regina Celia Spadari
- Laboratory of Stress Biology, Department of Biosciences, Campus Baixada Santista, Universidade Federal de São Paulo (UNIFESP), Santos, Brazil.
- Departamento de Biociências / Campus Baixada Santista, UNIFESP, Rua Silva Jardim 136, Santos, SP, 11015-020, Brazil.
| | - Claudia Cavadas
- Center for Neurosciences and Cell Biology (CNC) and School of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Ana Elisa T Saturi de Carvalho
- Laboratory of Stress Biology, Department of Biosciences, Campus Baixada Santista, Universidade Federal de São Paulo (UNIFESP), Santos, Brazil
| | - Daniela Ortolani
- Laboratory of Stress Biology, Department of Biosciences, Campus Baixada Santista, Universidade Federal de São Paulo (UNIFESP), Santos, Brazil
- Department of Physiological Science, Universidade Federal Do Espírito Santo (HUCAM, UFES), Vitória, Brazil
| | - Andre Luiz de Moura
- Laboratory of Stress Biology, Department of Biosciences, Campus Baixada Santista, Universidade Federal de São Paulo (UNIFESP), Santos, Brazil
| | - Paula Frizera Vassalo
- Department of Physiological Science, Universidade Federal Do Espírito Santo (HUCAM, UFES), Vitória, Brazil
- University Hospital Cassiano Antônio de Moraes, Universidade Federal Do Espírito Santo (HUCAM, UFES), Vitória, Brazil
| |
Collapse
|
37
|
Sudo RT, do Amaral RV, Monteiro CEDS, Pitta IDR, Lima MDC, Montes GC, Ririe DG, Hayashida K, Zapata-Sudo G. Antinociception induced by a novel α 2A adrenergic receptor agonist in rodents acute and chronic pain models. Eur J Pharmacol 2017; 815:210-218. [PMID: 28935564 DOI: 10.1016/j.ejphar.2017.09.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 09/06/2017] [Accepted: 09/14/2017] [Indexed: 01/05/2023]
Abstract
The mechanisms and antinociceptive effects of a novel α2A adrenoceptor agonist, 3-(2-chloro-6-fluorobenzil)-imidazolinide-2,4-dione (PT-31) were investigated using animal models of acute and chronic pain. The effects of PT-31 on pain responses were examined using hot plate and formalin tests in mice and spinal nerve ligation (SNL)-induced hyperalgesia in rats. The effects of antagonists acting on α adrenoceptor were assessed to investigate the interaction of these pathways upon PT-31 induced antinociception. PT-31 effects on motor activity/skills and on hemodynamic parameters were also evaluated. PT-31 had dose-dependent antinociception effects on hot-plate and formalin-injection induced pain responses. Thermal hyperalgesia and mechanical allodynia were reduced following a 7 d treatment with PT-31 (1, 5, and 10mg/kg/d, p.o.), and those effects were attenuated by yohimbine (5mg/kg), atropine (2mg/kg), L-nitro arginine methyl ester (L-NAME; 30mg/kg), or naloxone (2mg/kg). In contrast to clonidine, PT-31 did not have locomotor or hemodynamic effects in rats. The present results suggest that PT-31 represents a candidate for pain treatment with advantages over clonidine, namely no locomotor or hemodynamic impairments.
Collapse
Affiliation(s)
- Roberto Takashi Sudo
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, RJ, Brazil.
| | - Rachel Vieiralves do Amaral
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, RJ, Brazil
| | - Carlos Eduardo da Silva Monteiro
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, RJ, Brazil
| | - Ivan da Rocha Pitta
- Núcleo de Pesquisa em Inovação Terapêutica, Universidade Federal de Pernambuco, PE, Brazil
| | - Maria do Carmo Lima
- Núcleo de Pesquisa em Inovação Terapêutica, Universidade Federal de Pernambuco, PE, Brazil
| | - Guilherme Carneiro Montes
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, RJ, Brazil
| | - Douglas Gordon Ririe
- Department of Anesthesiology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Kenichiro Hayashida
- Department of Neurophysiology, Akita University School of Medicine, Akita, Japan
| | - Gisele Zapata-Sudo
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, RJ, Brazil
| |
Collapse
|
38
|
Artalejo AR, Olivos-Oré LA. Alpha2-adrenoceptors in adrenomedullary chromaffin cells: functional role and pathophysiological implications. Pflugers Arch 2017; 470:61-66. [PMID: 28836008 DOI: 10.1007/s00424-017-2059-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Revised: 08/12/2017] [Accepted: 08/15/2017] [Indexed: 12/20/2022]
Abstract
Chromaffin cells from the adrenal medulla participate in stress responses by releasing catecholamines into the bloodstream. Main control of adrenal catecholamine secretion is exerted both neurally (by the splanchnic nerve fibers) and humorally (by corticosteroids, circulating noradrenaline, etc.). It should be noted, however, that secretory products themselves (catecholamines, ATP, opioids, ascorbic acid, chromogranins) could also influence the secretory response in an autocrine/paracrine manner. This form of control is activity-dependent and can be either inhibitory or excitatory. Among the inhibitory influences, it stands out the one mediated by α2-adrenergic autoreceptors activated by released catecholamines. α2-adrenoceptors are G protein-coupled receptors capable to inhibit exocytotic secretion through a direct interaction of Gβγ subunits with voltage-gated Ca2+ channels. Interestingly, upon intense and/or prolonged stimulation, α2-adrenergic receptors become desensitized by the intervention of G protein-coupled receptor kinase 2 (GRK2). In several experimental models of heart failure, there has been reported the up-regulation of GRK2 and the loss of functioning of inhibitory α2-adrenoceptors resulting in enhanced release of adrenomedullary catecholamines. Given the importance of circulating catecholamines in the pathophysiology of heart failure, the recovery of α2-adrenergic modulation of the secretory response from chromaffin cells appears as a novel strategy for a better control of the patients with this cardiac disease.
Collapse
Affiliation(s)
- Antonio R Artalejo
- Institute for Research in Neurochemistry & Department of Toxicology and Pharmacology, Faculty of Veterinary, Universidad Complutense de Madrid, Avda. Puerta de Hierro s/n, 28029, Madrid, Spain.
| | - Luis Alcides Olivos-Oré
- Institute for Research in Neurochemistry & Department of Toxicology and Pharmacology, Faculty of Veterinary, Universidad Complutense de Madrid, Avda. Puerta de Hierro s/n, 28029, Madrid, Spain
| |
Collapse
|
39
|
Huhtinen A, Hongisto V, Laiho A, Löyttyniemi E, Pijnenburg D, Scheinin M. Gene expression profiles and signaling mechanisms in α 2B-adrenoceptor-evoked proliferation of vascular smooth muscle cells. BMC SYSTEMS BIOLOGY 2017; 11:65. [PMID: 28659168 PMCID: PMC5490158 DOI: 10.1186/s12918-017-0439-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 06/09/2017] [Indexed: 12/31/2022]
Abstract
BACKGROUND α2-adrenoceptors are important regulators of vascular tone and blood pressure. Regulation of cell proliferation is a less well investigated consequence of α2-adrenoceptor activation. We have previously shown that α2B-adrenoceptor activation stimulates proliferation of vascular smooth muscle cells (VSMCs). This may be important for blood vessel development and plasticity and for the pathology and therapeutics of cardiovascular disorders. The underlying cellular mechanisms have remained mostly unknown. This study explored pathways of regulation of gene expression and intracellular signaling related to α2B-adrenoceptor-evoked VSMC proliferation. RESULTS The cellular mechanisms and signaling pathways of α2B-adrenoceptor-evoked proliferation of VSMCs are complex and include redundancy. Functional enrichment analysis and pathway analysis identified differentially expressed genes associated with α2B-adrenoceptor-regulated VSMC proliferation. They included the upregulated genes Egr1, F3, Ptgs2 and Serpine1 and the downregulated genes Cx3cl1, Cav1, Rhoa, Nppb and Prrx1. The most highly upregulated gene, Lypd8, represents a novel finding in the VSMC context. Inhibitor library screening and kinase activity profiling were applied to identify kinases in the involved signaling pathways. Putative upstream kinases identified by two different screens included PKC, Raf-1, Src, the MAP kinases p38 and JNK and the receptor tyrosine kinases EGFR and HGF/HGFR. As a novel finding, the Src family kinase Lyn was also identified as a putative upstream kinase. CONCLUSIONS α2B-adrenoceptors may mediate their pro-proliferative effects in VSMCs by promoting the activity of bFGF and PDGF and the growth factor receptors EGFR, HGFR and VEGFR-1/2. The Src family kinase Lyn was also identified as a putative upstream kinase. Lyn is known to be expressed in VSMCs and has been identified as an important regulator of GPCR trafficking and GPCR effects on cell proliferation. Identified Ser/Thr kinases included several PKC isoforms and the β-adrenoceptor kinases 1 and 2. Cross-talk between the signaling mechanisms involved in α2B-adrenoceptor-evoked VSMC proliferation thus appears to involve PKC activation, subsequent changes in gene expression, transactivation of EGFR, and modulation of kinase activities and growth factor-mediated signaling. While many of the identified individual signals were relatively small in terms of effect size, many of them were validated by combining pathway analysis and our integrated screening approach.
Collapse
Affiliation(s)
- Anna Huhtinen
- Department of Pharmacology, Drug Development and Therapeutics, Institute of Biomedicine, University of Turku, Kiinamyllynkatu 10, FI-20520 Turku, Finland
- Unit of Clinical Pharmacology, Turku University Hospital, Turku, Finland
| | - Vesa Hongisto
- Toxicology Division, Misvik Biology Oy, Turku, Finland
| | - Asta Laiho
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Eliisa Löyttyniemi
- Department of Biostatistics, Department of Clinical Medicine, University of Turku, Turku, Finland
| | - Dirk Pijnenburg
- PamGene International BV, Wolvenhoek 10, 5211HH s’Hertogenbosch, The Netherlands
| | - Mika Scheinin
- Department of Pharmacology, Drug Development and Therapeutics, Institute of Biomedicine, University of Turku, Kiinamyllynkatu 10, FI-20520 Turku, Finland
- Unit of Clinical Pharmacology, Turku University Hospital, Turku, Finland
| |
Collapse
|
40
|
Zhang F, Gannon M, Chen Y, Zhou L, Jiao K, Wang Q. The amyloid precursor protein modulates α 2A-adrenergic receptor endocytosis and signaling through disrupting arrestin 3 recruitment. FASEB J 2017. [PMID: 28646018 DOI: 10.1096/fj.201700346r] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The amyloid precursor protein (APP) has long been appreciated for its role in Alzheimer's disease (AD) pathology. However, less is known about the physiologic function of APP outside of AD. Particularly, whether and how APP may regulate functions of cell surface receptors, including GPCRs, remains largely unclear. In this study, we identified a novel direct interaction between APP and the α2A-adrenergic receptor (α2AAR) that occurs at the intracellular domains of both proteins. The APP interaction with α2AAR is promoted by agonist stimulation and competes with arrestin 3 binding to the receptor. Consequently, the presence of APP attenuates α2AAR internalization and desensitization, which are arrestin-dependent processes. Furthermore, in neuroblastoma neuro-2A cells and primary superior cervical ganglion neurons, where APP is highly expressed, the lack of APP leads to a dramatic increase in plasma membrane recruitment of endogenous arrestin 3 following α2AAR activation. Concomitantly, agonist-induced internalization of α2AAR is significantly enhanced in these neuronal cells. Our study provided the first evidence that APP fine tunes GPCR signaling and trafficking. Given the important role of α2AAR in controlling norepinephrine release and response, this novel regulation of α2AAR by APP may have an impact on modulation of noradrenergic activity and sympathetic tone.-Zhang, F., Gannon, M., Chen, Y., Zhou, L., Jiao, K., Wang, Q. The amyloid precursor protein modulates α2A-adrenergic receptor endocytosis and signaling through disrupting arrestin 3 recruitment.
Collapse
Affiliation(s)
- Fang Zhang
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Mary Gannon
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Yunjia Chen
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Lufang Zhou
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Kai Jiao
- Department of Genetics, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Qin Wang
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA;
| |
Collapse
|
41
|
Dexmedetomidine preconditioning may attenuate myocardial ischemia/reperfusion injury by down-regulating the HMGB1-TLR4-MyD88-NF-кB signaling pathway. PLoS One 2017; 12:e0172006. [PMID: 28222157 PMCID: PMC5319750 DOI: 10.1371/journal.pone.0172006] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 01/30/2017] [Indexed: 12/21/2022] Open
Abstract
Aims To investigate whether dexmedetomidine (DEX) preconditioning could alleviate the inflammation caused by myocardial ischemia/reperfusion (I/R) injury by reducing HMGB1-TLR4-MyD88-NF-кB signaling. Methods Seventy rats were randomly assigned into five groups: sham group, myocardial I/R group (I/R), DEX+I/R group (DEX), DEX+yohimbine+I/R group (DEX/YOH), and yohimbine+I/R group (YOH). Animals were subjected to 30 min of ischemia induced by occluding the left anterior descending artery followed by 120 min of reperfusion. Myocardial infarct size and histological scores were evaluated. The levels of IL-6 and TNF-α in serum and myocardium were quantified by enzyme-linked immunosorbent assay, and expression of HMGB1, TLR4, MyD88, IκB and NF-κB in the myocardial I/R area were determined with Western blot and immunocytochemistry. Results Myocardial infarct sizes, histological scores, levels of circulating and myocardial IL-6 and TNF-α, the expression of HMGB1, TLR4, MyD88 and NF-κB, and the degradation of IκB were significantly increased in the I/R group compared with the sham group (P<0.01). DEX preconditioning significantly reduced the myocardial infarct size and histological scores (P<0.01 vs. I/R group). Similarly, the serum and myocardial levels of IL-6 and TNF-α, the expression of HMGB1, TLR4, MyD88 and NF-κB, and the degradation of IκB were significantly reduced in the DEX group (P<0.01 vs. I/R group). These effects were partly reversed by yohimbine, a selective α2-adrenergic receptor antagonist, while yohimbine alone had no significant effect on any of the above indicators. Conclusion DEX preconditioning reduces myocardial I/R injury in part by attenuating inflammation, which may be attributed to the downregulation of the HMGB1-TLR4-MyD88-NF-кB signaling pathway mediated by the α2-adrenergic receptor activation.
Collapse
|
42
|
Zinc in the Monoaminergic Theory of Depression: Its Relationship to Neural Plasticity. Neural Plast 2017; 2017:3682752. [PMID: 28299207 PMCID: PMC5337390 DOI: 10.1155/2017/3682752] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 01/24/2017] [Indexed: 12/21/2022] Open
Abstract
Preclinical and clinical studies have demonstrated that zinc possesses antidepressant properties and that it may augment the therapy with conventional, that is, monoamine-based, antidepressants. In this review we aim to discuss the role of zinc in the pathophysiology and treatment of depression with regard to the monoamine hypothesis of the disease. Particular attention will be paid to the recently described zinc-sensing GPR39 receptor as well as aspects of zinc deficiency. Furthermore, an attempt will be made to give a possible explanation of the mechanisms by which zinc interacts with the monoamine system in the context of depression and neural plasticity.
Collapse
|
43
|
Uys MM, Shahid M, Harvey BH. Therapeutic Potential of Selectively Targeting the α 2C-Adrenoceptor in Cognition, Depression, and Schizophrenia-New Developments and Future Perspective. Front Psychiatry 2017; 8:144. [PMID: 28855875 PMCID: PMC5558054 DOI: 10.3389/fpsyt.2017.00144] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 07/24/2017] [Indexed: 12/12/2022] Open
Abstract
α2A- and α2C-adrenoceptors (ARs) are the primary α2-AR subtypes involved in central nervous system (CNS) function. These receptors are implicated in the pathophysiology of psychiatric illness, particularly those associated with affective, psychotic, and cognitive symptoms. Indeed, non-selective α2-AR blockade is proposed to contribute toward antidepressant (e.g., mirtazapine) and atypical antipsychotic (e.g., clozapine) drug action. Both α2C- and α2A-AR share autoreceptor functions to exert negative feedback control on noradrenaline (NA) release, with α2C-AR heteroreceptors regulating non-noradrenergic transmission (e.g., serotonin, dopamine). While the α2A-AR is widely distributed throughout the CNS, α2C-AR expression is more restricted, suggesting the possibility of significant differences in how these two receptor subtypes modulate regional neurotransmission. However, the α2C-AR plays a more prominent role during states of low endogenous NA activity, while the α2A-AR is relatively more engaged during states of high noradrenergic tone. Although augmentation of conventional antidepressant and antipsychotic therapy with non-selective α2-AR antagonists may improve therapeutic outcome, animal studies report distinct yet often opposing roles for the α2A- and α2C-ARs on behavioral markers of mood and cognition, implying that non-selective α2-AR antagonism may compromise therapeutic utility both in terms of efficacy and side-effect liability. Recently, several highly selective α2C-AR antagonists have been identified that have allowed deeper investigation into the function and utility of the α2C-AR. ORM-13070 is a useful positron emission tomography ligand, ORM-10921 has demonstrated antipsychotic, antidepressant, and pro-cognitive actions in animals, while ORM-12741 is in clinical development for the treatment of cognitive dysfunction and neuropsychiatric symptoms in Alzheimer's disease. This review will emphasize the importance and relevance of the α2C-AR as a neuropsychiatric drug target in major depression, schizophrenia, and associated cognitive deficits. In addition, we will present new prospects and future directions of investigation.
Collapse
Affiliation(s)
- Madeleine Monique Uys
- Division of Pharmacology, Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom, South Africa
| | | | - Brian Herbert Harvey
- Division of Pharmacology, Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom, South Africa
| |
Collapse
|
44
|
Chronic postnatal monoamine oxidase inhibition affects affiliative behavior in rat pupso. Pharmacol Biochem Behav 2016; 153:60-68. [PMID: 27989747 DOI: 10.1016/j.pbb.2016.12.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 12/12/2016] [Accepted: 12/14/2016] [Indexed: 12/29/2022]
Abstract
Monoamine neurotransmitters serotonin (5-HT), dopamine (DA), and noradrenaline (NA) act as important modulators of mammalian brain development and represent neurobiological substrates of affiliative behavior reflected in rat pups as a tendency to huddle or produce ultrasonic vocalizations (USV) when separated from the nest. Monoamines are metabolized through oxidative deamination catalyzed by the mitochondrial enzyme monoamine oxidase (MAO). In this study, we examined the consequences of postnatal MAO inhibition on affiliative behavior in rat pups. Pups received daily injections of either an irreversible non-selective MAO inhibitor tranylcypromine (TCP) or saline, from post-natal day (PND) 1 to PND 22. Quantitative and qualitative components of USV were analyzed on PNDs 10, 13 and 16 in order to determine the level of separation-induced anxiety and the modality of vocal communication. In comparison to control pups, TCP-treated pups displayed higher cortical 5-HT, DA and NA levels, higher peripheral 5-HT concentration, lower body mass throughout the pre-weaning period, higher isolation-induced drop in body temperature, and reduced total number of calls. Furthermore, they produced lower pitched calls of longer average duration without a preferable waveform. Our results demonstrate that chronic MAO inhibition by TCP primarily affects 5-HT concentrations, but also raises central catecholamine levels. They further indicate that disturbed monoaminergic homeostasis during early postnatal development leads to decreased weight-gain, compromised thermoregulation, and altered affiliative behavior in pre-weaning pups as reflected in reduced separation anxiety and inadequate vocal communication. Finally, they suggest a need for thorough examination of the potential effects of TCP and other monoamine inhibitors on the developing human brain.
Collapse
|
45
|
Multidisciplinary Consideration of Potential Pathophysiologic Mechanisms of Paradoxical Erythema with Topical Brimonidine Therapy. Adv Ther 2016; 33:1885-1895. [PMID: 27562835 PMCID: PMC5083782 DOI: 10.1007/s12325-016-0404-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Indexed: 01/14/2023]
Abstract
Rosacea is a chronic inflammatory disease with transient and non-transient redness as key characteristics. Brimonidine is a selective α2-adrenergic receptor (AR) agonist approved for persistent facial erythema of rosacea based on significant efficacy and good safety data. The majority of patients treated with brimonidine report a benefit; however, there have been sporadic reports of worsening erythema after the initial response. A group of dermatologists, receptor physiology, and neuroimmunology scientists met to explore potential mechanisms contributing to side effects as well as differences in efficacy. We propose the following could contribute to erythema after application: (1) local inflammation and perivascular inflammatory cells with abnormally functioning ARs may lead to vasodilatation; (2) abnormal saturation and cells expressing different AR subtypes with varying ligand affinity; (3) barrier dysfunction and increased skin concentrations of brimonidine with increased actions at endothelial and presynaptic receptors, resulting in increased vasodilation; and (4) genetic predisposition and receptor polymorphism(s) leading to different smooth muscle responses. Approximately 80% of patients treated with brimonidine experience a significant improvement without erythema worsening as an adverse event. Attention to optimizing skin barrier function, setting patient expectations, and strategies to minimize potential problems may possibly reduce further the number of patients who experience side effects. FUNDING Galderma International S.A.S., Paris, France.
Collapse
|
46
|
Kornicka A, Wasilewska A, Sączewski J, Hudson AL, Boblewski K, Lehmann A, Gzella K, Belka M, Sączewski F, Gdaniec M, Rybczyńska A, Bączek T. 1-[(Imidazolidin-2-yl)imino]-1H-indoles as new hypotensive agents: synthesis andin vitroandin vivobiological studies. Chem Biol Drug Des 2016; 89:400-410. [DOI: 10.1111/cbdd.12846] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 07/13/2016] [Accepted: 08/19/2016] [Indexed: 11/28/2022]
Affiliation(s)
- Anita Kornicka
- Department of Chemical Technology of Drugs; Medical University of Gdańsk; Gdańsk Poland
| | - Aleksandra Wasilewska
- Department of Chemical Technology of Drugs; Medical University of Gdańsk; Gdańsk Poland
| | - Jarosław Sączewski
- Department of Organic Chemistry; Medical University of Gdańsk; Gdańsk Poland
| | - Alan L. Hudson
- Department of Pharmacology; University of Alberta; Edmonton Canada
| | - Konrad Boblewski
- Department of Pathophysiology; Medical University of Gdańsk; Gdańsk Poland
| | - Artur Lehmann
- Department of Pathophysiology; Medical University of Gdańsk; Gdańsk Poland
| | - Karol Gzella
- Department of Chemical Technology of Drugs; Medical University of Gdańsk; Gdańsk Poland
| | - Mariusz Belka
- Department of Pharmaceutical Chemistry; Medical University of Gdańsk; Gdańsk Poland
| | - Franciszek Sączewski
- Department of Chemical Technology of Drugs; Medical University of Gdańsk; Gdańsk Poland
| | - Maria Gdaniec
- Faculty of Chemistry; A. Mickiewicz University; Poznań Poland
| | | | - Tomasz Bączek
- Department of Pharmaceutical Chemistry; Medical University of Gdańsk; Gdańsk Poland
| |
Collapse
|
47
|
Hara M, Zhou ZY, Hemmings HC. α2-Adrenergic Receptor and Isoflurane Modulation of Presynaptic Ca2+ Influx and Exocytosis in Hippocampal Neurons. Anesthesiology 2016; 125:535-46. [PMID: 27337223 PMCID: PMC4988866 DOI: 10.1097/aln.0000000000001213] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Evidence indicates that the anesthetic-sparing effects of α2-adrenergic receptor (AR) agonists involve α2A-AR heteroreceptors on nonadrenergic neurons. Since volatile anesthetics inhibit neurotransmitter release by reducing synaptic vesicle (SV) exocytosis, the authors hypothesized that α2-AR agonists inhibit nonadrenergic SV exocytosis and thereby potentiate presynaptic inhibition of exocytosis by isoflurane. METHODS Quantitative imaging of fluorescent biosensors of action potential-evoked SV exocytosis (synaptophysin-pHluorin) and Ca influx (GCaMP6) were used to characterize presynaptic actions of the clinically used α2-AR agonists dexmedetomidine and clonidine, and their interaction with isoflurane, in cultured rat hippocampal neurons. RESULTS Dexmedetomidine (0.1 μM, n = 10) or clonidine (0.5 μM, n = 8) inhibited action potential-evoked exocytosis (54 ± 5% and 59 ± 8% of control, respectively; P < 0.001). Effects on exocytosis were blocked by the subtype-nonselective α2-AR antagonist atipamezole or the α2A-AR-selective antagonist BRL 44408 but not by the α2C-AR-selective antagonist JP 1302. Dexmedetomidine inhibited exocytosis and presynaptic Ca influx without affecting Ca coupling to exocytosis, consistent with an effect upstream of Ca-exocytosis coupling. Exocytosis coupled to both N-type and P/Q-type Ca channels was inhibited by dexmedetomidine or clonidine. Dexmedetomidine potentiated inhibition of exocytosis by 0.7 mM isoflurane (to 42 ± 5%, compared to 63 ± 8% for isoflurane alone; P < 0.05). CONCLUSIONS Hippocampal SV exocytosis is inhibited by α2A-AR activation in proportion to reduced Ca entry. These effects are additive with those of isoflurane, consistent with a role for α2A-AR presynaptic heteroreceptor inhibition of nonadrenergic synaptic transmission in the anesthetic-sparing effects of α2A-AR agonists.
Collapse
Affiliation(s)
- Masato Hara
- Department of Anesthesiology, Weill Cornell Medical College, New York, NY 10065, USA
- Department of Anesthesiology, Kurume University School of Medicine, Kurume, Fukuoka 830-0011, Japan
| | - Zhen-Yu Zhou
- Department of Anesthesiology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Hugh C. Hemmings
- Department of Anesthesiology, Weill Cornell Medical College, New York, NY 10065, USA
- Department of Pharmacology, Weill Cornell Medical College, New York, NY 10065, USA
| |
Collapse
|
48
|
Wehrwein EA, Orer HS, Barman SM. Overview of the Anatomy, Physiology, and Pharmacology of the Autonomic Nervous System. Compr Physiol 2016; 6:1239-78. [PMID: 27347892 DOI: 10.1002/cphy.c150037] [Citation(s) in RCA: 235] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Comprised of the sympathetic nervous system, parasympathetic nervous system, and enteric nervous system, the autonomic nervous system (ANS) provides the neural control of all parts of the body except for skeletal muscles. The ANS has the major responsibility to ensure that the physiological integrity of cells, tissues, and organs throughout the entire body is maintained (homeostasis) in the face of perturbations exerted by both the external and internal environments. Many commonly prescribed drugs, over-the-counter drugs, toxins, and toxicants function by altering transmission within the ANS. Autonomic dysfunction is a signature of many neurological diseases or disorders. Despite the physiological relevance of the ANS, most neuroscience textbooks offer very limited coverage of this portion of the nervous system. This review article provides both historical and current information about the anatomy, physiology, and pharmacology of the sympathetic and parasympathetic divisions of the ANS. The ultimate aim is for this article to be a valuable resource for those interested in learning the basics of these two components of the ANS and to appreciate its importance in both health and disease. Other resources should be consulted for a thorough understanding of the third division of the ANS, the enteric nervous system. © 2016 American Physiological Society. Compr Physiol 6:1239-1278, 2016.
Collapse
Affiliation(s)
- Erica A Wehrwein
- Department of Physiology, Michigan State University, East Lansing, Michigan, USA
| | - Hakan S Orer
- Department of Pharmacology, Koc University School of Medicine, Istanbul, Turkey
| | - Susan M Barman
- Department of Pharmacology &Toxicology, Michigan State University, East Lansing, Michigan, USA
| |
Collapse
|
49
|
Pytka K, Podkowa K, Rapacz A, Podkowa A, Żmudzka E, Olczyk A, Sapa J, Filipek B. The role of serotonergic, adrenergic and dopaminergic receptors in antidepressant-like effect. Pharmacol Rep 2015; 68:263-74. [PMID: 26922526 DOI: 10.1016/j.pharep.2015.08.007] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 07/29/2015] [Accepted: 08/12/2015] [Indexed: 01/11/2023]
Abstract
Depression is a serious global illness, becoming more and more common in developed countries. Because of specific symptoms it is considered as a leading cause of disability all over the world with a high death factor due to suicides. There are many antidepressants used in the therapy, but still more than 30% of patients do not respond to the treatment. The heterogeneous nature of the illness and its complex, unclear aetiology may be responsible for these difficulties. Next to the main monoaminergic hypothesis of depression there are also many other approaches connected with the pathophysiology of the disease, including hypothalamic-pituitary-adrenal axis dysregulation, dopaminergic, cholinergic, glutamatergic or GABA-ergic neurotransmission. Nevertheless, it can be unambiguously stated that serotonergic, noradrenergic and dopaminergic systems are precisely connected with pathogenesis of depression, and should be therefore considered as valuable targets in patients' treatment. Bearing that in mind, this review presents the role of serotonergic, adrenergic and dopaminergic receptors in antidepressant-like effect.
Collapse
Affiliation(s)
- Karolina Pytka
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Kraków, Poland.
| | - Karolina Podkowa
- Department of Neurobiology, Institute of Pharmacology Polish Academy of Sciences, Kraków, Poland
| | - Anna Rapacz
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Kraków, Poland
| | - Adrian Podkowa
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Kraków, Poland
| | - Elżbieta Żmudzka
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Kraków, Poland
| | - Adrian Olczyk
- Institute of Automatic Control, Silesian University of Technology, Gliwice, Poland
| | - Jacek Sapa
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Kraków, Poland
| | - Barbara Filipek
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Kraków, Poland
| |
Collapse
|
50
|
Yaksh TL, Woller SA, Ramachandran R, Sorkin LS. The search for novel analgesics: targets and mechanisms. F1000PRIME REPORTS 2015; 7:56. [PMID: 26097729 PMCID: PMC4447049 DOI: 10.12703/p7-56] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The management of the pain state is of great therapeutic relevance to virtually every medical specialty. Failure to manage its expression has deleterious consequence to the well-being of the organism. An understanding of the complex biology of the mechanisms underlying the processing of nociceptive information provides an important pathway towards development of novel and robust therapeutics. Importantly, preclinical models have been of considerable use in determining the linkage between mechanism and the associated behaviorally defined pain state. This review seeks to provide an overview of current thinking targeting pain biology, the use of preclinical models and the development of novel pain therapeutics. Issues pertinent to the strengths and weaknesses of current development strategies for analgesics are considered.
Collapse
|