1
|
da Silva SB, Feitosa SGD, de L Alves SM, Santos RCA, Dos Anjos JV, Araújo AV. A Concise and Useful Guide to Understand How Alpha1 Adrenoceptor Antagonists Work. Mini Rev Med Chem 2022; 22:2383-2405. [PMID: 35507746 DOI: 10.2174/1389557522666220504141949] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 02/23/2022] [Accepted: 03/16/2022] [Indexed: 11/22/2022]
Abstract
Adrenoceptors are the receptors for the catecholamines, adrenaline and noradrenaline. They are divided in α (α1 and α2) and β (β1, β2 and β3). α1-Adrenoceptors are subdivided in α1A, α1B and α1D. Most tissues express mixtures of α1-adrenoceptors subtypes, which appear to coexist in different densities and ratios, and in most cases their responses are probably due to the activation of more than one type. The three subtypes of α1-adrenoceptors are G-protein-coupled receptors (GPCR), specifically coupled to Gq/11. Additionally, the activation of these receptors may activate other signaling pathways or different components of these pathways, which leads to a great variety of possible cellular effects. The first clinically used α1 antagonist was Prazosin, for Systemic Arterial Hypertension (SAH). It was followed by its congeners, Terazosin and Doxazosin. Nowadays, there are many classes of α-adrenergic antagonists with different selectivity profiles. In addition to SAH, the α1-adrenoceptors are used for the treatment of Benign Prostatic Hyperplasia (BPH) and urolithiasis. This antagonism may be part of the mechanism of action of tricyclic antidepressants. Moreover, the activation of these receptors may lead to adverse effects such as orthostatic hypotension, similar to what happens with the antidepressants and with some antipsychotic. Structure-activity relationships can explain, in part, how antagonists work and how selective they can be for each one of the subtypes. However, it is necessary to develop new molecules which antagonize the α1-adrenoceptors or make chemical modifications in these molecules to improve the selectivity, pharmacokinetic profile and/or reduce the adverse effects of known drugs.
Collapse
Affiliation(s)
- Sidiane B da Silva
- Laboratório de Nutrição, Atividade Física e Plasticidade Fenotípica - Centro Acadêmico de Vitória - Universidade Federal de Pernambuco. R. Alto do Reservatório, s/n Bela Vista - Vitória de Santo Antão, PE, 50608-680, Brazil
| | - Sidney G D Feitosa
- Departamento de Química Fundamental - Universidade Federal de Pernambuco. Av. Jornalista Aníbal Fernandes, s/n, Cidade Universitária - Recife, PE, 50740-560, Brazil
| | - Silvia M de L Alves
- Laboratório de Nutrição, Atividade Física e Plasticidade Fenotípica - Centro Acadêmico de Vitória - Universidade Federal de Pernambuco. R. Alto do Reservatório, s/n Bela Vista - Vitória de Santo Antão, PE, 50608-680, Brazil
| | - Ruth C A Santos
- Laboratório de Nutrição, Atividade Física e Plasticidade Fenotípica - Centro Acadêmico de Vitória - Universidade Federal de Pernambuco. R. Alto do Reservatório, s/n Bela Vista - Vitória de Santo Antão, PE, 50608-680, Brazil
| | - Janaína V Dos Anjos
- Departamento de Química Fundamental - Universidade Federal de Pernambuco. Av. Jornalista Aníbal Fernandes, s/n, Cidade Universitária - Recife, PE, 50740-560, Brazil
| | - Alice V Araújo
- Núcleo de Saúde Pública, Centro Acadêmico de Vitória - Universidade Federal de Pernambuco R. Alto do Reservatório, s/n Bela Vista - Vitória de Santo Antão, PE, 50608-680, Brazil
| |
Collapse
|
2
|
Chmielarz P, Kuśmierczyk J, Rafa-Zabłocka K, Chorązka K, Kowalska M, Satała G, Nalepa I. Antidepressants Differentially Regulate Intracellular Signaling from α1-Adrenergic Receptor Subtypes In Vitro. Int J Mol Sci 2021; 22:ijms22094817. [PMID: 34062902 PMCID: PMC8124549 DOI: 10.3390/ijms22094817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/28/2021] [Accepted: 04/29/2021] [Indexed: 11/16/2022] Open
Abstract
Currently utilized antidepressants have limited effectiveness and frequently incur undesired effects. Most antidepressants are thought to act via the inhibition of monoamine reuptake; however, direct binding to monoaminergic receptors has been proposed to contribute to both their clinical effectiveness and their side effects, or lack thereof. Among the target receptors of antidepressants, α1‑adrenergic receptors (ARs) have been implicated in depression etiology, antidepressant action, and side effects. However, differences in the direct effects of antidepressants on signaling from the three subtypes of α1-ARs, namely, α1A-, α1B- and α1D‑ARs, have been little explored. We utilized cell lines overexpressing α1A-, α1B- or α1D-ARs to investigate the effects of the antidepressants imipramine (IMI), desipramine (DMI), mianserin (MIA), reboxetine (REB), citalopram (CIT) and fluoxetine (FLU) on noradrenaline-induced second messenger generation by those receptors. We found similar orders of inhibition at α1A-AR (IMI < DMI < CIT < MIA < REB) and α1D‑AR (IMI = DMI < CIT < MIA), while the α1B-AR subtype was the least engaged subtype and was inhibited with low potency by three drugs (MIA < IMI = DMI). In contrast to their direct antagonistic effects, prolonged incubation with IMI and DMI increased the maximal response of the α1B-AR subtype, and the CIT of both the α1A- and the α1B-ARs. Our data demonstrate a complex, subtype-specific modulation of α1-ARs by antidepressants of different groups.
Collapse
Affiliation(s)
- Piotr Chmielarz
- Department of Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland; (P.C.); (J.K.); (K.R.-Z.); (K.C.); (M.K.)
| | - Justyna Kuśmierczyk
- Department of Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland; (P.C.); (J.K.); (K.R.-Z.); (K.C.); (M.K.)
| | - Katarzyna Rafa-Zabłocka
- Department of Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland; (P.C.); (J.K.); (K.R.-Z.); (K.C.); (M.K.)
| | - Katarzyna Chorązka
- Department of Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland; (P.C.); (J.K.); (K.R.-Z.); (K.C.); (M.K.)
| | - Marta Kowalska
- Department of Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland; (P.C.); (J.K.); (K.R.-Z.); (K.C.); (M.K.)
| | - Grzegorz Satała
- Department of Medicinal Chemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland;
| | - Irena Nalepa
- Department of Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland; (P.C.); (J.K.); (K.R.-Z.); (K.C.); (M.K.)
- Correspondence: ; Tel.: +48-12-6623225
| |
Collapse
|
3
|
Delcourte S, Etievant A, Haddjeri N. Role of central serotonin and noradrenaline interactions in the antidepressants' action: Electrophysiological and neurochemical evidence. PROGRESS IN BRAIN RESEARCH 2021; 259:7-81. [PMID: 33541681 DOI: 10.1016/bs.pbr.2021.01.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The development of antidepressant drugs, in the last 6 decades, has been associated with theories based on a deficiency of serotonin (5-HT) and/or noradrenaline (NA) systems. Although the pathophysiology of major depression (MD) is not fully understood, numerous investigations have suggested that treatments with various classes of antidepressant drugs may lead to an enhanced 5-HT and/or adapted NA neurotransmissions. In this review, particular morpho-physiological aspects of these systems are first considered. Second, principal features of central 5-HT/NA interactions are examined. In this regard, the effects of the acute and sustained antidepressant administrations on these systems are discussed. Finally, future directions including novel therapeutic strategies are proposed.
Collapse
Affiliation(s)
- Sarah Delcourte
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, Bron, France
| | - Adeline Etievant
- Integrative and Clinical Neurosciences EA481, University of Bourgogne Franche-Comté, Besançon, France
| | - Nasser Haddjeri
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, Bron, France.
| |
Collapse
|
4
|
Mahmoodkhani M, Amini M, Derafshpour L, Ghasemi M, Mehranfard N. Negative relationship between brain α 1A-AR neurotransmission and βArr2 levels in anxious adolescent rats subjected to early life stress. Exp Brain Res 2020; 238:2833-2844. [PMID: 33025031 DOI: 10.1007/s00221-020-05937-1] [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: 07/02/2020] [Accepted: 09/27/2020] [Indexed: 01/06/2023]
Abstract
Early-life stress is correlated with the development of anxiety-related behavior in adolescence, but underlying mechanisms remain poorly known. The α1A-adrenergic receptor (AR) is linked to mood regulation and its function is assumed to be regulated by β-arrestins (βArrs) via desensitization and downregulation. Here, we investigated correlation between changes in α1A-AR and βArr2 levels in the prefrontal cortex (PFC) and hippocampus of adolescent and adult male rats subjected to maternal separation (MS) and their relationship with anxiety-like behavior in adolescence. MS was performed 3 h per day from postnatal days 2-11 and anxiety-like behavior was evaluated in the elevated plus-maze and open field tests. The protein levels were examined using western blot assay. MS decreased α1A-AR expression and increased βArr2 expression in both brain regions of adolescent rats, while induced reverse changes in adulthood. MS adolescent rats demonstrated higher anxiety-type behavior and lower activity in behavioral tests than controls. Decreased α1A-AR levels in MS adolescence strongly correlated with reduced time spent in the open field central area, consistent with increased anxiety-like behavior. An anxiety-like phenotype was mimicked by acute and chronic treatment of developing rats with prazosin, an α1A-AR antagonist, suggesting α1A-AR downregulation may facilitate anxiety behavior in MS adolescent rats. Together, our results indicate a negative correlation between α1A-AR neurotransmission and βArr2 levels in both adults and anxious-adolescent rats and suggest that increased βArr2 levels may contribute to posttranslational regulation of α1A-AR and modulation of anxiety-like behavior in adolescent rats. This may provide a path to develop more effective anxiolytic treatments.
Collapse
Affiliation(s)
- Maryam Mahmoodkhani
- Neurophysiology Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Mohammad Amini
- Neurophysiology Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Leila Derafshpour
- Neurophysiology Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Maedeh Ghasemi
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nasrin Mehranfard
- Neurophysiology Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
5
|
Wang YJ, Liu L, Wang Y, Wang JL, Gao TT, Wang H, Chen TT, Guan W, Jiang B. Imipramine exerts antidepressant-like effects in chronic stress models of depression by promoting CRTC1 expression in the mPFC. Brain Res Bull 2020; 164:257-268. [PMID: 32905805 DOI: 10.1016/j.brainresbull.2020.08.028] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 08/24/2020] [Accepted: 08/26/2020] [Indexed: 10/23/2022]
Abstract
Recent studies have suggested that CREB-regulated transcription coactivator 1 (CRTC1) plays a role in the pathophysiology of depression. Although imipramine is thought to prevent the reuptake of synaptic serotonin and norepinephrine, its antidepressant-like mechanisms remain elusive. In this study, the effects of imipramine on CRTC1 were studied in several models of depression, including the chronic restraint stress (CRS), chronic unpredictable mild stress (CUMS) and chronic social defeat stress (CSDS) models. We examined whether repeated imipramine administration can reverse the effects of CRS, CUMS and CSDS on CRTC1 expression in both the hippocampus and medial prefrontal cortex (mPFC). Furthermore, genetic knockdown of CRTC1 by CRTC1-shRNA was used to determine whether CRTC1 is necessary for the antidepressant-like effects of imipramine in mice. Our results showed that imipramine reversed the down-regulating effects of CRS, CUMS and CSDS on CRTC1 expression in the mPFC but not the hippocampus, and that CRTC1-shRNA fully abolished the antidepressant-like actions of imipramine in mice. In conclusion, CRTC1 in the mPFC is involved in the antidepressant mechanism of imipramine.
Collapse
Affiliation(s)
- Ying-Jie Wang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu, China; Provincial Key Laboratory of Inflammation and Molecular Drug Target, Jiangsu, China
| | - Ling Liu
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu, China; Provincial Key Laboratory of Inflammation and Molecular Drug Target, Jiangsu, China
| | - Yuan Wang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu, China; Provincial Key Laboratory of Inflammation and Molecular Drug Target, Jiangsu, China
| | - Jin-Liang Wang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu, China; Provincial Key Laboratory of Inflammation and Molecular Drug Target, Jiangsu, China
| | - Ting-Ting Gao
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu, China; Provincial Key Laboratory of Inflammation and Molecular Drug Target, Jiangsu, China
| | - Hao Wang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu, China; Provincial Key Laboratory of Inflammation and Molecular Drug Target, Jiangsu, China
| | - Ting-Ting Chen
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu, China; Provincial Key Laboratory of Inflammation and Molecular Drug Target, Jiangsu, China
| | - Wei Guan
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu, China; Provincial Key Laboratory of Inflammation and Molecular Drug Target, Jiangsu, China
| | - Bo Jiang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu, China; Provincial Key Laboratory of Inflammation and Molecular Drug Target, Jiangsu, China.
| |
Collapse
|
6
|
Stimulation of noradrenergic transmission by reboxetine is beneficial for a mouse model of progressive parkinsonism. Sci Rep 2019; 9:5262. [PMID: 30918302 PMCID: PMC6437187 DOI: 10.1038/s41598-019-41756-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 03/14/2019] [Indexed: 01/23/2023] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder and is characterized by motor deficits such as tremor, rigidity and bradykinesia. These symptoms are directly caused by the loss of dopaminergic neurons. However, a wealth of clinical evidence indicates that the dopaminergic system is not the only system affected in PD. Postmortem studies of brains from PD patients have revealed the degeneration of noradrenergic neurons in the locus coeruleus (LC) to the same or even greater extent than that observed in the dopaminergic neurons of substantia nigra (SN) and ventral tegmental area (VTA). Moreover, studies performed on rodent models suggest that enhancement of noradrenergic transmission may attenuate the PD-like phenotype induced by MPTP administration, a neurotoxin-based PD model. The aim of this study was to investigate whether chronic treatment with either of two compounds targeting the noradrenergic system (reboxetine or atipamezole) possess the ability to reduce the progression of a PD-like phenotype in a novel mouse model of progressive dopaminergic neurodegeneration induced by the genetic inhibition of rRNA synthesis in dopaminergic neurons, mimicking a PD-like phenotype. The results showed that reboxetine improved the parkinsonian phenotype associated with delayed progression of SN/VTA dopaminergic neurodegeneration and higher dopamine content in the striatum. Moreover, the alpha1-adrenergic agonist phenylephrine enhanced survival of TH+ neurons in primary cell cultures, supporting the putative neuroprotective effects of noradrenergic stimulation. Our results provide new insights regarding the possible influence of the noradrenergic system on dopaminergic neuron survival and strongly support the hypothesis regarding the neuroprotective role of noradrenaline.
Collapse
|
7
|
Mesripour A, Sajadian S, Hajhashemi V. Antidepressant-like effect of vitamin B6 in mice forced swimming test and the possible involvement of the noradrenergic system. JOURNAL OF REPORTS IN PHARMACEUTICAL SCIENCES 2019. [DOI: 10.4103/jrptps.jrptps_52_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|
8
|
Dijkstra AA, Lin LC, Nana AL, Gaus SE, Seeley WW. Von Economo Neurons and Fork Cells: A Neurochemical Signature Linked to Monoaminergic Function. Cereb Cortex 2018; 28:131-144. [PMID: 27913432 PMCID: PMC6075576 DOI: 10.1093/cercor/bhw358] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 09/29/2016] [Indexed: 12/13/2022] Open
Abstract
The human anterior cingulate and frontoinsular cortices are distinguished by 2 unique Layer 5 neuronal morphotypes, the von Economo neurons (VENs) and fork cells, whose biological identity remains mysterious. Insights could impact research on diverse neuropsychiatric diseases to which these cells have been linked. Here, we leveraged the Allen Brain Atlas to evaluate mRNA expression of 176 neurotransmitter-related genes and identified vesicular monoamine transporter 2 (VMAT2), gamma-aminobutyric acid (GABA) receptor subunit θ (GABRQ), and adrenoreceptor α-1A (ADRA1A) expression in human VENs, fork cells, and a minority of neighboring Layer 5 neurons. We confirmed these results using immunohistochemistry or in situ hybridization. VMAT2 and GABRQ expression was absent in mouse cerebral cortex. Although VMAT2 is known to package monoamines into synaptic vesicles, in VENs and fork cells its expression occurs in the absence of monoamine-synthesizing enzymes or reuptake transporters. Thus, VENs and fork cells may possess a novel, uncharacterized mode of cortical monoaminergic function that distinguishes them from most other mammalian Layer 5 neurons.
Collapse
Affiliation(s)
- Anke A Dijkstra
- Memory and Aging Center, Department of Neurology, University of California, San Francisco
| | - Li-Chun Lin
- Memory and Aging Center, Department of Neurology, University of California, San Francisco
| | - Alissa L Nana
- Memory and Aging Center, Department of Neurology, University of California, San Francisco
| | - Stephanie E Gaus
- Memory and Aging Center, Department of Neurology, University of California, San Francisco
| | - William W Seeley
- Memory and Aging Center, Department of Neurology, University of California, San Francisco
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
9
|
Atzori M, Cuevas-Olguin R, Esquivel-Rendon E, Garcia-Oscos F, Salgado-Delgado RC, Saderi N, Miranda-Morales M, Treviño M, Pineda JC, Salgado H. Locus Ceruleus Norepinephrine Release: A Central Regulator of CNS Spatio-Temporal Activation? Front Synaptic Neurosci 2016; 8:25. [PMID: 27616990 PMCID: PMC4999448 DOI: 10.3389/fnsyn.2016.00025] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2016] [Accepted: 08/05/2016] [Indexed: 12/22/2022] Open
Abstract
Norepinephrine (NE) is synthesized in the Locus Coeruleus (LC) of the brainstem, from where it is released by axonal varicosities throughout the brain via volume transmission. A wealth of data from clinics and from animal models indicates that this catecholamine coordinates the activity of the central nervous system (CNS) and of the whole organism by modulating cell function in a vast number of brain areas in a coordinated manner. The ubiquity of NE receptors, the daunting number of cerebral areas regulated by the catecholamine, as well as the variety of cellular effects and of their timescales have contributed so far to defeat the attempts to integrate central adrenergic function into a unitary and coherent framework. Since three main families of NE receptors are represented-in order of decreasing affinity for the catecholamine-by: α2 adrenoceptors (α2Rs, high affinity), α1 adrenoceptors (α1Rs, intermediate affinity), and β adrenoceptors (βRs, low affinity), on a pharmacological basis, and on the ground of recent studies on cellular and systemic central noradrenergic effects, we propose that an increase in LC tonic activity promotes the emergence of four global states covering the whole spectrum of brain activation: (1) sleep: virtual absence of NE, (2) quiet wake: activation of α2Rs, (3) active wake/physiological stress: activation of α2- and α1-Rs, (4) distress: activation of α2-, α1-, and β-Rs. We postulate that excess intensity and/or duration of states (3) and (4) may lead to maladaptive plasticity, causing-in turn-a variety of neuropsychiatric illnesses including depression, schizophrenic psychoses, anxiety disorders, and attention deficit. The interplay between tonic and phasic LC activity identified in the LC in relationship with behavioral response is of critical importance in defining the short- and long-term biological mechanisms associated with the basic states postulated for the CNS. While the model has the potential to explain a large number of experimental and clinical findings, a major challenge will be to adapt this hypothesis to integrate the role of other neurotransmitters released during stress in a centralized fashion, like serotonin, acetylcholine, and histamine, as well as those released in a non-centralized fashion, like purines and cytokines.
Collapse
Affiliation(s)
- Marco Atzori
- Neurobiology of Stress Laboratory, Facultad de Ciencias, Universidad Autónoma de San Luis PotosíSan Luis Potosí, Mexico; School for Behavior and Brain Sciences, University of Texas at DallasRichardson, TX, USA
| | - Roberto Cuevas-Olguin
- Neurobiology of Stress Laboratory, Facultad de Ciencias, Universidad Autónoma de San Luis Potosí San Luis Potosí, Mexico
| | - Eric Esquivel-Rendon
- Neurobiology of Stress Laboratory, Facultad de Ciencias, Universidad Autónoma de San Luis Potosí San Luis Potosí, Mexico
| | | | - Roberto C Salgado-Delgado
- Neurobiology of Stress Laboratory, Facultad de Ciencias, Universidad Autónoma de San Luis Potosí San Luis Potosí, Mexico
| | - Nadia Saderi
- Neurobiology of Stress Laboratory, Facultad de Ciencias, Universidad Autónoma de San Luis Potosí San Luis Potosí, Mexico
| | - Marcela Miranda-Morales
- Neurobiology of Stress Laboratory, Facultad de Ciencias, Universidad Autónoma de San Luis Potosí San Luis Potosí, Mexico
| | - Mario Treviño
- Laboratory of Cortical Plasticity and Learning, Universidad de Guadalajara Guadalajara, Mexico
| | - Juan C Pineda
- Electrophysiology Laboratory, Centro de Investigaciones Regionales "Dr. Hideyo Noguchi", Universidad Autónoma de Yucatán Mérida, Mexico
| | - Humberto Salgado
- Electrophysiology Laboratory, Centro de Investigaciones Regionales "Dr. Hideyo Noguchi", Universidad Autónoma de Yucatán Mérida, Mexico
| |
Collapse
|
10
|
A lack of α1A-adrenergic receptor-mediated antidepressant-like effects of S-(+)-niguldipine and B8805-033 in the forced swim test. Behav Pharmacol 2016; 27:397-401. [DOI: 10.1097/fbp.0000000000000204] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
11
|
Zalewska T, Bielawski A, Stanaszek L, Wieczerzak K, Ziemka-Nałęcz M, Nalepa I. Imipramine administration induces changes in the phosphorylation of FAK and PYK2 and modulates signaling pathways related to their activity. Biochim Biophys Acta Gen Subj 2015; 1860:424-33. [PMID: 26620976 DOI: 10.1016/j.bbagen.2015.11.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Revised: 09/26/2015] [Accepted: 11/22/2015] [Indexed: 02/05/2023]
Abstract
BACKGROUND Antidepressants can modify neuronal functioning by affecting many levels of signal transduction pathways that are involved in neuroplasticity. We investigated whether the phosphorylation status of focal adhesion kinase (FAK/PTK2) and its homolog, PYK2/PTK2B, and their complex with the downstream effectors (Src kinase, p130Cas, and paxillin) are affected by administration of the antidepressant drug, imipramine. The treatment influence on the levels of ERK1/2 kinases and their phosphorylated forms (pERK1/2) or the Gαq, Gα11 and Gα12 proteins were also assessed. METHODS Rats were injected with imipramine (10 mg/kg, twice daily) for 21 days. The levels of proteins investigated in their prefrontal cortices were measured by Western blotting. RESULTS Imipramine induced contrasting changes in the phosphorylation of FAK and PYK2 at Tyr397 and Tyr402, respectively. The decreased FAK phosphorylation and increased PYK2 phosphorylation were reflected by changes in the levels of their complex with Src and p130Cas, which was observed predominantly after chronic imipramine treatment. Similarly only chronic imipramine decreased the Gαq expression while Gα11 and Gα12 proteins were untouched. Acute and chronic treatment with imipramine elevated ERK1 and ERK2 total protein levels, whereas only the pERK1 was significantly affected by the drug. CONCLUSION The enhanced activation of PYK2 observed here could function as compensation for FAK inhibition. GENERAL SIGNIFICANCE These data demonstrate that treatment with imipramine, which is a routine in counteracting depressive disorders, enhances the phosphorylation of PYK2, a non-receptor kinase instrumental in promoting synaptic plasticity. This effect documents as yet not considered target in the mechanism of imipramine action.
Collapse
Affiliation(s)
- Teresa Zalewska
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Adam Bielawski
- Department of Brain Biochemistry, Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Luiza Stanaszek
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Krzysztof Wieczerzak
- Department of Brain Biochemistry, Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Małgorzata Ziemka-Nałęcz
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Irena Nalepa
- Department of Brain Biochemistry, Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland.
| |
Collapse
|
12
|
Brief maternal separation affects brain α1-adrenoceptors and apoptotic signaling in adult mice. Prog Neuropsychopharmacol Biol Psychiatry 2014; 48:161-9. [PMID: 24128685 DOI: 10.1016/j.pnpbp.2013.10.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Revised: 09/21/2013] [Accepted: 10/03/2013] [Indexed: 01/24/2023]
Abstract
Exposure to adversity during early life is a risk factor for the development of different mood and psychiatric disorders, including depressive-like behaviors. Here, neonatal mice were temporarily but repeatedly (day 1 to day 13) separated from mothers and placed in a testing environment containing a layer of odorless clean bedding (CB). We assessed in adult animals the impact of this early experience on binding sites and mRNA expression of α1-adrenergic receptor subtypes, heat shock proteins (HSPs) and proapoptotic and antiapoptotic members of the Bcl-2 family proteins in different brain regions involved in processing of olfactory information and rewarding stimuli. We found that repeated exposure to CB experience produced anhedonic-like behavior in terms of reduced saccharin intake and α1-adrenoceptor downregulation in piriform and somatosensory cortices, hippocampus, amygdala and discrete thalamic nuclei. We also found a selective decrease of α1B-adrenoceptor binding sites in the cingulate cortex and hippocampus and an increase of hippocampal α1A and α1B receptor, but not of α1D-adrenoceptor, mRNA levels. Moreover, while a significant decrease of antiapoptotic heat shock proteins Hsp72 and Hsp90 was identified in the prefrontal cortex, a parallel increase of antiapoptotic members of Bcl-2 family proteins was found at the hippocampal level. Together, these data provide evidence that the early exposure to CB experience produced enduring downregulation of α1-adrenoceptors in the prefrontal-limbic forebrain/limbic midbrain network, which plays a key role in the processing of olfactory information and reaction to rewarding stimuli. Finally, these data show that CB experience can "prime" the hippocampal circuitry and promote the expression of antiapoptotic factors that can confer potential neuroprotection to subsequent adversity.
Collapse
|
13
|
Sakata K, Duke SM. Lack of BDNF expression through promoter IV disturbs expression of monoamine genes in the frontal cortex and hippocampus. Neuroscience 2013; 260:265-75. [PMID: 24345476 DOI: 10.1016/j.neuroscience.2013.12.013] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 11/29/2013] [Accepted: 12/06/2013] [Indexed: 02/07/2023]
Abstract
Brain-derived neurotrophic factor (BDNF) is implicated in the pathophysiology of psychiatric conditions including major depression and schizophrenia. Mice lacking activity-driven BDNF expression through promoter IV (knock-in promoter IV: KIV) exhibit depression-like behavior, inflexible learning, and impaired response inhibition. Monoamine systems (serotonin, dopamine, and noradrenaline) are suggested to be involved in depression and schizophrenia since many of the current antidepressants and antipsychotics increase the brain levels of monoamines and/or act on monoamine receptors. To elucidate the impact of activity-driven BDNF on the monoamine systems, we examined mRNA levels for 30 monoamine-related genes, including receptors, transporters, and synthesizing enzymes, in KIV and control wild-type mice by using quantitative reverse-transcription polymerase chain reaction (qRT-PCR). mRNA levels were measured in the frontal cortex and hippocampus, which are regions related to depression and schizophrenia and where promoter IV is active. The frontal cortex of KIV mice showed reduced levels of mRNA expression for serotonin receptors 1b, 2a, and 5b (5HTR1b, 5HTR2a, 5HTR5b), dopamine D2 receptors (DRD2), and adrenergic receptors alpha 1a and 1d (AdRα1a and AdRα1b), but increased levels for serotonin synthesizing enzyme, tryptophan hydroxylase (TPH), and dopamine D4 receptor (DRD4) when compared to control wild-type mice. The hippocampus of KIV mice showed decreased levels of 5HTR5b. Our results provide causal evidence that lack of promoter IV-driven BDNF disturbs expression of monoaminergic genes in the frontal cortex and hippocampus. These disturbed expression changes in the monoamine systems may mediate the depression- and schizophrenia-like behavior of KIV mice. Our results also suggest that antidepressant and antipsychotic treatments may actually interfere with and normalize the disturbed monoamine systems caused by reduced activity-dependent BDNF, while the treatment responses to these drugs may differ in the subject with reduced BDNF levels caused by stress and lack of neuronal activity.
Collapse
Affiliation(s)
- K Sakata
- Department of Pharmacology, University of Tennessee Health Science Center, Memphis, TN, USA.
| | - S M Duke
- Department of Pharmacology, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
14
|
α1-Adrenergic receptor subtypes in the central nervous system: insights from genetically engineered mouse models. Pharmacol Rep 2013; 65:1489-97. [DOI: 10.1016/s1734-1140(13)71509-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Revised: 09/20/2013] [Indexed: 11/18/2022]
|
15
|
Effects of the noradrenergic neurotoxin DSP-4 on the expression of α1-adrenoceptor subtypes after antidepressant treatment. Pharmacol Rep 2012; 63:1349-58. [PMID: 22358083 DOI: 10.1016/s1734-1140(11)70699-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2011] [Revised: 11/14/2011] [Indexed: 11/23/2022]
Abstract
We have previously reported that chronic imipramine and electroconvulsive treatments increase the α(1A)-adrenoceptor (but not the α(1B) subtype) mRNA level and the receptor density in the rat cerebral cortex. Furthermore, we have also shown that chronic treatment with citalopram does not affect the expression of either the α(1A)- or the α(1B)-adrenoceptor, indicating that the previously observed up-regulation of α(1A)-adrenoceptor may depend on the noradrenergic component of the pharmacological mechanism of action of these antidepressants. Here, we report that previous noradrenergic depletion with DSP-4 (50 mg/kg) (a neurotoxin selective for the noradrenergic nerve terminals) significantly attenuated the increase of α(1A)-adrenoceptor mRNA induced by a 14-day treatment with imipramine (IMI, 20 mg/kg, ip) and abolished the effect of electroconvulsive shock (ECS, 150 mA, 0.5 s) in the prefrontal cortex of the rat brain. The changes in the receptor protein expression (as reflected by its density) that were induced by IMI and ECS treatments were differently modulated by DSP-4 lesioning, and only the ECS-induced increase in α(1A)-adrenoceptor level was abolished. This study provides further evidence corroborating our initial hypothesis that the noradrenergic component of the action of antidepressant agents plays an essential role in the modulation of α(1A)-adrenoceptor in the rat cerebral cortex.
Collapse
|
16
|
Wakabayashi C, Kiyama Y, Kunugi H, Manabe T, Iwakura Y. Age-dependent regulation of depression-like behaviors through modulation of adrenergic receptor α1A subtype expression revealed by the analysis of interleukin-1 receptor antagonist knockout mice. Neuroscience 2011; 192:475-84. [DOI: 10.1016/j.neuroscience.2011.06.031] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2010] [Revised: 05/29/2011] [Accepted: 06/09/2011] [Indexed: 11/29/2022]
|
17
|
Differential modulation of α-1 adrenoceptor subtypes by antidepressants in the rat brain. J Neural Transm (Vienna) 2010; 117:1423-30. [PMID: 21136124 DOI: 10.1007/s00702-010-0522-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2010] [Accepted: 10/31/2010] [Indexed: 10/18/2022]
Abstract
The aim of the present study was to examine the effect of chronic antidepressants treatment on the density of α₁-adrenoceptor (AR) subtypes in rat brain. Density of total α₁ and α(1A)- and α(1Β)-ARs was measured in cortex and cerebellum of rats treated with amitriptyline (AMI), desipramine (DMI) and fluoxetine (FLX), (10 mg/kg body wt), for 30 days, using [³H]prazosin in presence and absence of WB-4101. The density of cortical total α₁-ARs was significantly decreased with AMI (54%) and DMI (25%) treatment, without altering the affinity of the receptor. Fluoxetine did not alter the density of cortical α₁-ARs. The density of cortical α(1A)-ARs was also significantly decreased with AMI (85%) and DMI (50%) treatment, without affecting the affinity. The density of cerebellar total α₁-ARs was significantly decreased with AMI (37%), DMI (50%) and FLX (70%) treatment, without affecting the affinity for [³H]prazosin. The density of α(1A)-ARs was significantly decreased with AMI (67%), DMI (59%) and FLX (92%) treatment. α(1B)-AR density was decreased only with FLX (47%) and DMI (47%) treatment. Correspondingly the basal IP3 and NE (10 μM) stimulated IP3 levels were significantly decreased in AMI (47%), DMI (22%) and FLX (48%) treated rat cortex. The results suggest that chronic antidepressant (AD) treatment down-regulates the cortical and cerebellar total α₁-ARs in rat brain. However, α(1A) subtype is predominantly down-regulated by AMI and DMI, where as FLX affects cerebellar α(1A)-ARs. The region-specific and subtype specific down-regulation of α₁-ARs density, which occurs after prolonged AD treatment, may underline the therapeutic mechanism of action.
Collapse
|
18
|
Abstract
Antidepressant drugs represent one of the main forms of effective treatment for the amelioration of depressive symptoms. Most available antidepressants increase extracellular levels of monoamines. However, it is now recognized that monoamine levels and availability are only part of the story, and that antidepressants whose mechanism of action is mainly based on the modulation of monoaminergic systems may not be able to satisfy the unmet needs of depression. Therefore, a number of compounds, developed for their potential antidepressant activity, are endowed with putative mechanisms of action not affecting traditional monoamine targets. This article briefly reviews, within a mechanistic perspective, the pharmacological profiles of representative antidepressants from each class, including monoamine oxidase inhibitors, tricyclics, norepinephrine reuptake inhibitors, selective serotonin reuptake inhibitors, norepinephrine and serotonin reuptake inhibitors, antidepressants interacting with dopaminergic, melatonergic, glutamatergic, or neuropeptide systems. The undesirable side effects of currently used antidepressants, which can often be a reason for lack of compliance, are also considered.
Collapse
|
19
|
Nojimoto FD, Mueller A, Hebeler-Barbosa F, Akinaga J, Lima V, Kiguti LRDA, Pupo AS. The tricyclic antidepressants amitriptyline, nortriptyline and imipramine are weak antagonists of human and rat alpha1B-adrenoceptors. Neuropharmacology 2010; 59:49-57. [PMID: 20363235 DOI: 10.1016/j.neuropharm.2010.03.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2010] [Revised: 03/24/2010] [Accepted: 03/26/2010] [Indexed: 12/11/2022]
Abstract
Although it is long known that the tricyclic antidepressants amitriptyline, nortriptyline and imipramine inhibit the noradrenaline transporter and alpha(1)-adrenoceptors with similar affinities, which may lead to self-cancelling actions, the selectivity of these drugs for alpha(1)-adrenoceptor subtypes is unknown. The present study investigates the selectivity of amitriptyline, nortriptyline and imipramine for human recombinant and rat native alpha(1)-adrenoceptor subtypes. The selectivity of amitriptyline, nortriptyline and imipramine was investigated in HEK-293 cells expressing each of the human alpha(1)-subtypes and in rat native receptors from the vas deferens (alpha(1A)), spleen (alpha(1B)) and aorta (alpha(1D)) through [(3)H]prazosin binding, and noradrenaline-induced intracellular Ca(2+) increases and contraction assays. Amitriptyline, nortriptyline and imipramine showed considerably higher affinities for alpha(1A)- (approximately 25- to 80-fold) and alpha(1D)-adrenoceptors (approximately 10- to 25-fold) than for alpha(1B)-adrenoceptors in both contraction and [(3)H]prazosin binding assays with rat native and human receptors, respectively. In addition, amitriptyline, nortriptyline and imipramine were substantially more potent in the inhibition of noradrenaline-induced intracellular Ca(2+) increases in HEK-293 cells expressing alpha(1A)- or a truncated version of alpha(1D)-adrenoceptors which traffics more efficiently towards the cell membrane than in cells expressing alpha(1B)-adrenoceptors. Amitriptyline, nortriptyline and imipramine are much weaker antagonists of rat and human alpha(1B)-adrenoceptors than of alpha(1A)- and alpha(1D)-adrenoceptors. The differential affinities for these receptors indicate that the alpha(1)-adrenoceptor subtype which activation is most increased by the augmented noradrenaline availability resultant from the blockade of neuronal reuptake is the alpha(1B)-adrenoceptor. This may be important for the behavioural effects of these drugs.
Collapse
Affiliation(s)
- F D Nojimoto
- Department of Pharmacology, Instituto de Biociências, UNESP, Botucatu, SP, Brazil
| | | | | | | | | | | | | |
Collapse
|
20
|
Doze VA, Handel EM, Jensen KA, Darsie B, Luger EJ, Haselton JR, Talbot JN, Rorabaugh BR. alpha(1A)- and alpha(1B)-adrenergic receptors differentially modulate antidepressant-like behavior in the mouse. Brain Res 2009; 1285:148-57. [PMID: 19540213 PMCID: PMC2720445 DOI: 10.1016/j.brainres.2009.06.035] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2008] [Revised: 06/09/2009] [Accepted: 06/11/2009] [Indexed: 01/23/2023]
Abstract
Tricyclic antidepressant (TCA) drugs are used for the treatment of chronic depression, obsessive-compulsive disorder (OCD), and anxiety-related disorders. Chronic use of TCA drugs increases the expression of alpha(1)-adrenergic receptors (alpha(1)-ARs). Yet, it is unclear whether increased alpha(1)-AR expression contributes to the antidepressant effects of these drugs or if this effect is unrelated to their therapeutic benefit. In this study, mice expressing constitutively active mutant alpha(1A)-ARs (CAM alpha(1A)-AR) or CAM alpha(1B)-ARs were used to examine the effects of alpha(1A)- and alpha(1B)-AR signaling on rodent behavioral models of depression, OCD, and anxiety. CAM alpha(1A)-AR mice, but not CAM alpha(1B)-AR mice, exhibited antidepressant-like behavior in the tail suspension test and forced swim test. This behavior was reversed by prazosin, a selective alpha(1)-AR inverse agonist, and mimicked by chronically treating wild type mice with cirazoline, an alpha(1A)-AR agonist. Marble burying behavior, commonly used to model OCD in rodents, was significantly decreased in CAM alpha(1A)-AR mice but not in CAM alpha(1B)-AR mice. In contrast, no significant differences in anxiety-related behavior were observed between wild type, CAM alpha(1A)-AR, and CAM alpha(1B)-AR animals in the elevated plus maze and light/dark box. This is the first study to demonstrate that alpha(1A)- and alpha(1B)-ARs differentially modulate antidepressant-like behavior in the mouse. These data suggest that alpha(1A)-ARs may be a useful therapeutic target for the treatment of depression.
Collapse
MESH Headings
- Adrenergic alpha-Agonists/pharmacology
- Animals
- Antidepressive Agents/pharmacology
- Behavior, Animal/drug effects
- Behavior, Animal/physiology
- Brain/drug effects
- Brain/metabolism
- Brain/physiopathology
- Catecholamines/metabolism
- Depressive Disorder/drug therapy
- Depressive Disorder/metabolism
- Depressive Disorder/physiopathology
- Disease Models, Animal
- Female
- Imidazoles/pharmacology
- Male
- Maze Learning/drug effects
- Maze Learning/physiology
- Mice
- Mice, Inbred CBA
- Neuropsychological Tests
- Prazosin/pharmacology
- Receptors, Adrenergic, alpha-1/drug effects
- Receptors, Adrenergic, alpha-1/metabolism
- Stress, Psychological/complications
- Stress, Psychological/metabolism
- Stress, Psychological/physiopathology
Collapse
Affiliation(s)
- Van A Doze
- Department of Pharmacology, Physiology and Therapeutics, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58202, USA
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Selken J, Nichols DE. Alpha1-adrenergic receptors mediate the locomotor response to systemic administration of (+/-)-3,4-methylenedioxymethamphetamine (MDMA) in rats. Pharmacol Biochem Behav 2007; 86:622-30. [PMID: 17363047 PMCID: PMC1976288 DOI: 10.1016/j.pbb.2007.02.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2006] [Revised: 02/08/2007] [Accepted: 02/08/2007] [Indexed: 10/23/2022]
Abstract
The recreational drug 3,4-methylenedioxymethamphetamine (MDMA, ecstasy) increases locomotor activity when administered to rats. Although the published pharmacology of MDMA has focused almost exclusively on the roles of serotonin and dopamine, in vitro studies indicate that MDMA induces serotonin and norepinephrine release with equal potency. The present experiments tested the hypothesis that blockade of alpha(1)-adrenoceptors with systemic or local administration of the antagonist prazosin would attenuate the locomotor response to systemic administration of (+/-)-MDMA. Pretreatment with systemic prazosin (0.5 mg/kg) or microinjections into either the prefrontal cortex or ventral tegmental area completely blocked the locomotor stimulant effects of 5 mg/kg (+/-)-MDMA, assessed using a computerized Behavioral Pattern Monitor. Prazosin was more potent in blocking the locomotor stimulant effects of (+/-)-MDMA than a 2 mg/kg dose of (+)-amphetamine that produced a similar locomotor activity increase. These results indicate that activation of alpha(1)-adrenoceptors in both the prefrontal cortex and ventral tegmental areas modulates the locomotor response to MDMA.
Collapse
Affiliation(s)
- Jennifer Selken
- Department of Medicinal Chemistry and Molecular Pharmacology, School of Pharmacy and Pharmaceutical Sciences, Purdue University, West Lafayette, IN 47907, USA
| | | |
Collapse
|
22
|
Dziedzicka-Wasylewska M, Faron-Górecka A, Kuśmider M, Drozdowska E, Rogóz Z, Siwanowicz J, Caron MG, Bönisch H. Effect of antidepressant drugs in mice lacking the norepinephrine transporter. Neuropsychopharmacology 2006; 31:2424-32. [PMID: 16554743 DOI: 10.1038/sj.npp.1301064] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
One of the main theories concerning the mechanism of action of antidepressant drugs (ADs) is based on the notion that the neurochemical background of depression involves an impairment of central noradrenergic transmission with a concomitant decrease of the norepinephrine (NE) in the synaptic gap. Many ADs increase synaptic NE availability by inhibition of the reuptake of NE. Using mice lacking NE transporter (NET-/-) we examined their baseline phenotype as well as the response in the forced swim test (FST) and in the tail suspension test (TST) upon treatment with ADs that display different pharmacological profiles. In both tests, the NET-/- mice behaved like wild-type (WT) mice acutely treated with ADs. Autoradiographic studies showed decreased binding of the beta-adrenergic ligand [3H]CGP12177 in the cerebral cortex of NET-/- mice, indicating the changes at the level of beta-adrenergic receptors similar to those obtained with ADs treatment. The binding of [3H]prazosin to alpha1-adrenergic receptors in the cerebral cortex of NET-/- mice was also decreased, most probably as an adaptive response to the sustained elevation of extracellular NE levels observed in these mice. A pronounced NET knockout-induced shortening of the immobility time in the TST (by ca 50%) compared to WT mice was not reduced any further by NET-inhibiting ADs such as reboxetine, desipramine, and imipramine. Citalopram, which is devoid of affinity for the NET, exerted a significant reduction of immobility time in the NET-/- mice. In the FST, reboxetine, desipramine, imipramine, and citalopram administered acutely did not reduce any further the immobility time shortened by NET knockout itself (ca 25%); however, antidepressant-like action of repeatedly (7 days) administered desipramine was observed in NET-/- mice, indicating that the chronic presence of this drug may also affect other neurochemical targets involved in the behavioral reactions monitored by this test. From the present study, it may be concluded that mice lacking the NET may represent a good model of some aspects of depression-resistant behavior, paralleled with alterations in the expression of adrenergic receptors, which result as an adaptation to elevated levels of extracellular NE.
Collapse
|
23
|
Millan MJ. Multi-target strategies for the improved treatment of depressive states: Conceptual foundations and neuronal substrates, drug discovery and therapeutic application. Pharmacol Ther 2006; 110:135-370. [PMID: 16522330 DOI: 10.1016/j.pharmthera.2005.11.006] [Citation(s) in RCA: 389] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2005] [Accepted: 11/28/2005] [Indexed: 12/20/2022]
Abstract
Major depression is a debilitating and recurrent disorder with a substantial lifetime risk and a high social cost. Depressed patients generally display co-morbid symptoms, and depression frequently accompanies other serious disorders. Currently available drugs display limited efficacy and a pronounced delay to onset of action, and all provoke distressing side effects. Cloning of the human genome has fuelled expectations that symptomatic treatment may soon become more rapid and effective, and that depressive states may ultimately be "prevented" or "cured". In pursuing these objectives, in particular for genome-derived, non-monoaminergic targets, "specificity" of drug actions is often emphasized. That is, priority is afforded to agents that interact exclusively with a single site hypothesized as critically involved in the pathogenesis and/or control of depression. Certain highly selective drugs may prove effective, and they remain indispensable in the experimental (and clinical) evaluation of the significance of novel mechanisms. However, by analogy to other multifactorial disorders, "multi-target" agents may be better adapted to the improved treatment of depressive states. Support for this contention is garnered from a broad palette of observations, ranging from mechanisms of action of adjunctive drug combinations and electroconvulsive therapy to "network theory" analysis of the etiology and management of depressive states. The review also outlines opportunities to be exploited, and challenges to be addressed, in the discovery and characterization of drugs recognizing multiple targets. Finally, a diversity of multi-target strategies is proposed for the more efficacious and rapid control of core and co-morbid symptoms of depression, together with improved tolerance relative to currently available agents.
Collapse
Affiliation(s)
- Mark J Millan
- Institut de Recherches Servier, Centre de Recherches de Croissy, Psychopharmacology Department, 125, Chemin de Ronde, 78290-Croissy/Seine, France.
| |
Collapse
|
24
|
Millan MJ. The role of monoamines in the actions of established and "novel" antidepressant agents: a critical review. Eur J Pharmacol 2005; 500:371-84. [PMID: 15464046 DOI: 10.1016/j.ejphar.2004.07.038] [Citation(s) in RCA: 190] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/01/2004] [Indexed: 11/19/2022]
Abstract
Monoaminergic pathways are highly responsive to aversive stimuli and play a crucial role in the control of affect, cognition, endocrine secretion, chronobiotic rhythms, appetite, and motor function, all of which are profoundly disrupted in depressive states. Accordingly, a perturbation of monoaminergic transmission is implicated in the aetiology of depressive disorders, and all clinically available antidepressants increase corticolimbic availability of monoamines. However, their limited efficacy, delayed onset of action, and undesirable side effects underlie ongoing efforts to identify improved therapeutic agents. Sequencing the human genome has raised the hope not only of better symptomatic control of depression, but even of the prevention or cure of depressive states. In the pursuit of these goals, there is currently a tendency to focus on selective ligands of "novel" nonmonoaminergic targets. However, certain classes of novel agent (such as neurokinin(1) receptor antagonists) indirectly modulate the activity of monoaminergic networks. Others may act "downstream" of them, converging onto common cellular substrates controlling gene expression, synaptic plasticity, and neurogenesis. Further, by analogy to the broad-based actions of currently employed drugs, multitarget agents may be better adapted than selective agents to the management of depression-a complex disorder with hereditary, developmental, and environmental origins. It is, thus, important to continue the creative exploration of clinically validated and innovative monoaminergic strategies within a multitarget framework. In this light, drugs combining monoaminergic and nonmonoaminergic mechanisms of action may be of particular interest. The present article provides a critical overview of monoaminergic strategies for the treatment of depressive states, both established and under development, and discusses interactions of novel "nonmonoaminergic" antidepressants with monoaminergic mechanisms.
Collapse
Affiliation(s)
- Mark J Millan
- Psychopharmacology Department, IdR Servier, 125 Chemin de Ronde, Croissy/Seine, Paris 78290, France.
| |
Collapse
|
25
|
Abstract
The past decade has seen a rapid progression in our knowledge of the neurobiological basis of fear and anxiety. Specific neurochemical and neuropeptide systems have been demonstrated to play important roles in the behaviors associated with fear and anxiety-producing stimuli. Long-term dysregulation of these systems appears to contribute to the development of anxiety disorders, including panic disorder, posttraumatic stress disorder (PTSD), and social anxiety disorder. These neurochemical and neuropeptide systems have been shown to have effects on distinct cortical and subcortical brain areas that are relevant to the mediation of the symptoms associated with anxiety disorders. Moreover, advances in molecular genetics portend the identification of the genes that underlie the neurobiological disturbances that increase the vulnerability to anxiety disorders. This chapter reviews clinical research pertinent to the neurobiological basis of anxiety disorders. The implications of this synthesis for the discovery of anxiety disorder vulnerability genes and novel psychopharmacological approaches will also be discussed.
Collapse
Affiliation(s)
- A Neumeister
- Mood and Anxiety Disorders Research Program, National Institute of Mental Health/NIH, 15K North Drive, MSC 2670, Bethesda MD, 20892-2670, USA.
| | | | | |
Collapse
|
26
|
Abstract
Fear is an adaptive component of the acute "stress" response to potentially-dangerous (external and internal) stimuli which threaten to perturb homeostasis. However, when disproportional in intensity, chronic and/or irreversible, or not associated with any genuine risk, it may be symptomatic of a debilitating anxious state: for example, social phobia, panic attacks or generalized anxiety disorder. In view of the importance of guaranteeing an appropriate emotional response to aversive events, it is not surprising that a diversity of mechanisms are involved in the induction and inhibition of anxious states. Apart from conventional neurotransmitters, such as monoamines, gamma-amino-butyric acid (GABA) and glutamate, many other modulators have been implicated, including: adenosine, cannabinoids, numerous neuropeptides, hormones, neurotrophins, cytokines and several cellular mediators. Accordingly, though benzodiazepines (which reinforce transmission at GABA(A) receptors), serotonin (5-HT)(1A) receptor agonists and 5-HT reuptake inhibitors are currently the principle drugs employed in the management of anxiety disorders, there is considerable scope for the development of alternative therapies. In addition to cellular, anatomical and neurochemical strategies, behavioral models are indispensable for the characterization of anxious states and their modulation. Amongst diverse paradigms, conflict procedures--in which subjects experience opposing impulses of desire and fear--are of especial conceptual and therapeutic pertinence. For example, in the Vogel Conflict Test (VCT), the ability of drugs to release punishment-suppressed drinking behavior is evaluated. In reviewing the neurobiology of anxious states, the present article focuses in particular upon: the multifarious and complex roles of individual modulators, often as a function of the specific receptor type and neuronal substrate involved in their actions; novel targets for the management of anxiety disorders; the influence of neurotransmitters and other agents upon performance in the VCT; data acquired from complementary pharmacological and genetic strategies and, finally, several open questions likely to orientate future experimental- and clinical-research. In view of the recent proliferation of mechanisms implicated in the pathogenesis, modulation and, potentially, treatment of anxiety disorders, this is an opportune moment to survey their functional and pathophysiological significance, and to assess their influence upon performance in the VCT and other models of potential anxiolytic properties.
Collapse
Affiliation(s)
- Mark J Millan
- Psychopharmacology Department, Centre de Rescherches de Croissy, Institut de Recherches (IDR) Servier, 125 Chemin de Ronde, 78290 Croissy-sur-Seine, Paris, France.
| |
Collapse
|