1
|
Fuhr A, Roediger R, Simelitidis M, Gamper-Tsigaras J, Templin M, Kormann M, Antkowiak B, Rudolph U, Köhler D, Rosenberger P, Ngamsri KC, Konrad FM. Regulation of neutrophil migration in acute pulmonary inflammation by extraneuronal α1 gamma-aminobutyric acid A receptors. Cell Death Dis 2025; 16:313. [PMID: 40251174 PMCID: PMC12008292 DOI: 10.1038/s41419-025-07488-1] [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: 11/07/2024] [Revised: 02/03/2025] [Accepted: 02/26/2025] [Indexed: 04/20/2025]
Abstract
Acute pulmonary inflammation is a common disease on intensive care. Due to the lack of specific treatments, lethality is still very high. The ionotropic GABAA-receptors are known from the central nervous system (CNS) and have recently been detected in the lung. These receptors have been shown to influence inflammatory processes. Opposing data has been reported, where both, GABA site agonists and antagonists achieved anti-inflammatory effects. The distribution of the 19 known GABAA-receptor subunits (α1-6, β1-3, γ1-3, δ, ε, π, θ and ρ1-3) and their role in inflammation remain unclear. In murine models of LPS- and bacteria-induced inflammation, Muscimol (GABAA-receptor agonist) and Bicuculline (antagonist) were administered before the onset of inflammation. Transcription of GABAA-receptor subunits was evaluated by real-time polymerase chain reaction. Neutrophil counts and adhesion molecule expression in wild type and GABAα1 knock-in mice were determined by flow-cytometry. Myeloperoxidase, neutrophil extracellular traps and cytokines were determined. In a model of ventilator-induced lung injury, blood gas analysis was performed using arterial blood. A multiplex western blot was done to assess signaling proteins. Muscimol and Bicuculline inhibited neutrophil influx in the bronchoalveolar lavage but did not change neutrophil activation. Both altered surface expression of adhesion molecules on neutrophils and reduced release of interleukin-6 (IL-6). The increased α1 subunit expression on lung epithelium and endothelium after inflammation was abolished by Muscimol and Bicuculline. In GABAα1-knock-in mice the protective effects of both agents were no longer observed. Only Muscimol lowered protein extravasation, improved blood gas analysis and lung function. A multiplex assay ascribed these anti-inflammatory effects to the influence of the IL-6 and phosphoinositide 3-kinase signaling pathways. In conclusion, Muscimol and Bicuculline exert various protective effects in two murine models of acute pulmonary inflammation. The multiple effects of Muscimol were linked to the inhibition of the proinflammatory signaling pathways IL-6 and PI3K.
Collapse
Affiliation(s)
- Anika Fuhr
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Tuebingen, Tübingen, Germany
| | - Robin Roediger
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Tuebingen, Tübingen, Germany
| | - Mariana Simelitidis
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Tuebingen, Tübingen, Germany
| | - Jutta Gamper-Tsigaras
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Tuebingen, Tübingen, Germany
| | - Markus Templin
- NMI Natural and Medical Sciences Institute, University Tuebingen, Tübingen, Germany
| | - Michael Kormann
- Stem Cell Network Tuebingen, University Tuebingen, Tübingen, Germany
| | - Bernd Antkowiak
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Tuebingen, Tübingen, Germany
| | - Uwe Rudolph
- Department of Comparative Biosicences, College of Veterinary Medicine, and Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Champaign, IL, USA
| | - David Köhler
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Tuebingen, Tübingen, Germany
| | - Peter Rosenberger
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Tuebingen, Tübingen, Germany
| | - Kristian-Christos Ngamsri
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Tuebingen, Tübingen, Germany
| | - Franziska M Konrad
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Tuebingen, Tübingen, Germany.
| |
Collapse
|
2
|
Cardona-Acosta AM, Meisser N, Vardeleon NI, Steiner H, Bolaños-Guzmán CA. Mother's little helper turned a foe: Alprazolam use, misuse, and abuse. Prog Neuropsychopharmacol Biol Psychiatry 2025; 136:111137. [PMID: 39260815 DOI: 10.1016/j.pnpbp.2024.111137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 08/27/2024] [Accepted: 09/04/2024] [Indexed: 09/13/2024]
Abstract
Benzodiazepines are effective in managing anxiety and related disorders when used properly (short-term). Their inappropriate use, however, carries significant risks, involving amnesia, rebound insomnia, rebound anxiety, depression, dependence, abuse, addiction, and an intense and exceedingly prolonged withdrawal, among other complications. Benzodiazepines also amplify the effects of opioids and, consequently, have been implicated in approximately 30 % of opioid overdose deaths. Despite their unfavorable profile, sharp increases in medical and non-medical use of benzodiazepines have been steadily reported worldwide. Alprazolam (Xanax®), a potent, short-acting benzodiazepine, is among the most prescribed and abused anxiolytics in the United States. This medication is commonly co-abused with opioids, increasing the likelihood for oversedation, overdose, and death. Notwithstanding these risks, it is surprising that research investigating how benzodiazepines, such as alprazolam, interact with opioids is severely lacking in clinical and preclinical settings. This review therefore aims to present our current knowledge of benzodiazepine use and misuse, with an emphasis on alprazolam when data is available, and particularly in populations at higher risk for developing substance use disorders. Additionally, the potential mechanism(s) surrounding tolerance, dependence and abuse liability are discussed. Despite their popularity, our understanding of how benzodiazepines and opioids interact is less than adequate. Therefore, it is now more important than ever to understand the short- and long-term consequences of benzodiazepine/alprazolam use.
Collapse
Affiliation(s)
- Astrid M Cardona-Acosta
- Department of Psychological and Brain Sciences, and Institute for Neuroscience, Texas A&M University, College Station, TX 77843, USA
| | - Noelle Meisser
- Department of Psychological and Brain Sciences, and Institute for Neuroscience, Texas A&M University, College Station, TX 77843, USA
| | - Nathan I Vardeleon
- Department of Psychological and Brain Sciences, and Institute for Neuroscience, Texas A&M University, College Station, TX 77843, USA
| | - Heinz Steiner
- Stanson Toshok Center for Brain Function and Repair, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA; Discipline of Cellular and Molecular Pharmacology, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| | - Carlos A Bolaños-Guzmán
- Department of Psychological and Brain Sciences, and Institute for Neuroscience, Texas A&M University, College Station, TX 77843, USA.
| |
Collapse
|
3
|
Batallán Burrowes AA, Moisan É, Garrone A, Buynack LM, Chapman CA. 17β-Estradiol reduces inhibitory synaptic currents in entorhinal cortex neurons through G protein-coupled estrogen receptor-1 activation of extracellular signal-regulated kinase. Hippocampus 2024; 34:454-463. [PMID: 39150316 DOI: 10.1002/hipo.23621] [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: 01/30/2024] [Revised: 05/02/2024] [Accepted: 06/05/2024] [Indexed: 08/17/2024]
Abstract
Estrogens are believed to modulate cognitive functions in part through the modulation of synaptic transmission in the cortex and hippocampus. Administration of 17β-estradiol (E2) can rapidly enhance excitatory synaptic transmission in the hippocampus and facilitate excitatory synaptic transmission in rat lateral entorhinal cortex via activation of the G protein-coupled estrogen receptor-1 (GPER1). To assess the mechanisms through which GPER1 activation facilitates synaptic transmission, we assessed the effects of acute 10 nM E2 administration on pharmacologically isolated evoked excitatory and inhibitory synaptic currents in layer II/III entorhinal neurons. Female Long-Evans rats were ovariectomized between postnatal day (PD) 63 and 74 and implanted with a subdermal E2 capsule to maintain continuous low levels of E2. Electrophysiological recordings were obtained between 7 and 20 days after ovariectomy. Application of E2 for 20 min did not significantly affect AMPA or NMDA receptor-mediated excitatory synaptic currents. However, GABA receptor-mediated inhibitory synaptic currents (IPSCs) were markedly reduced by E2 and returned towards baseline levels during the 20-min washout period. The inhibition of GABA-mediated IPSCs was blocked in the presence of the GPER1 receptor antagonist G15. GPER1 can modulate protein kinase A (PKA), but blocking PKA with intracellular KT5720 did not prevent the E2-induced reduction in IPSCs. GPER1 can also stimulate extracellular signal-regulated kinase (ERK), a negative modulator of GABAA receptors, and blocking activation of ERK with PD90859 prevented the E2-induced reduction of IPSCs. E2 can therefore result in a rapid GPER1 and ERK signaling-mediated reduction in GABA-mediated IPSCs. This provides a novel mechanism through which E2 can rapidly modulate synaptic excitability in entorhinal layer II/III neurons and may also contribute to E2 and ERK-dependent alterations in synaptic transmission in other brain areas.
Collapse
Affiliation(s)
- Ariel A Batallán Burrowes
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montreal, Québec, Canada
| | - Élyse Moisan
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montreal, Québec, Canada
| | - Aurelie Garrone
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montreal, Québec, Canada
| | - Lauren M Buynack
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montreal, Québec, Canada
| | - C Andrew Chapman
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montreal, Québec, Canada
| |
Collapse
|
4
|
Lacroix A, Proteau-Lemieux M, Côté S, Near J, Hui SC, Edden RA, Lippé S, Çaku A, Corbin F, Lepage JF. Multimodal assessment of the GABA system in patients with fragile-X syndrome and neurofibromatosis of type 1. Neurobiol Dis 2022; 174:105881. [DOI: 10.1016/j.nbd.2022.105881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 09/12/2022] [Accepted: 10/02/2022] [Indexed: 11/24/2022] Open
|
5
|
Effects of docosanyl ferulate, a constituent of Withania somnifera, on ethanol- and morphine-elicited conditioned place preference and ERK phosphorylation in the accumbens shell of CD1 mice. Psychopharmacology (Berl) 2022; 239:795-806. [PMID: 35088095 PMCID: PMC8891193 DOI: 10.1007/s00213-022-06069-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 01/17/2022] [Indexed: 10/27/2022]
Abstract
BACKGROUND Docosanyl ferulate (DF) is a behaviourally active GABAA receptor complex (GABAAR) agonist, recently isolated from the standardized methanolic extract of Withania somnifera Dunal (WSE) root. Previous studies have shown that WSE prevents both ethanol- and morphine-dependent acquisition and expression of conditioned place preference (CPP) and stimulation of dopamine release in the nucleus accumbens shell (AcbSh). AIMS The study aimed at determining (a) whether DF contributes to WSE's ability to affect the acquisition and expression of ethanol- and morphine-elicited CPP and, given that phosphorylation of extracellular signal-regulated kinase (pERK) in the AcbSh is involved in associative learning and motivated behaviours, (b) whether WSE and DF may affect ethanol- and morphine-induced ERKs phosphorylation in the AcbSh. METHODS In adult male CD1 mice, DF's effects on the acquisition and expression of ethanol- and morphine-elicited CPP were evaluated by a classical place conditioning paradigm, whereas the effects of WSE and DF on ethanol- and morphine-elicited pERK in the AcbSh were evaluated by immunohistochemistry. RESULTS AND CONCLUSIONS The study shows that DF, differently from WSE, affects only the acquisition but not the expression of ethanol- and morphine-induced CPP. Moreover, the study shows that both WSE and DF can prevent ethanol- and morphine-elicited pERK expression in the AcbSh. Overall, these results highlight subtle but critical differences for the role of GABAARs in the mechanism by which WSE affects these ethanol- and morphine-dependent behavioural and molecular/cellular responses and support the suggestion of WSE and DF for the control of different components of drug addiction.
Collapse
|
6
|
Jiang C, Wang Z, Donnelly CR, Wang K, Andriessen AS, Tao X, Matsuda M, Zhao J, Ji RR. PD-1 Regulates GABAergic Neurotransmission and GABA-Mediated Analgesia and Anesthesia. iScience 2020; 23:101570. [PMID: 33083737 PMCID: PMC7530307 DOI: 10.1016/j.isci.2020.101570] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 09/01/2020] [Accepted: 09/14/2020] [Indexed: 12/22/2022] Open
Abstract
The immune checkpoint inhibitor programmed cell death protein 1 (PD-1) plays a critical role in immune regulation. Recent studies have demonstrated functional PD-1 expression in peripheral sensory neurons, which contributes to neuronal excitability, pain, and opioid analgesia. Here we report neuronal expression and function of PD-1 in the central nervous system (CNS), including the spinal cord, thalamus, and cerebral cortex. Notably, GABA-induced currents in spinal dorsal horn neurons, thalamic neurons, and cortical neurons are suppressed by the PD-1-neutralizing immunotherapeutic Nivolumab in spinal cord slices, brain slices, and dissociated cortical neurons. Reductions in GABA-mediated currents in CNS neurons were also observed in Pd1−/− mice without changes in GABA receptor expression. Mechanistically, Nivolumab binds spinal cord neurons and elicits ERK phosphorylation to suppress GABA currents. Finally, both GABA-mediated analgesia and anesthesia are impaired by Pd1 deficiency. Our findings reveal PD-1 as a CNS-neuronal inhibitor that regulates GABAergic signaling and GABA-mediated behaviors. Pd1 mRNA and PD-1 protein are widely expressed in spinal cord and brain neurons GABA-induced currents in CNS neurons are suppressed by PD-1 blockade with Nivolumab Nivolumab binds neuronal PD-1 to induce ERK activation and GABAergic inhibition GABA-mediated pain inhibition and anesthesia is impaired after Pd1 deficiency
Collapse
Affiliation(s)
- Changyu Jiang
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Zilong Wang
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Christopher R Donnelly
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Kaiyuan Wang
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Amanda S Andriessen
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Xueshu Tao
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Megumi Matsuda
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Junli Zhao
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Ru-Rong Ji
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA.,Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA.,Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
7
|
The role of hippocampal GABA A receptors on anxiolytic effects of Echium amoenum extract in a mice model of restraint stress. Mol Biol Rep 2020; 47:6487-6496. [PMID: 32778988 DOI: 10.1007/s11033-020-05699-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 07/26/2020] [Indexed: 01/01/2023]
Abstract
Echium amoenum (EA), a popular medicinal plant in Persian medicine, has anxiolytic, antioxidant, sedative, and anti-inflammatory effects. This study examined whether GABA-ergic signaling is involved in the anxiolytic effects of EA in mice. Sixty BALB/c mice (25-30 g) were divided into six groups (n = 10) as follows: the (I) control group received 10 ml/kg normal saline (NS). In the stress groups, the animals underwent 14 consecutive days of restraint stress (RS), and received following treatments simultaneously; (II) RS + NS; (III) RS + Diaz (Diazepam); (IV) RS + EA; (V) RS + Flu (Flumazenil) + EA; (VI) RS + Flu + Diaz. Behavioral tests including the open field test (OFT) and elevated plus maze (EPM) were performed to evaluate anxiety-like behaviors and the effects of the regimens. The plasma level of corticosterone and the hippocampal protein expressions of IL-1β, TNF-α, CREB, and BDNF, as well as p-GABAA/GABAA ratio, were also assessed. The findings revealed that chronic administration of EA alone produced anxiolytic effects in both behavioral tests, while diazepam alone or in combination with Flu failed to decrease the anxiety-like behaviors. Furthermore, the p-GABAA/GABAA and p-CREB/CREB ratios, and protein levels of BDNF were significantly increased in the EA-received group. On the other hand, plasma corticosterone levels and the hippocampal IL-1β and TNF-α levels were significantly decreased by EA. However, pre-treatment with GABAA receptors (GABAA Rs) antagonist, Flu, reversed the anxiolytic and molecular effects of EA in the RS-subjected animals. Our findings confirmed that alternation of GABAAR is involved in the effects of EA against RS-induced anxiety-like behaviors, HPA axis activation, and neuroinflammation.
Collapse
|
8
|
The molecular and cellular mechanisms of depression: a focus on reward circuitry. Mol Psychiatry 2019; 24:1798-1815. [PMID: 30967681 PMCID: PMC6785351 DOI: 10.1038/s41380-019-0415-3] [Citation(s) in RCA: 143] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 02/18/2019] [Accepted: 03/18/2019] [Indexed: 12/16/2022]
Abstract
Depression is a complex disorder that takes an enormous toll on individual health. As affected individuals display a wide variation in their clinical symptoms, the precise neural mechanisms underlying the development of depression remain elusive. Although it is impossible to phenocopy every symptom of human depression in rodents, the preclinical field has had great success in modeling some of the core affective and neurovegetative depressive symptoms, including social withdrawal, anhedonia, and weight loss. Adaptations in select cell populations may underlie these individual depressive symptoms and new tools have expanded our ability to monitor and manipulate specific cell types. This review outlines some of the most recent preclinical discoveries on the molecular and neurophysiological mechanisms in reward circuitry that underlie the expression of behavioral constructs relevant to depressive symptoms.
Collapse
|
9
|
Marottoli FM, Priego M, Flores-Barrera E, Pisharody R, Zaldua S, Fan KD, Ekkurthi GK, Brady ST, Morfini GA, Tseng KY, Tai LM. EGF Treatment Improves Motor Behavior and Cortical GABAergic Function in the R6/2 Mouse Model of Huntington's Disease. Mol Neurobiol 2019; 56:7708-7718. [PMID: 31104296 DOI: 10.1007/s12035-019-1634-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 04/24/2019] [Indexed: 01/13/2023]
Abstract
Recent evidence indicates that disruption of epidermal growth factor (EGF) signaling by mutant huntingtin (polyQ-htt) may contribute to the onset of behavioral deficits observed in Huntington's disease (HD) through a variety of mechanisms, including cerebrovascular dysfunction. Yet, whether EGF signaling modulates the development of HD pathology and the associated behavioral impairments remain unclear. To gain insight on this issue, we used the R6/2 mouse model of HD to assess the impact of chronic EGF treatment on behavior, and cerebrovascular and cortical neuronal functions. We found that bi-weekly treatment with a low dose of EGF (300 µg/kg, i.p.) for 6 weeks was sufficient to effectively improve motor behavior in R6/2 mice and diminish mortality, compared to vehicle-treated littermates. These beneficial effects of EGF treatment were dissociated from changes in cerebrovascular leakiness, a result that was surprising given that EGF ameliorates this deficit in other neurodegenerative diseases. Rather, the beneficial effect of EGF on R6/2 mice behavior was concomitant with a marked amelioration of cortical GABAergic function. As GABAergic transmission in cortical circuits is disrupted in HD, these novel data suggest a potential mechanistic link between deficits in EGF signaling and GABAergic dysfunction in the progression of HD.
Collapse
Affiliation(s)
- Felecia M Marottoli
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Mercedes Priego
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Eden Flores-Barrera
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Rohan Pisharody
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Steve Zaldua
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Kelly D Fan
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Giri K Ekkurthi
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Scott T Brady
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Gerardo A Morfini
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Kuei Y Tseng
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Leon M Tai
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA.
| |
Collapse
|
10
|
|
11
|
Lee YM, Choi JH, Min WK, Han JK, Oh JW. Induction of functional erythropoietin and erythropoietin receptor gene expression by gamma-aminobutyric acid and piperine in kidney epithelial cells. Life Sci 2018; 215:207-215. [PMID: 30439377 DOI: 10.1016/j.lfs.2018.11.024] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 11/02/2018] [Accepted: 11/10/2018] [Indexed: 11/30/2022]
Abstract
AIMS The aim of this study was to evaluate gamma-aminobutyric acid (GABA)- and piperine-induced erythropoietin (EPO) and EPO-receptor expression. MATERIALS AND METHODS The effect of GABA and piperine on cell viability was examined using kidney epithelial cells. Expression levels of EPO and EPO-R mRNA and protein were evaluated in response to GABA and piperine treatments. GABA- and piperine-mediated activation of the mitogen-activated protein kinase (MAPK) signaling pathway was investigated. Additionally, EPO function was evaluated using conditioned media containing EPO. The GABA receptor type involved in this process was identified. KEY FINDINGS Messenger RNA and protein expression levels of EPO and EPO-R significantly increased in response to treatment with GABA, piperine, or the combination of both, compared with control. GABA plus piperine synergistically enhanced EPO and EPO-R expression through p38 and c-Jun N-terminal kinase (JNK) MAPK signaling pathways, but not through the extracellular signal-regulated kinase (ERK) MAPK pathway. SB203580 and SP600125 (p38 and JNK pathway inhibitors, respectively) attenuated GABA plus piperine-induced EPO and EPO-R expression. Treatment of macrophages with EPO-containing conditioned media induced mRNA expression of interleukin (IL)-10 and nuclear factor (NF)-κB due to the interaction between EPO and EPO-R. Interestingly, GABA-induced EPO and EPO-R expression was mediated through GABAA, not GABAB, receptor activation. SIGNIFICANCE These findings demonstrate that GABA plus piperine-mediated p38 and JNK MAPK activation increases EPO and EPO-R expression, resulting in up-regulation of IL-10 and NF-κB.
Collapse
Affiliation(s)
- Yoon-Mi Lee
- Department of Animal Biotechnology, College of Animal Bioscience and Technology, Konkuk University, Seoul 05029, Republic of Korea
| | - Jun-Ha Choi
- Department of Animal Biotechnology, College of Animal Bioscience and Technology, Konkuk University, Seoul 05029, Republic of Korea
| | - Wan-Kwon Min
- Department of Animal Biotechnology, College of Animal Bioscience and Technology, Konkuk University, Seoul 05029, Republic of Korea
| | - Jong-Kwon Han
- Department of Research and Development Center, Milae Resource ML Co. Ltd., Seoul 05836, Republic of Korea
| | - Jae-Wook Oh
- Department of Animal Biotechnology, College of Animal Bioscience and Technology, Konkuk University, Seoul 05029, Republic of Korea.
| |
Collapse
|
12
|
El Amki M, Binder N, Steffen R, Schneider H, Luft AR, Weller M, Imthurn B, Merki-Feld GS, Wegener S. Contraceptive drugs mitigate experimental stroke-induced brain injury. Cardiovasc Res 2018; 115:637-646. [DOI: 10.1093/cvr/cvy248] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 10/01/2018] [Accepted: 10/04/2018] [Indexed: 12/14/2022] Open
Abstract
AbstractAimsEffective stroke treatments beyond reperfusion remain scant. The natural steroid hormone progesterone has shown protective effects in experimental models of brain injury and cardiovascular disease. However, unfavourable bioavailability limits its clinical use. Desogestrel and drospirenone are new generation progestins with progesterone-like properties, developed as oral contraceptives with excellent bioavailability and safety profile. We investigated the neuroprotective properties of these progestins in vivo using transient middle cerebral artery occlusion (MCAO) and in vitro using an oxygen-glucose deprivation and reoxygenation (OGD/R) model in primary neuronal cells.Methods and resultsMCAO was induced in female, female ovariectomized (modelling postmenopausal females) and male mice. Treatment with the progestins resulted in less severe strokes after MCAO and less neuronal death in OGD/R. Desogestrel and drospirenone induced higher expression levels of GABAAR α4 and delta subunits within the brain, suggesting changes in GABAAR configuration favouring tonic inhibition as potential mechanism of action. Treatment with the GABAAR blocker picrotoxin abolished the protection afforded by the progestins in vivo and in vitro.ConclusionFor the first time, here, we delineate a potential role of desogestrel and drospirenone, both clinically approved and safe drugs in mitigating the consequences of stroke. Contraception with desogestrel and drospirenone in progestin-only preparations may be particularly beneficial for women at risk of stroke.
Collapse
Affiliation(s)
- Mohamad El Amki
- Department of Neurology, University Hospital and University of Zurich, Frauenklinikstrasse 26, 8091 Zurich, Switzerland
| | - Nadine Binder
- Department of Neurology, University Hospital and University of Zurich, Frauenklinikstrasse 26, 8091 Zurich, Switzerland
| | - Riccardo Steffen
- Department of Neurology, University Hospital and University of Zurich, Frauenklinikstrasse 26, 8091 Zurich, Switzerland
| | - Hannah Schneider
- Department of Neurology, University Hospital and University of Zurich, Frauenklinikstrasse 26, 8091 Zurich, Switzerland
| | - Andreas R Luft
- Department of Neurology, University Hospital and University of Zurich, Frauenklinikstrasse 26, 8091 Zurich, Switzerland
| | - Michael Weller
- Department of Neurology, University Hospital and University of Zurich, Frauenklinikstrasse 26, 8091 Zurich, Switzerland
| | - Bruno Imthurn
- Department of Reproductive Endocrinology, University Hospital Zurich, Switzerland
| | | | - Susanne Wegener
- Department of Neurology, University Hospital and University of Zurich, Frauenklinikstrasse 26, 8091 Zurich, Switzerland
| |
Collapse
|
13
|
Porcher C, Medina I, Gaiarsa JL. Mechanism of BDNF Modulation in GABAergic Synaptic Transmission in Healthy and Disease Brains. Front Cell Neurosci 2018; 12:273. [PMID: 30210299 PMCID: PMC6121065 DOI: 10.3389/fncel.2018.00273] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 08/06/2018] [Indexed: 12/18/2022] Open
Abstract
In the mature healthy mammalian neuronal networks, γ-aminobutyric acid (GABA) mediates synaptic inhibition by acting on GABAA and GABAB receptors (GABAAR, GABABR). In immature networks and during numerous pathological conditions the strength of GABAergic synaptic inhibition is much less pronounced. In these neurons the activation of GABAAR produces paradoxical depolarizing action that favors neuronal network excitation. The depolarizing action of GABAAR is a consequence of deregulated chloride ion homeostasis. In addition to depolarizing action of GABAAR, the GABABR mediated inhibition is also less efficient. One of the key molecules regulating the GABAergic synaptic transmission is the brain derived neurotrophic factor (BDNF). BDNF and its precursor proBDNF, can be released in an activity-dependent manner. Mature BDNF operates via its cognate receptors tropomyosin related kinase B (TrkB) whereas proBDNF binds the p75 neurotrophin receptor (p75NTR). In this review article, we discuss recent finding illuminating how mBDNF-TrkB and proBDNF-p75NTR signaling pathways regulate GABA related neurotransmission under physiological conditions and during epilepsy.
Collapse
Affiliation(s)
- Christophe Porcher
- Aix Marseille University, Marseille, France.,Institut National de la Santé et de la Recherche Médicale (INSERM) U901, Marseille, France.,Institut de Neurobiologie de la Méditerranée (INMED), Marseille, France
| | - Igor Medina
- Aix Marseille University, Marseille, France.,Institut National de la Santé et de la Recherche Médicale (INSERM) U901, Marseille, France.,Institut de Neurobiologie de la Méditerranée (INMED), Marseille, France
| | - Jean-Luc Gaiarsa
- Aix Marseille University, Marseille, France.,Institut National de la Santé et de la Recherche Médicale (INSERM) U901, Marseille, France.,Institut de Neurobiologie de la Méditerranée (INMED), Marseille, France
| |
Collapse
|
14
|
Álvaro-Bartolomé M, Salort G, García-Sevilla JA. Disruption of brain MEK-ERK sequential phosphorylation and activation during midazolam-induced hypnosis in mice: Roles of GABA A receptor, MEK1 inactivation, and phosphatase MKP-3. Prog Neuropsychopharmacol Biol Psychiatry 2017; 75:84-93. [PMID: 28111292 DOI: 10.1016/j.pnpbp.2017.01.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 01/11/2017] [Accepted: 01/13/2017] [Indexed: 01/08/2023]
Abstract
Midazolam is a positive allosteric modulator at GABAA receptor that induces a short hypnosis and neuroplasticity, in which the sequential phosphorylation of MEK1/2 and ERK1/2 was shown to play a role. This study investigated the parallel activation of p-MEK and p-ERK and regulatory mechanisms induced by midazolam through the stimulation of GABAA receptors in the mouse brain. During the time course of midazolam (60mg/kg)-induced sleep in mice (lasting for about 2h) p-Ser217/221 MEK1/2 was increased (+146% to +258%) whereas, unexpectedly, p-Tyr204/Thr202 ERK1/2 was found decreased (-16% to -38%), revealing uncoupling of MEK to ERK signals in various brain regions. Midazolam-induced p-MEK1/2 upregulation was prevented by pretreatment (30min) with flumazenil (10mg/kg), indicating the involvement of GABAA receptors. Also unexpectedly, midazolam-induced p-ERK1/2 downregulation was not prevented by flumazenil (10 or 30mg/kg). Notably, during midazolam-induced sleep the content of inactivated p-Thr286 MEK1, which can dampen ERK1/2 activation, was increased (+33% to +149%) through a mechanism sensitive to flumazenil (10mg/kg). Midazolam also increased MKP-3 (+13% to +73%) content and this upregulation was prevented by flumazenil (10mg/kg); an effect suggesting ERK inactivation because MKP-3 is the phosphatase selective for ERK1/2 dephosphorylation. The results indicate that during midazolam-induced sleep in mice there is an uncoupling of p-MEK (increased) to p-ERK (decreased) signals. p-ERK1/2 downregulation (not involving GABAA receptors) is the result of increased inactivated MEK1 and phosphatase MKP-3 (both effects involving GABAA receptors). These findings are relevant for the neurobiology and clinical use of benzodiazepines.
Collapse
Affiliation(s)
- María Álvaro-Bartolomé
- Laboratory of Neuropharmacology, IUNICS-IdISPa, University of the Balearic Islands (UIB), Palma de Mallorca, Spain; Redes Temáticas de Investigación Cooperativa en Salud-Red de Trastornos Adictivos (RETICS-RTA), ISCIII, Madrid, Spain
| | - Glòria Salort
- Laboratory of Neuropharmacology, IUNICS-IdISPa, University of the Balearic Islands (UIB), Palma de Mallorca, Spain; Redes Temáticas de Investigación Cooperativa en Salud-Red de Trastornos Adictivos (RETICS-RTA), ISCIII, Madrid, Spain
| | - Jesús A García-Sevilla
- Laboratory of Neuropharmacology, IUNICS-IdISPa, University of the Balearic Islands (UIB), Palma de Mallorca, Spain; Redes Temáticas de Investigación Cooperativa en Salud-Red de Trastornos Adictivos (RETICS-RTA), ISCIII, Madrid, Spain.
| |
Collapse
|
15
|
Evans MC, Anderson GM. Neuroendocrine integration of nutritional signals on reproduction. J Mol Endocrinol 2017; 58:R107-R128. [PMID: 28057770 DOI: 10.1530/jme-16-0212] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 01/05/2017] [Indexed: 12/28/2022]
Abstract
Reproductive function in mammals is energetically costly and therefore tightly regulated by nutritional status. To enable this integration of metabolic and reproductive function, information regarding peripheral nutritional status must be relayed centrally to the gonadotropin-releasing hormone (GNRH) neurons that drive reproductive function. The metabolically relevant hormones leptin, insulin and ghrelin have been identified as key mediators of this 'metabolic control of fertility'. However, the neural circuitry through which they act to exert their control over GNRH drive remains incompletely understood. With the advent of Cre-LoxP technology, it has become possible to perform targeted gene-deletion and gene-rescue experiments and thus test the functional requirement and sufficiency, respectively, of discrete hormone-neuron signaling pathways in the metabolic control of reproductive function. This review discusses the findings from these investigations, and attempts to put them in context with what is known from clinical situations and wild-type animal models. What emerges from this discussion is clear evidence that the integration of nutritional signals on reproduction is complex and highly redundant, and therefore, surprisingly difficult to perturb. Consequently, the deletion of individual hormone-neuron signaling pathways often fails to cause reproductive phenotypes, despite strong evidence that the targeted pathway plays a role under normal physiological conditions. Although transgenic studies rarely reveal a critical role for discrete signaling pathways, they nevertheless prove to be a good strategy for identifying whether a targeted pathway is absolutely required, critically involved, sufficient or dispensable in the metabolic control of fertility.
Collapse
Affiliation(s)
- Maggie C Evans
- Centre for Neuroendocrinology and Department of AnatomyUniversity of Otago School of Medical Sciences, Dunedin, New Zealand
| | - Greg M Anderson
- Centre for Neuroendocrinology and Department of AnatomyUniversity of Otago School of Medical Sciences, Dunedin, New Zealand
| |
Collapse
|
16
|
Osmanovic-Barilar J, Salkovic-Petrisi M. Evaluating the Role of Hormone Therapy in Postmenopausal Women with Alzheimer’s Disease. Drugs Aging 2016; 33:787-808. [DOI: 10.1007/s40266-016-0407-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
17
|
Molecular basis of the alternative recruitment of GABAA versus glycine receptors through gephyrin. Nat Commun 2014; 5:5767. [DOI: 10.1038/ncomms6767] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 11/04/2014] [Indexed: 01/17/2023] Open
|
18
|
Singh M, Su C, Ng S. Non-genomic mechanisms of progesterone action in the brain. Front Neurosci 2013; 7:159. [PMID: 24065876 PMCID: PMC3776940 DOI: 10.3389/fnins.2013.00159] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Accepted: 08/19/2013] [Indexed: 01/10/2023] Open
Abstract
Progesterone is a gonadal steroid hormone whose physiological effects extend well beyond the strict confines of reproductive function. In fact, progesterone can have important effects on a variety of tissues, including the bone, the heart and the brain. Mechanistically, progesterone has been thought to exert its effects through the progesterone receptor (PR), a member of the nuclear steroid hormone superfamily, and as such, acts through specific progesterone response elements (PRE) within the promoter region of target genes to regulate transcription of such genes. This has been often described as the “genomic” mechanism of progesterone action. However, just as progesterone has a diverse range of tissue targets, the mechanisms through which progesterone elicits its effects are equally diverse. For example, progesterone can activate alternative receptors, such as membrane-associated PRs (distinct from the classical PR), to elicit the activation of several signaling pathways that in turn, can influence cell function. Here, we review various non-nuclear (i.e., non-genomic) signaling mechanisms that progesterone can recruit to elicit its effects, focusing our discussion primarily on those signaling mechanisms by which progesterone influences cell viability in the brain.
Collapse
Affiliation(s)
- Meharvan Singh
- Department of Pharmacology and Neuroscience, Center FOR HER, Institute for Aging and Alzheimer's Disease Research, University of North Texas Health Science Center at Fort Worth Fort Worth, TX, USA
| | | | | |
Collapse
|
19
|
Li W, Huang R, Shetty RA, Thangthaeng N, Liu R, Chen Z, Sumien N, Rutledge M, Dillon GH, Yuan F, Forster MJ, Simpkins JW, Yang SH. Transient focal cerebral ischemia induces long-term cognitive function deficit in an experimental ischemic stroke model. Neurobiol Dis 2013; 59:18-25. [PMID: 23845275 DOI: 10.1016/j.nbd.2013.06.014] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Revised: 06/19/2013] [Accepted: 06/25/2013] [Indexed: 01/27/2023] Open
Abstract
Vascular dementia ranks as the second leading cause of dementia in the United States. However, its underlying pathophysiological mechanism is not fully understood and no effective treatment is available. The purpose of the current study was to evaluate long-term cognitive deficits induced by transient middle cerebral artery occlusion (tMCAO) in rats and to investigate the underlying mechanism. Sprague-Dawley rats were subjected to tMCAO or sham surgery. Behavior tests for locomotor activity and cognitive function were conducted at 7 or 30days after stroke. Hippocampal long term potentiation (LTP) and involvement of GABAergic neurotransmission were evaluated at 30days after sham surgery or stroke. Immunohistochemistry and Western blot analyses were conducted to determine the effect of tMCAO on cell signaling in the hippocampus. Transient MCAO induced a progressive deficiency in spatial performance. At 30days after stroke, no neuron loss or synaptic marker change in the hippocampus were observed. LTP in both hippocampi was reduced at 30days after stroke. This LTP impairment was prevented by blocking GABAA receptors. In addition, ERK activity was significantly reduced in both hippocampi. In summary, we identified a progressive decline in spatial learning and memory after ischemic stroke that correlates with suppression of hippocampal LTP, elevation of GABAergic neurotransmission, and inhibition of ERK activation. Our results indicate that the attenuation of GABAergic activity or enhancement of ERK/MAPK activation in the hippocampus might be potential therapeutic approaches to prevent or attenuate cognitive impairment after ischemic stroke.
Collapse
Affiliation(s)
- Wenjun Li
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Curia G, Gualtieri F, Bartolomeo R, Vezzali R, Biagini G. Resilience to audiogenic seizures is associated with p-ERK1/2 dephosphorylation in the subiculum of Fmr1 knockout mice. Front Cell Neurosci 2013; 7:46. [PMID: 23630463 PMCID: PMC3635025 DOI: 10.3389/fncel.2013.00046] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Accepted: 04/03/2013] [Indexed: 12/30/2022] Open
Abstract
Young, but not adult, fragile X mental retardation gene (Fmr1) knockout (KO) mice display audiogenic seizures (AGS) that can be prevented by inhibiting extracellular signal-regulated kinases 1/2 (ERK1/2) phosphorylation. In order to identify the cerebral regions involved in these phenomena, we characterized the response to AGS in Fmr1 KO mice and wild type (WT) controls at postnatal day (P) 45 and P90. To characterize the diverse response to AGS in various cerebral regions, we evaluated the activity markers FosB/ΔFosB and phosphorylated ERK1/2 (p-ERK1/2). Wild running (100% of tested mice) followed by clonic/tonic seizures (30%) were observed in P45 Fmr1 KO mice, but not in WT mice. In P90 Fmr1 KO mice, wild running was only present in 25% of tested animals. Basal FosB/ΔFosB immunoreactivity was higher (P < 0.01 vs. WT) in the CA1 and subiculum of P45 Fmr1 KO mice. Following the AGS test, FosB/ΔFosB expression consistently increased in most of the analyzed regions in both groups at P45, but not at P90. Interestingly, FosB/ΔFosB immunoreactivity was significantly higher in P45 Fmr1 KO mice in the medial geniculate body (P < 0.05 vs. WT) and CA3 (P < 0.01). Neurons presenting with immunopositivity to p-ERK1/2 were more abundant in the subiculum of Fmr1 KO mice in control condition (P < 0.05 vs. WT, in both age groups). In this region, p-ERK1/2-immunopositive cells significantly decreased (–75%, P < 0.01) in P90 Fmr1 KO mice exposed to the AGS test, but no changes were found in P45 mice or in other brain regions. In both age groups of WT mice, p-ERK1/2-immunopositive cells increased in the subiculum after exposure to the acoustic test. Our findings illustrate that FosB/ΔFosB markers are overexpressed in the medial geniculate body and CA3 in Fmr1 KO mice experiencing AGS, and that p-ERK1/2 is markedly decreased in the subiculum of Fmr1 KO mice resistant to AGS induction. These findings suggest that resilience to AGS is associated with dephosphorylation of p-ERK1/2 in the subiculum of mature Fmr1 KO mice.
Collapse
Affiliation(s)
- Giulia Curia
- Laboratory of Experimental Epileptology, Department of Biomedical, Metabolic, and Neural Sciences, University of Modena and Reggio Emilia Modena, Italy
| | | | | | | | | |
Collapse
|
21
|
Abstract
Numerous studies aimed at identifying the role of estrogen on the brain have used the ovariectomized rodent as the experimental model. And while estrogen intervention in these animals has, at least partially, restored cholinergic, neurotrophin and cognitive deficits seen in the ovariectomized animal, it is worth considering that the removal of the ovaries results in the loss of not only circulating estrogen but of circulating progesterone as well. As such, the various deficits associated with ovariectomy may be attributed to the loss of progesterone as well. Similarly, one must also consider the fact that the human menopause results in the precipitous decline of not just circulating estrogens, but in circulating progesterone as well and as such, the increased risk for diseases such as Alzheimer's disease during the postmenopausal period could also be contributed by this loss of progesterone. In fact, progesterone has been shown to exert neuroprotective effects, both in cell models, animal models and in humans. Here, we review the evidence that supports the neuroprotective effects of progesterone and discuss the various mechanisms that are thought to mediate these protective effects. We also discuss the receptor pharmacology of progesterone's neuroprotective effects and present a conceptual model of progesterone action that supports the complementary effects of membrane-associated and classical intracellular progesterone receptors. In addition, we discuss fundamental differences in the neurobiology of progesterone and the clinically used, synthetic progestin, medroxyprogesterone acetate that may offer an explanation for the negative findings of the combined estrogen/progestin arm of the Women's Health Initiative-Memory Study (WHIMS) and suggest that the type of progestin used may dictate the outcome of either pre-clinical or clinical studies that addresses brain function.
Collapse
Affiliation(s)
- Meharvan Singh
- Department of Pharmacology and Neuroscience, Institute for Aging and Alzheimer's Disease Research, Center FOR HER, University of North Texas Health Science Center at Fort Worth, Fort Worth, TX 76107, USA.
| | | |
Collapse
|
22
|
Singh M, Su C. Progesterone-induced neuroprotection: factors that may predict therapeutic efficacy. Brain Res 2013; 1514:98-106. [PMID: 23340161 DOI: 10.1016/j.brainres.2013.01.027] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Accepted: 01/15/2013] [Indexed: 12/12/2022]
Abstract
Both progesterone and estradiol have well-described neuroprotective effects against numerous insults in a variety of cell culture models, animal models and in humans. However, the efficacy of these hormones may depend on a variety of factors, including the type of hormone used (ex. progesterone versus medroxyprogesterone acetate), the duration of the postmenopausal period prior to initiating the hormone intervention, and potentially, the age of the subject. The latter two factors relate to the proposed existence of a "window of therapeutic opportunity" for steroid hormones in the brain. While such a window of opportunity has been described for estrogen, there is a paucity of information to address whether such a window of opportunity exists for progesterone and its related progestins. Here, we review known cellular mechanisms likely to underlie the protective effects of progesterone and furthermore, describe key differences in the neurobiology of progesterone and the synthetic progestin, medroxyprogesterone acetate (MPA). Based on the latter, we offer a model that defines some of the key cellular and molecular players that predict the neuroprotective efficacy of progesterone. Accordingly, we suggest how changes in the expression or function of these cellular and molecular targets of progesterone with age or prolonged duration of hormone withdrawal (such as following surgical or natural menopause) may impact the efficacy of progesterone. This article is part of a Special Issue entitled Hormone Therapy.
Collapse
Affiliation(s)
- Meharvan Singh
- Department of Pharmacology and Neuroscience, Institute for Aging and Alzheimer's Disease Research, Center FOR HER, University of North Texas Health Science Center at Fort Worth, Fort Worth, TX 76107, USA.
| | | |
Collapse
|
23
|
Ibrahim BM, Abdel-Rahman AA. Enhancement of rostral ventrolateral medulla neuronal nitric-oxide synthase-nitric-oxide signaling mediates the central cannabinoid receptor 1-evoked pressor response in conscious rats. J Pharmacol Exp Ther 2012; 341:579-86. [PMID: 22366659 PMCID: PMC3362886 DOI: 10.1124/jpet.112.192369] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Accepted: 02/23/2012] [Indexed: 11/22/2022] Open
Abstract
Our recent studies implicated brainstem GABAergic signaling in the central cannabinoid receptor 1 (CB(1)R)-mediated pressor response in conscious rats. Given the well established link between neuronal nitric-oxide synthase (nNOS)/nitric oxide (NO) signaling and GABAergic transmission in brainstem cardiovascular regulating areas, we elucidated the role of nNOS-generated NO in the central CB(1)R-elicited pressor response. Compared with vehicle, intracisternal (i.c.) microinjection of the CB(1)R agonist (R)-(+)-[2,3-dihydro-5-methyl-3[(4-morpholinyl)methyl]pyrrolo[1,2,3-de]-1,4-benzoxazinyl]-(1-naphthalenyl) methanone mesylate (WIN55212-2) (15 μg/rat) significantly enhanced nNOS phosphorylation as well as the total nitrate and nitrite content in the rostral ventrolateral medulla (RVLM) at 5, 10, and 30 min, which paralleled the elicited pressor response. These findings were corroborated by: 1) the parallel dose-related increases in blood pressure and RVLM-NO levels, measured in real time by in vivo electrochemistry, elicited by intra-RVLM WIN55212-2 (100, 200, or 300 pmol /80 nl; n = 5) in conscious rats; and 2) the significantly higher phosphorylated nNOS (p-nNOS) levels in the WIN55212-2-injected RVLM compared with the contralateral RVLM. Subsequent neurochemical studies showed that WIN55212-2 (15 μg/rat i.c.) significantly increased the number and percentage of neurons immunostained for nNOS (nitroxidergic neurons) and c-Fos (marker of neuronal activity) within the RVLM. The increases in blood pressure and the neurochemical responses elicited by intracisternal WIN55212-2 were attenuated by prior central CB(1)R blockade by N-(piperidin-1-yl)-5-(4-iodophenyl)-1-(2,4-dichlorophenyl)-4-methyl-1H-pyrazole-3-carboxamide (AM251; 30 μg/rat i.c.) or selective nNOS inhibition by N(ω)-propyl-(L)-arginine (1 μg/rat i.c.). These findings implicate RVLM p-nNOS/NO signaling as a molecular mechanism in the central CB(1)R-evoked pressor effect in conscious rats.
Collapse
Affiliation(s)
- Badr Mostafa Ibrahim
- Department of Pharmacology and Toxicology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | | |
Collapse
|
24
|
Wefers B, Hitz C, Hölter SM, Trümbach D, Hansen J, Weber P, Pütz B, Deussing JM, de Angelis MH, Roenneberg T, Zheng F, Alzheimer C, Silva A, Wurst W, Kühn R. MAPK signaling determines anxiety in the juvenile mouse brain but depression-like behavior in adults. PLoS One 2012; 7:e35035. [PMID: 22529971 PMCID: PMC3329550 DOI: 10.1371/journal.pone.0035035] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2011] [Accepted: 03/08/2012] [Indexed: 11/18/2022] Open
Abstract
MAP kinase signaling has been implicated in brain development, long-term memory, and the response to antidepressants. Inducible Braf knockout mice, which exhibit protein depletion in principle forebrain neurons, enabled us to unravel a new role of neuronal MAPK signaling for emotional behavior. Braf mice that were induced during adulthood showed normal anxiety but increased depression-like behavior, in accordance with pharmacological findings. In contrast, the inducible or constitutive inactivation of Braf in the juvenile brain leads to normal depression-like behavior but decreased anxiety in adults. In juvenile, constitutive mutants we found no alteration of GABAergic neurotransmission but reduced neuronal arborization in the dentate gyrus. Analysis of gene expression in the hippocampus revealed nine downregulated MAPK target genes that represent candidates to cause the mutant phenotype. Our results reveal the differential function of MAPK signaling in juvenile and adult life phases and emphasize the early postnatal period as critical for the determination of anxiety in adults. Moreover, these results validate inducible gene inactivation as a new valuable approach, allowing it to discriminate between gene function in the adult and the developing postnatal brain.
Collapse
Affiliation(s)
- Benedikt Wefers
- German Research Center for Environmental Health, Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg/Munich, Germany
| | - Christiane Hitz
- German Research Center for Environmental Health, Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg/Munich, Germany
| | - Sabine M. Hölter
- German Research Center for Environmental Health, Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg/Munich, Germany
| | - Dietrich Trümbach
- German Research Center for Environmental Health, Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg/Munich, Germany
| | - Jens Hansen
- German Research Center for Environmental Health, Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg/Munich, Germany
| | - Peter Weber
- Molecular Neurogenetics, Max-Planck-Institute of Psychiatry, Munich, Germany
| | - Benno Pütz
- Molecular Neurogenetics, Max-Planck-Institute of Psychiatry, Munich, Germany
| | - Jan M. Deussing
- Molecular Neurogenetics, Max-Planck-Institute of Psychiatry, Munich, Germany
| | - Martin Hrabé de Angelis
- Institute of Experimental Genetics, German Research Center for Environmental Health, Helmholtz Zentrum München, GmbH, Neuherberg/Munich, Germany
| | - Till Roenneberg
- Institute of Medical Psychology, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Fang Zheng
- Institute of Physiology and Pathophysiology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Christian Alzheimer
- Institute of Physiology and Pathophysiology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Alcino Silva
- Department of Neurobiology, University of California Los Angeles, Los Angeles, United States of America
| | - Wolfgang Wurst
- German Research Center for Environmental Health, Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg/Munich, Germany
- Molecular Neurogenetics, Max-Planck-Institute of Psychiatry, Munich, Germany
- Lehrstuhl für Entwicklungsgenetik, Technical University München-Weihenstephan, Neuherberg/Munich, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen e.V. (DZNE) Site Munich, München, Germany
| | - Ralf Kühn
- German Research Center for Environmental Health, Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg/Munich, Germany
- Lehrstuhl für Entwicklungsgenetik, Technical University München-Weihenstephan, Neuherberg/Munich, Germany
- * E-mail:
| |
Collapse
|
25
|
Zhou A, Zhang S. Regulation of cell signaling and porcine reproductive and respiratory syndrome virus. Cell Signal 2012; 24:973-80. [PMID: 22274732 DOI: 10.1016/j.cellsig.2012.01.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2011] [Revised: 12/26/2011] [Accepted: 01/10/2012] [Indexed: 01/21/2023]
Abstract
In order to successfully survive in host and persistent infection, porcine reproductive and respiratory syndrome virus (PRRSV) utilized sophisticated mechanisms to suppress or escape from the host' innate and adaptive immune systems, and then changed host gene expression. Signaling pathways play a pivotal role in the regulation of diverse biological processes. Once signaling pathways are activated by a variety of different stimuli, immune responses will be triggered by the activation of chemokines, transcription factors, and inflammatory cytokines to adjust the aggressive replication and dissemination of viruses. PRRSV infection is able to get many signaling pathways activation that facilitates distinct cell functions to modulate immune responses. In addition, the cross-talk of cell signaling pathways also can regulate PRRSV replication and also is present in this review by recent finding.
Collapse
Affiliation(s)
- Ao Zhou
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
| | | |
Collapse
|
26
|
Le Merrer J, Befort K, Gardon O, Filliol D, Darcq E, Dembele D, Becker JAJ, Kieffer BL. Protracted abstinence from distinct drugs of abuse shows regulation of a common gene network. Addict Biol 2012; 17:1-12. [PMID: 21955143 DOI: 10.1111/j.1369-1600.2011.00365.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Addiction is a chronic brain disorder. Prolonged abstinence from drugs of abuse involves dysphoria, high stress responsiveness and craving. The neurobiology of drug abstinence, however, is poorly understood. We previously identified a unique set of hundred mu-opioid receptor-dependent genes in the extended amygdala, a key site for hedonic and stress processing in the brain. Here we examined these candidate genes either immediately after chronic morphine, nicotine, Δ9-tetrahydrocannabinol or alcohol, or following 4 weeks of abstinence. Regulation patterns strongly differed among chronic groups. In contrast, gene regulations strikingly converged in the abstinent groups and revealed unforeseen common adaptations within a novel huntingtin-centered molecular network previously unreported in addiction research. This study demonstrates that, regardless the drug, a specific set of transcriptional regulations develops in the abstinent brain, which possibly contributes to the negative affect characterizing protracted abstinence. This transcriptional signature may represent a hallmark of drug abstinence and a unitary adaptive molecular mechanism in substance abuse disorders.
Collapse
Affiliation(s)
- Julie Le Merrer
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, INSERM and CNRS, Illkirch-Graffenstaden, France
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Ibrahim BM, Abdel-Rahman AA. Differential modulation of brainstem phosphatidylinositol 3-kinase/Akt and extracellular signal-regulated kinase 1/2 signaling underlies WIN55,212-2 centrally mediated pressor response in conscious rats. J Pharmacol Exp Ther 2012; 340:11-8. [PMID: 21946192 PMCID: PMC3251017 DOI: 10.1124/jpet.111.186858] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Accepted: 09/23/2011] [Indexed: 02/01/2023] Open
Abstract
Our recent study demonstrated that central cannabinoid receptor 1 (CB₁R) activation caused dose-related pressor response in conscious rats, and reported studies implicated the brainstem phosphatidylinositol 3-kinase (PI3K)/Akt-extracellular signal-regulated kinase 1/2 (ERK1/2) pathway in blood pressure control. Therefore, in this study, we tested the hypothesis that the modulation of brainstem PI3K/Akt-ERK1/2 signaling plays a critical role in the central CB(1)R-mediated pressor response. In conscious freely moving rats, the pressor response elicited by intracisternal (i.c.) (R)-(+)-[2,3-dihydro-5-methyl-3[(4-morpholinyl)methyl]pyrrolo[1,2,3-de]-1,4-benzoxazinyl]-(1-naphthalenyl) methanone mesylate salt (WIN55,212-2) (15 μg) was associated with significant increases in ERK1/2 phosphorylation in the rostral ventrolateral medulla (RVLM) and the nucleus tractus solitarius (NTS). In contrast, Akt phosphorylation was significantly reduced in the same neuronal pools. Pretreatment with the selective CB₁R antagonist N-(piperidin-1-yl)-5-(4-iodophenyl)-1-(2,4-dichlorophenyl)-4-methyl-1H-pyrazole-3-carboxamide (AM251) (30 μg i.c.) attenuated the neurochemical responses elicited by central CB₁R activation. Furthermore, pretreatment with the ERK/mitogen-activated protein kinase kinase inhibitor 2'-amino-3'-methoxyflavone (PD98059) (5 μg i.c.) abrogated WIN55,212-2-evoked increases in blood pressure and neuronal ERK1/2 phosphorylation but not the reduction in Akt phosphorylation. On the other hand, prior PI3K inhibition with wortmannin (0.4 μg i.c.) exacerbated the WIN55,212-2 (7.5 and 15 μg i.c.) dose-related increases in blood pressure and ERK1/2 phosphorylation in the RVLM. The present neurochemical and integrative studies yield new insight into the critical role of two brainstem kinases, PI3K and ERK1/2, in the pressor response elicited by central CB₁R activation in conscious rats.
Collapse
Affiliation(s)
- Badr Mostafa Ibrahim
- Department of Pharmacology and Toxicology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | | |
Collapse
|
28
|
Bansal P, Wang S, Liu S, Xiang YY, Lu WY, Wang Q. GABA coordinates with insulin in regulating secretory function in pancreatic INS-1 β-cells. PLoS One 2011; 6:e26225. [PMID: 22031825 PMCID: PMC3198728 DOI: 10.1371/journal.pone.0026225] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2011] [Accepted: 09/22/2011] [Indexed: 11/18/2022] Open
Abstract
Pancreatic islet β-cells produce large amounts of γ-aminobutyric acid (GABA), which is co-released with insulin. GABA inhibits glucagon secretion by hyperpolarizing α-cells via type-A GABA receptors (GABA(A)Rs). We and others recently reported that islet β-cells also express GABA(A)Rs and that activation of GABA(A)Rs increases insulin release. Here we investigate the effects of insulin on the GABA-GABA(A)R system in the pancreatic INS-1 cells using perforated-patch recording. The results showed that GABA produces a rapid inward current and depolarizes INS-1 cells. However, pre-treatment of the cell with regular insulin (1 µM) suppressed the GABA-induced current (I(GABA)) by 43%. Zinc-free insulin also suppressed I(GABA) to the same extent of inhibition by regular insulin. The inhibition of I(GABA) occurs within 30 seconds after application of insulin. The insulin-induced inhibition of I(GABA) persisted in the presence of PI3-kinase inhibitor, but was abolished upon inhibition of ERK, indicating that insulin suppresses GABA(A)Rs through a mechanism that involves ERK activation. Radioimmunoassay revealed that the secretion of C-peptide was enhanced by GABA, which was blocked by pre-incubating the cells with picrotoxin (50 µM, p<0.01) and insulin (1 µM, p<0.01), respectively. Together, these data suggest that autocrine GABA, via activation of GABA(A)Rs, depolarizes the pancreatic β-cells and enhances insulin secretion. On the other hand, insulin down-regulates GABA-GABA(A)R signaling presenting a feedback mechanism for fine-tuning β-cell secretion.
Collapse
Affiliation(s)
- Paul Bansal
- Departments of Physiology and Medicine, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Division of Endocrinology and Metabolism, The Keenan Research Centre in the Li Ka-Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Shuanglian Wang
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
| | - Shenghao Liu
- Departments of Physiology and Medicine, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Division of Endocrinology and Metabolism, The Keenan Research Centre in the Li Ka-Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Yun-Yan Xiang
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
| | - Wei-Yang Lu
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
- * E-mail: (W-YL); (QW)
| | - Qinghua Wang
- Departments of Physiology and Medicine, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Division of Endocrinology and Metabolism, The Keenan Research Centre in the Li Ka-Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada
- * E-mail: (W-YL); (QW)
| |
Collapse
|
29
|
Kang SU, Heo S, Lubec G. Mass spectrometric analysis of GABAA receptor subtypes and phosphorylations from mouse hippocampus. Proteomics 2011; 11:2171-81. [PMID: 21538884 DOI: 10.1002/pmic.201000374] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2010] [Revised: 02/02/2011] [Accepted: 02/21/2011] [Indexed: 11/05/2022]
Abstract
The brain GABA(A) receptor (GABA(A) R) is a key element of signaling and neural transmission in health and disease. Recently, complete sequence analysis of the recombinant GABA(A) R has been reported, separation and mass spectrometrical (MS) characterisation from tissue, however, has not been published so far. Hippocampi were homogenised, put on a sucrose gradient 10-69% and the layer from 10 to 20% was used for extraction of membrane proteins by a solution of Triton X-100, 1.5 M aminocaproic acid in the presence of 0.3 M Bis-Tris. This mixture was subsequently loaded onto blue native PAGE (BN-PAGE) with subsequent analysis on denaturing gel systems. Spots from the 3-DE electrophoretic run were stained with Colloidal Coomassie Brilliant Blue, and spots with an apparent molecular weight between 40 and 60 kDa were picked and in-gel digested with trypsin, chymotrypsin and subtilisin. The resulting peptides were analysed by nano-LC-ESI-MS/MS (ion trap) and protein identification was carried out using MASCOT searches. In addition, known GABA(A) R-specific MS information taken from own previous studies was used for searches of GABA(A) R subunits. β-1, β-2 and β-3, θ and ρ-1 subunits were detected and six novel phosphorylation sites were observed and verified by phosphatase treatment. The method used herein enables identification of several GABA(A) R subunits from mouse hippocampus along with phosphorylations of β-1 (T227, Y230), β-2 (Y215, T439) and β-3 (T282, S406) subunits. The procedure forms the basis for GABA(A) R studies at the protein chemical rather than at the immunochemical level in health and disease.
Collapse
Affiliation(s)
- Sung Ung Kang
- Division of Neuroproteomics, Department of Pediatrics, Medical University of Vienna, Waehringer Guertel, Vienna, Austria
| | | | | |
Collapse
|
30
|
The involvement of GABA(A) receptor in the molecular mechanisms of combined selective serotonin reuptake inhibitor-antipsychotic treatment. Int J Neuropsychopharmacol 2011; 14:143-55. [PMID: 20181299 DOI: 10.1017/s1461145710000106] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
There is evidence that combining selective serotonin reuptake inhibitor (SSRI) antidepressant and antipsychotic drugs may improve negative symptoms in schizophrenia and resistant symptoms in obsessive-compulsive and affective disorders. To examine the mechanism of action of this treatment we investigated the molecular modulation of γ-aminobutyric acid-A (GABA(A)) receptor components and biochemical pathways associated with GABA(A) receptor function following administration of the SSRI fluvoxamine (Flu) combined with the first-generation antipsychotic haloperidol (Hal) and compared it to the individual drugs and the atypical antipsychotic clozapine (Clz). We analysed prefrontal cortices of Sprague-Dawley rats injected intraperitoneally (i.p.) with the combination of Flu (10 mg/kg) and Hal (1 mg/kg), each drug alone, or Clz (10 mg/kg) after 30 min and 1 h. We found that haloperidol plus fluvoxamine (Hal-Flu) co-administration, and Clz, decreased the level of GABAAβ2/3 receptor subunit in the cytosolic fraction, and increased it in the membrane compartment in rat PFC. Flu or Hal alone did not produce changes in GABAAβ2/3 receptor protein expression. Additionally, Hal-Flu and Clz regulated molecular signalling pathways that modulate GABA(A) receptor function, including protein kinase C (PKC) and extracellular signal-regulated kinase-2 (ERK2). In primary cortical culture, short-term treatment (15 min) with Hal-Flu combination and Clz increased GABAAβ subunit phosphorylation levels. Pretreatment of the cells with PKC inhibitor abolished the effect of the combined treatment, or Clz on phosphorylation of GABA(A) receptor. Inhibition of ERK2 did not alter the effect of drugs on GABA(A) receptor phosphorylation levels. Our findings provide evidence that the combined treatment regulates GABA(A) receptor function and does so via a PKC-dependent pathway.
Collapse
|
31
|
Iñiguez SD, Vialou V, Warren BL, Cao JL, Alcantara LF, Davis LC, Manojlovic Z, Neve RL, Russo SJ, Han MH, Nestler EJ, Bolaños-Guzmán CA. Extracellular signal-regulated kinase-2 within the ventral tegmental area regulates responses to stress. J Neurosci 2010; 30:7652-63. [PMID: 20519540 PMCID: PMC2895424 DOI: 10.1523/jneurosci.0951-10.2010] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2010] [Revised: 04/09/2010] [Accepted: 04/15/2010] [Indexed: 12/12/2022] Open
Abstract
Neurotrophic factors and their signaling pathways have been implicated in the neurobiological adaptations in response to stress and the regulation of mood-related behaviors. A candidate signaling molecule implicated in mediating these cellular responses is the extracellular signal-regulated kinase (ERK1/2), although its functional role in mood regulation remains to be fully elucidated. Here we show that acute (1 d) or chronic (4 weeks) exposure to unpredictable stress increases phosphorylation of ERK1/2 and of two downstream targets (ribosomal S6 kinase and mitogen- and stress-activated protein kinase 1) within the ventral tegmental area (VTA), an important substrate for motivated behavior and mood regulation. Using herpes simplex virus-mediated gene transfer to assess the functional significance of this ERK induction, we show that overexpressing ERK2 within the VTA increases susceptibility to stress as measured in the forced swim test, responses to unconditioned nociceptive stimuli, and elevated plus maze in Sprague Dawley male rats, and in the tail suspension test and chronic social defeat stress procedure in C57BL/6 male mice. In contrast, blocking ERK2 activity in the VTA produces stress-resistant behavioral responses in these same assays and also blocks a chronic stress-induced reduction in sucrose preference. The effects induced by ERK2 blockade were accompanied by decreases in the firing frequency of VTA dopamine neurons, an important electrophysiological hallmark of resilient-like behavior. Together, these results strongly implicate a role for ERK2 signaling in the VTA as a key modulator of responsiveness to stress and mood-related behaviors.
Collapse
MESH Headings
- Action Potentials/physiology
- Analysis of Variance
- Animals
- Animals, Genetically Modified
- Behavior, Animal/physiology
- Dominance-Subordination
- Electroshock/adverse effects
- Escape Reaction/physiology
- Food Preferences/physiology
- Gene Expression Regulation, Enzymologic/physiology
- Green Fluorescent Proteins/genetics
- Hindlimb Suspension/methods
- In Vitro Techniques
- Male
- Maze Learning/physiology
- Mice
- Mice, Inbred C57BL
- Mitogen-Activated Protein Kinase 1/genetics
- Mitogen-Activated Protein Kinase 1/metabolism
- Motor Activity
- Neurons/physiology
- Oncogene Proteins, Fusion
- Pain/enzymology
- Pain/etiology
- Pain/pathology
- Phosphorylation/physiology
- Rats
- Rats, Sprague-Dawley
- Receptors, Fibroblast Growth Factor
- Signal Transduction/physiology
- Simplexvirus/physiology
- Stress, Psychological/enzymology
- Stress, Psychological/etiology
- Stress, Psychological/pathology
- Sucrose/administration & dosage
- Sweetening Agents/administration & dosage
- Swimming/psychology
- Time Factors
- Transduction, Genetic/methods
- Tyrosine 3-Monooxygenase/metabolism
- Ventral Tegmental Area/enzymology
- Ventral Tegmental Area/pathology
Collapse
Affiliation(s)
- Sergio D. Iñiguez
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, Florida 32306-4301
| | | | - Brandon L. Warren
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, Florida 32306-4301
| | - Jun-Li Cao
- Department of Pharmacology and Systems Therapeutics, Mount Sinai School of Medicine, New York, New York 10029-6574, and
| | - Lyonna F. Alcantara
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, Florida 32306-4301
| | - Lindsey C. Davis
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, Florida 32306-4301
| | - Zarko Manojlovic
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, Florida 32306-4301
| | - Rachael L. Neve
- Massachusetts Institute of Technology, Cambridge, Massachusetts 02139-4307
| | | | - Ming-Hu Han
- Fishberg Department of Neuroscience and
- Department of Pharmacology and Systems Therapeutics, Mount Sinai School of Medicine, New York, New York 10029-6574, and
| | | | - Carlos A. Bolaños-Guzmán
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, Florida 32306-4301
| |
Collapse
|
32
|
Blockage of A2A and A3 adenosine receptors decreases the desensitization of human GABA(A) receptors microtransplanted to Xenopus oocytes. Proc Natl Acad Sci U S A 2009; 106:15927-31. [PMID: 19721003 DOI: 10.1073/pnas.0907324106] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
We previously found that the endogenous anticonvulsant adenosine, acting through A(2A) and A(3) adenosine receptors (ARs), alters the stability of currents (I(GABA)) generated by GABA(A) receptors expressed in the epileptic human mesial temporal lobe (MTLE). Here we examined whether ARs alter the stability (desensitization) of I(GABA) expressed in focal cortical dysplasia (FCD) and in periglioma epileptic tissues. The experiments were performed with tissues from 23 patients, using voltage-clamp recordings in Xenopus oocytes microinjected with membranes isolated from human MTLE and FCD tissues or using patch-clamp recordings of pyramidal neurons in epileptic tissue slices. On repetitive activation, the epileptic GABA(A) receptors revealed instability, manifested by a large I(GABA) rundown, which in most of the oocytes (approximately 70%) was obviously impaired by the new A(2A) antagonists ANR82, ANR94, and ANR152. In most MTLE tissue-microtransplanted oocytes, a new A(3) receptor antagonist (ANR235) significantly improved I(GABA) stability. Moreover, patch-clamped pyramidal neurons from human neocortical slices of periglioma epileptic tissues exhibited altered I(GABA) rundown on ANR94 treatment. Our findings indicate that antagonizing A(2A) and A(3) receptors increases the I(GABA) stability in different epileptic tissues and suggest that adenosine derivatives may offer therapeutic opportunities in various forms of human epilepsy.
Collapse
|
33
|
Peng J, Wagle M, Mueller T, Mathur P, Lockwood BL, Bretaud S, Guo S. Ethanol-modulated camouflage response screen in zebrafish uncovers a novel role for cAMP and extracellular signal-regulated kinase signaling in behavioral sensitivity to ethanol. J Neurosci 2009; 29:8408-18. [PMID: 19571131 PMCID: PMC2722107 DOI: 10.1523/jneurosci.0714-09.2009] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2009] [Revised: 05/11/2009] [Accepted: 05/25/2009] [Indexed: 11/21/2022] Open
Abstract
Ethanol, a widely abused substance, elicits evolutionarily conserved behavioral responses in a concentration-dependent manner in vivo. The molecular mechanisms underlying such behavioral sensitivity to ethanol are poorly understood. While locomotor-based behavioral genetic screening is successful in identifying genes in invertebrate models, such complex behavior-based screening has proven difficult for recovering genes in vertebrates. Here we report a novel and tractable ethanol response in zebrafish. Using this ethanol-modulated camouflage response as a screening assay, we have identified a zebrafish mutant named fantasma (fan), which displays reduced behavioral sensitivity to ethanol. Positional cloning reveals that fan encodes type 5 adenylyl cyclase (AC5). fan/ac5 is required to maintain the phosphorylation of extracellular signal-regulated kinase (ERK) in the forebrain structures, including the telencephalon and hypothalamus. Partial inhibition of phosphorylation of ERK in wild-type zebrafish mimics the reduction in sensitivity to stimulatory effects of ethanol observed in the fan mutant, whereas, strikingly, strong inhibition of phosphorylation of ERK renders a stimulatory dose of ethanol sedating. Since previous studies in Drosophila and mice show a role of cAMP signaling in suppressing behavioral sensitivity to ethanol, our findings reveal a novel, isoform-specific role of AC signaling in promoting ethanol sensitivity, and suggest that the phosphorylation level of the downstream effector ERK is a critical "gatekeeper" of behavioral sensitivity to ethanol.
Collapse
Affiliation(s)
- Jisong Peng
- Department of Biopharmaceutical Sciences, Programs in Human Genetics and Biological Sciences, The Wheeler Center for the Neurobiology of Addiction, University of California, San Francisco, San Francisco, California 94143-2811
| | - Mahendra Wagle
- Department of Biopharmaceutical Sciences, Programs in Human Genetics and Biological Sciences, The Wheeler Center for the Neurobiology of Addiction, University of California, San Francisco, San Francisco, California 94143-2811
| | - Thomas Mueller
- Department of Biopharmaceutical Sciences, Programs in Human Genetics and Biological Sciences, The Wheeler Center for the Neurobiology of Addiction, University of California, San Francisco, San Francisco, California 94143-2811
| | - Priya Mathur
- Department of Biopharmaceutical Sciences, Programs in Human Genetics and Biological Sciences, The Wheeler Center for the Neurobiology of Addiction, University of California, San Francisco, San Francisco, California 94143-2811
| | - Brent L. Lockwood
- Department of Biopharmaceutical Sciences, Programs in Human Genetics and Biological Sciences, The Wheeler Center for the Neurobiology of Addiction, University of California, San Francisco, San Francisco, California 94143-2811
| | - Sandrine Bretaud
- Department of Biopharmaceutical Sciences, Programs in Human Genetics and Biological Sciences, The Wheeler Center for the Neurobiology of Addiction, University of California, San Francisco, San Francisco, California 94143-2811
| | - Su Guo
- Department of Biopharmaceutical Sciences, Programs in Human Genetics and Biological Sciences, The Wheeler Center for the Neurobiology of Addiction, University of California, San Francisco, San Francisco, California 94143-2811
| |
Collapse
|
34
|
Roseti C, Martinello K, Fucile S, Piccari V, Mascia A, Di Gennaro G, Quarato PP, Manfredi M, Esposito V, Cantore G, Arcella A, Simonato M, Fredholm BB, Limatola C, Miledi R, Eusebi F. Adenosine receptor antagonists alter the stability of human epileptic GABAA receptors. Proc Natl Acad Sci U S A 2008; 105:15118-23. [PMID: 18809912 PMCID: PMC2567502 DOI: 10.1073/pnas.0807277105] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2008] [Indexed: 12/26/2022] Open
Abstract
We examined how the endogenous anticonvulsant adenosine might influence gamma-aminobutyric acid type A (GABA(A)) receptor stability and which adenosine receptors (ARs) were involved. Upon repetitive activation (GABA 500 microM), GABA(A) receptors, microtransplanted into Xenopus oocytes from neurosurgically resected epileptic human nervous tissues, exhibited an obvious GABA(A)-current (I(GABA)) run-down, which was consistently and significantly reduced by treatment with the nonselective adenosine receptor antagonist CGS15943 (100 nM) or with adenosine deaminase (ADA) (1 units/ml), that inactivates adenosine. It was also found that selective antagonists of A2B (MRS1706, 10 nM) or A3 (MRS1334, 30 nM) receptors reduced I(GABA) run-down, whereas treatment with the specific A1 receptor antagonist DPCPX (10 nM) was ineffective. The selective A2A receptor antagonist SCH58261 (10 nM) reduced or potentiated I(GABA) run-down in approximately 40% and approximately 20% of tested oocytes, respectively. The ADA-resistant, AR agonist 2-chloroadenosine (2-CA) (10 microM) potentiated I(GABA) run-down but only in approximately 20% of tested oocytes. CGS15943 administration again decreased I(GABA) run-down in patch-clamped neurons from either human or rat neocortex slices. I(GABA) run-down in pyramidal neurons was equivalent in A1 receptor-deficient and wt neurons but much larger in neurons from A2A receptor-deficient mice, indicating that, in mouse cortex, GABA(A)-receptor stability is tonically influenced by A2A but not by A1 receptors. I(GABA) run-down from wt mice was not affected by 2-CA, suggesting maximal ARs activity by endogenous adenosine. Our findings strongly suggest that cortical A2-A3 receptors alter the stability of GABA(A) receptors, which could offer therapeutic opportunities.
Collapse
Affiliation(s)
- Cristina Roseti
- *Istituto Pasteur–Fondazione Cenci Bolognetti and Dipartimento di Fisiologia Umana e Farmacologia, Centro di Eccellenza di Biologia e Medicina Molecolare, Università di Roma “La Sapienza”, Piazzale A. Moro 5, I-00185 Rome, Italy
| | | | - Sergio Fucile
- *Istituto Pasteur–Fondazione Cenci Bolognetti and Dipartimento di Fisiologia Umana e Farmacologia, Centro di Eccellenza di Biologia e Medicina Molecolare, Università di Roma “La Sapienza”, Piazzale A. Moro 5, I-00185 Rome, Italy
- Neuromed I.R.C.C.S., Via Atinese 18, I-86077 Isernia, Italy
| | - Vanessa Piccari
- *Istituto Pasteur–Fondazione Cenci Bolognetti and Dipartimento di Fisiologia Umana e Farmacologia, Centro di Eccellenza di Biologia e Medicina Molecolare, Università di Roma “La Sapienza”, Piazzale A. Moro 5, I-00185 Rome, Italy
- Neuromed I.R.C.C.S., Via Atinese 18, I-86077 Isernia, Italy
| | | | | | | | - Mario Manfredi
- Neuromed I.R.C.C.S., Via Atinese 18, I-86077 Isernia, Italy
| | | | | | | | - Michele Simonato
- Department of Clinical and Experimental Medicine, Section of Pharmacology, Neuroscience Center, University of Ferrara and National Institute of Neuroscience, I-44100 Ferrara, Italy
| | - Bertil B. Fredholm
- Department of Physiology and Pharmacology, Karolinska Institutet, S-171 77 Stockholm, Sweden
| | - Cristina Limatola
- *Istituto Pasteur–Fondazione Cenci Bolognetti and Dipartimento di Fisiologia Umana e Farmacologia, Centro di Eccellenza di Biologia e Medicina Molecolare, Università di Roma “La Sapienza”, Piazzale A. Moro 5, I-00185 Rome, Italy
- Neuromed I.R.C.C.S., Via Atinese 18, I-86077 Isernia, Italy
| | - Ricardo Miledi
- **Instituto de Neurobiología, Campus UNAM-Juriquilla, Universidad Nacional Autónoma de México, AP1-1141 Querétaro, Mexico; and
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697-4550
| | - Fabrizio Eusebi
- *Istituto Pasteur–Fondazione Cenci Bolognetti and Dipartimento di Fisiologia Umana e Farmacologia, Centro di Eccellenza di Biologia e Medicina Molecolare, Università di Roma “La Sapienza”, Piazzale A. Moro 5, I-00185 Rome, Italy
- Neuromed I.R.C.C.S., Via Atinese 18, I-86077 Isernia, Italy
| |
Collapse
|
35
|
Williams DB. A novel, rapid, inhibitory effect of insulin on alpha1beta2gamma2s gamma-aminobutyric acid type A receptors. Neurosci Lett 2008; 443:27-31. [PMID: 18672028 PMCID: PMC2597579 DOI: 10.1016/j.neulet.2008.07.038] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2008] [Revised: 07/09/2008] [Accepted: 07/16/2008] [Indexed: 10/21/2022]
Abstract
In the CNS, GABA and insulin seem to contribute to similar processes, including neuronal survival; learning and reward; and energy balance and food intake. It is likely then that insulin and GABA may interact, perhaps at the GABA(A) receptor. One such interaction has already been described [Q. Wan, Z.G. Xiong, H.Y. Man, C.A. Ackerley, J. Braunton, W.Y. Lu, L.E. Becker, J.F. MacDonald, Y.T. Wang, Recruitment of functional GABA(A) receptors to postsynaptic domains by insulin, Nature 388 (1997) 686-690]; in it a micromolar concentration of insulin causes the insertion of GABA(A) receptors into the cell membrane, increasing GABA current. I have discovered another effect of insulin on GABA(A) currents. Using a receptor isoform, alpha(1)beta(2)gamma(2s) that is the likely main neuronal GABA(A) isoform expressed recombinantly in Xenopus oocytes, insulin inhibits GABA-induced current when applied simultaneously with low concentrations of GABA. Insulin will significantly inhibit currents induced by EC(30-50) concentrations of GABA by about 38%. Insulin is potent in this effect; IC(50) of insulin was found to be about 4.3 x 10(-10) M. The insulin effect on the GABA dose responses looked like that of an antagonist similar to bicuculline or beta-carbolines. However, an effect of phosphorylation on the GABA(A) receptor from the insulin receptor signal transduction pathway cannot yet be dismissed.
Collapse
Affiliation(s)
- Daniel B Williams
- Department of Life Sciences, Winston-Salem St. University, 601 Martin Luther King Jr Dr, WBA 402, Winston-Salem, NC 27110, United States.
| |
Collapse
|