1
|
Jung SH, Olsen LK, Jones KA, Moore RJ, Harshman SW, Hatcher-Solis CN. VNS paired with training enhances recognition memory: mechanistic insights from proteomic analysis of the hippocampal synapse. Front Mol Neurosci 2024; 17:1452327. [PMID: 39741691 PMCID: PMC11685747 DOI: 10.3389/fnmol.2024.1452327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 08/27/2024] [Indexed: 01/03/2025] Open
Abstract
Introduction Recognition memory, an essential component of cognitive health, can suffer from biological limitations of stress, aging, or neurodegenerative disease. Vagus nerve stimulation (VNS) is a neuromodulation therapy with the potential to improve cognitive function. This study investigated the effectiveness of multiple sessions of VNS to enhance recognition memory in healthy rodents and the underlying cognitive benefits of VNS by proteomic analysis of the synaptosome. Methods Rats demonstrated VNS-induced recognition memory improvements using a novel object recognition (NOR) task. Using the LC-MS/MS method, roughly 3,000 proteins in the synaptosome of the hippocampus were analyzed. Results Protein-protein interaction (PPI) enrichment analysis found differentially expressed proteins related to synaptic signaling and neurotransmitter pathways. PPI network analysis identified six unique protein clusters, including a cluster of synaptic signaling related pathways. Using ingenuity pathway analysis (IPA), rapamycin-insensitive companion of mTOR was identified as an upstream regulator of synaptosome changes due to VNS-paired training. Discussion Based on these results, it is proposed that VNS may mediate cognitive enhancement via increases in glutamatergic signaling and early LTP during the consolidation period, followed by sustained synaptic plasticity via modified post-synaptic receptor expression and dendritic outgrowth. Further investigation is required to determine if VNS is a good candidate to ameliorate cognitive impairment.
Collapse
Affiliation(s)
- Seung H. Jung
- Cognitive Neuroscience, 711th Human Performance Wing, Air Force Research Laboratory, Wright-Patterson AFB, OH, United States
- DCS Infoscitex, Dayton, OH, United States
| | - Laura K. Olsen
- Cognitive Neuroscience, 711th Human Performance Wing, Air Force Research Laboratory, Wright-Patterson AFB, OH, United States
- Oak Ridge Institute for Science and Education, Oak Ridge, TN, United States
| | - Krysten A. Jones
- Cognitive Neuroscience, 711th Human Performance Wing, Air Force Research Laboratory, Wright-Patterson AFB, OH, United States
- Integrative Health & Performance Sciences, UES, Inc., Blue Halo, Dayton, OH, United States
| | - Raquel J. Moore
- Cognitive Neuroscience, 711th Human Performance Wing, Air Force Research Laboratory, Wright-Patterson AFB, OH, United States
- DCS Infoscitex, Dayton, OH, United States
| | - Sean W. Harshman
- Analytical Chemistry, 711th Human Performance Wing, Air Force Research Laboratory, Wright-Patterson AFB, OH, United States
| | - Candice N. Hatcher-Solis
- Cognitive Neuroscience, 711th Human Performance Wing, Air Force Research Laboratory, Wright-Patterson AFB, OH, United States
| |
Collapse
|
2
|
Szabo B. Presynaptic Adrenoceptors. Handb Exp Pharmacol 2024; 285:185-245. [PMID: 38755350 DOI: 10.1007/164_2024_714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2024]
Abstract
Presynaptic α2-adrenoceptors are localized on axon terminals of many noradrenergic and non-noradrenergic neurons in the peripheral and central nervous systems. Their activation by exogenous agonists leads to inhibition of the exocytotic release of noradrenaline and other transmitters from the neurons. Most often, the α2A-receptor subtype is involved in this inhibition. The chain of molecular events between receptor occupation and inhibition of the exocytotic release of transmitters has been determined. Physiologically released endogenous noradrenaline elicits retrograde autoinhibition of its own release. Some clonidine-like α2-receptor agonists have been used to treat hypertension. Dexmedetomidine is used for prolonged sedation in the intensive care; It also has a strong analgesic effect. The α2-receptor antagonist mirtazapine increases the noradrenaline concentration in the synaptic cleft by interrupting physiological autoinhibion of release. It belongs to the most effective antidepressive drugs. β2-Adrenoceptors are also localized on axon terminals in the peripheral and central nervous systems. Their activation leads to enhanced transmitter release, however, they are not activated by endogenous adrenaline.
Collapse
MESH Headings
- Animals
- Humans
- Receptors, Adrenergic, alpha-2/metabolism
- Receptors, Adrenergic, alpha-2/drug effects
- Receptors, Adrenergic, alpha-2/physiology
- Presynaptic Terminals/drug effects
- Presynaptic Terminals/metabolism
- Adrenergic alpha-2 Receptor Agonists/pharmacology
- Receptors, Presynaptic/drug effects
- Receptors, Presynaptic/physiology
- Receptors, Presynaptic/metabolism
- Synaptic Transmission/drug effects
- Receptors, Adrenergic, beta-2/metabolism
- Receptors, Adrenergic, beta-2/drug effects
Collapse
Affiliation(s)
- Bela Szabo
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany.
| |
Collapse
|
3
|
Theriault JE, Shaffer C, Dienel GA, Sander CY, Hooker JM, Dickerson BC, Barrett LF, Quigley KS. A functional account of stimulation-based aerobic glycolysis and its role in interpreting BOLD signal intensity increases in neuroimaging experiments. Neurosci Biobehav Rev 2023; 153:105373. [PMID: 37634556 PMCID: PMC10591873 DOI: 10.1016/j.neubiorev.2023.105373] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 07/28/2023] [Accepted: 08/23/2023] [Indexed: 08/29/2023]
Abstract
In aerobic glycolysis, oxygen is abundant, and yet cells metabolize glucose without using it, decreasing their ATP per glucose yield by 15-fold. During task-based stimulation, aerobic glycolysis occurs in localized brain regions, presenting a puzzle: why produce ATP inefficiently when, all else being equal, evolution should favor the efficient use of metabolic resources? The answer is that all else is not equal. We propose that a tradeoff exists between efficient ATP production and the efficiency with which ATP is spent to transmit information. Aerobic glycolysis, despite yielding little ATP per glucose, may support neuronal signaling in thin (< 0.5 µm), information-efficient axons. We call this the efficiency tradeoff hypothesis. This tradeoff has potential implications for interpretations of task-related BOLD "activation" observed in fMRI. We hypothesize that BOLD "activation" may index local increases in aerobic glycolysis, which support signaling in thin axons carrying "bottom-up" information, or "prediction error"-i.e., the BIAPEM (BOLD increases approximate prediction error metabolism) hypothesis. Finally, we explore implications of our hypotheses for human brain evolution, social behavior, and mental disorders.
Collapse
Affiliation(s)
- Jordan E Theriault
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA; Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA.
| | - Clare Shaffer
- Northeastern University, Department of Psychology, Boston, MA, USA
| | - Gerald A Dienel
- Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, AR, USA; Department of Cell Biology and Physiology, University of New Mexico, Albuquerque, NM, USA
| | - Christin Y Sander
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA; Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Jacob M Hooker
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA; Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Bradford C Dickerson
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA; Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA; Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Lisa Feldman Barrett
- Northeastern University, Department of Psychology, Boston, MA, USA; Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA; Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Karen S Quigley
- Northeastern University, Department of Psychology, Boston, MA, USA; VA Bedford Healthcare System, Bedford, MA, USA
| |
Collapse
|
4
|
Li R, Wang Y, Yang Y, Wu Z, Wang L, Tang G, Yang J, Liu J. The α1 and γ2 subunit-containing GABA A receptor-mediated inhibitory transmission in the anteroventral bed nucleus of stria terminalis is involved in the regulation of anxiety in rats with substantia nigra lesions. Neuropharmacology 2023:109645. [PMID: 37392819 DOI: 10.1016/j.neuropharm.2023.109645] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 06/08/2023] [Accepted: 06/22/2023] [Indexed: 07/03/2023]
Abstract
The anteroventral bed nucleus of the stria terminalis (avBNST) is widely acknowledged as a key brain structure that regulates negative emotional states, such as anxiety. At present, it is still unclear whether GABAA receptor-mediated inhibitory transmission in the avBNST is involved in Parkinson's disease (PD)-related anxiety. In this study, unilateral 6-hydroxydopamine (6-OHDA) lesions of the substantia nigra pars compacta (SNc) in rats induced anxiety-like behaviors, increased GABA synthesis and release, and upregulated expression of GABAA receptor subunits in the avBNST, as well as decreased level of dopamine (DA) in the basolateral amygdala (BLA). In both sham and 6-OHDA rats, intra-avBNST injection of GABAA receptor agonist muscimol induced the following changes: (i) anxiolytic-like responses, (ii) inhibition of the firing activity of GABAergic neurons in the avBNST, (iii) excitation of dopaminergic neurons in the ventral tegmental area (VTA) and serotonergic neurons in the dorsal raphe nucleus (DRN), and (iv) increase of DA and 5-HT release in the BLA, whereas antagonist bicuculline induced the opposite effects. Collectively, these findings suggest that degeneration of the nigrostriatal pathway enhances GABAA receptor-mediated inhibitory transmission in the avBNST, which is involved in PD-related anxiety. Further, activation and blockade of avBNST GABAA receptors affect the firing activity of VTA dopaminergic and DRN serotonergic neurons, and then change release of BLA DA and 5-HT, thereby regulating anxiety-like behaviors.
Collapse
Affiliation(s)
- Ruotong Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Yong Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Yaxin Yang
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710004, China
| | - Zhongheng Wu
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710004, China
| | - Ling Wang
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710004, China
| | - Guoyi Tang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Jie Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Jian Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China.
| |
Collapse
|
5
|
Gyawali U, Martin DA, Sun F, Li Y, Calu D. Dopamine in the dorsal bed nucleus of stria terminalis signals Pavlovian sign-tracking and reward violations. eLife 2023; 12:e81980. [PMID: 37232554 PMCID: PMC10219648 DOI: 10.7554/elife.81980] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 05/05/2023] [Indexed: 05/27/2023] Open
Abstract
Midbrain and striatal dopamine signals have been extremely well characterized over the past several decades, yet novel dopamine signals and functions in reward learning and motivation continue to emerge. A similar characterization of real-time sub-second dopamine signals in areas outside of the striatum has been limited. Recent advances in fluorescent sensor technology and fiber photometry permit the measurement of dopamine binding correlates, which can divulge basic functions of dopamine signaling in non-striatal dopamine terminal regions, like the dorsal bed nucleus of the stria terminalis (dBNST). Here, we record GRABDA signals in the dBNST during a Pavlovian lever autoshaping task. We observe greater Pavlovian cue-evoked dBNST GRABDA signals in sign-tracking (ST) compared to goal-tracking/intermediate (GT/INT) rats and the magnitude of cue-evoked dBNST GRABDA signals decreases immediately following reinforcer-specific satiety. When we deliver unexpected rewards or omit expected rewards, we find that dBNST dopamine signals encode bidirectional reward prediction errors in GT/INT rats, but only positive prediction errors in ST rats. Since sign- and goal-tracking approach strategies are associated with distinct drug relapse vulnerabilities, we examined the effects of experimenter-administered fentanyl on dBNST dopamine associative encoding. Systemic fentanyl injections do not disrupt cue discrimination but generally potentiate dBNST dopamine signals. These results reveal multiple dBNST dopamine correlates of learning and motivation that depend on the Pavlovian approach strategy employed.
Collapse
Affiliation(s)
- Utsav Gyawali
- Program in Neuroscience, University of Maryland School of MedicineBaltimoreUnited States
- Department of Anatomy and Neurobiology, University of Maryland School of MedicineBaltimoreUnited States
| | - David A Martin
- Department of Anatomy and Neurobiology, University of Maryland School of MedicineBaltimoreUnited States
| | - Fangmiao Sun
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences; PKU-IDG/McGovern Institute for Brain Research; Peking-Tsinghua Center for Life SciencesBeijingChina
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences; PKU-IDG/McGovern Institute for Brain Research; Peking-Tsinghua Center for Life SciencesBeijingChina
| | - Donna Calu
- Program in Neuroscience, University of Maryland School of MedicineBaltimoreUnited States
- Department of Anatomy and Neurobiology, University of Maryland School of MedicineBaltimoreUnited States
| |
Collapse
|
6
|
Perdikaris P, Dermon CR. Behavioral and neurochemical profile of MK-801 adult zebrafish model: Forebrain β 2-adrenoceptors contribute to social withdrawal and anxiety-like behavior. Prog Neuropsychopharmacol Biol Psychiatry 2022; 115:110494. [PMID: 34896197 DOI: 10.1016/j.pnpbp.2021.110494] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 11/22/2021] [Accepted: 12/03/2021] [Indexed: 01/29/2023]
Abstract
Deficits in social communication and interaction are core clinical symptoms characterizing multiple neuropsychiatric conditions, including autism spectrum disorder (ASD) and schizophrenia. Interestingly, elevated anxiety levels are a common comorbid psychopathology characterizing individuals with aberrant social behavior. Despite recent progress, the underlying neurobiological mechanisms that link anxiety with social withdrawal remain poorly understood. The present study developed a zebrafish pharmacological model displaying social withdrawal behavior, following a 3-h exposure to 4 μΜ (+)-MK-801, a non-competitive N-methyl-d-aspartate (NMDA) receptor antagonist, for 7 days. Interestingly, MK-801-treated zebrafish displayed elevated anxiety levels along with higher frequency of stereotypical behaviors, rendering this zebrafish model appropriate to unravel a possible link of catecholaminergic and ASD-like phenotypes. MK-801-treated zebrafish showed increased telencephalic protein expression of metabotropic glutamate 5 receptor (mGluR5), dopamine transporter (DAT) and β2-adrenergic receptors (β2-ARs), supporting the presence of excitation/inhibition imbalance along with altered dopaminergic and noradrenergic activity. Interestingly, β2-ARs expression, was differentially regulated across the Social Decision-Making (SDM) network nodes, exhibiting increased levels in ventral telencephalic area (Vv), a key-area integrating reward and social circuits but decreased expression in dorso-medial telencephalic area (Dm) and anterior tuberal nucleus (ATN). Moreover, the co-localization of β2-ARs with elements of GABAergic and glutamatergic systems, as well as with GAP-43, a protein indicating increased brain plasticity potential, support the key-role of β2-ARs in the MK-801 zebrafish social dysfunctions. Our results highlight the importance of the catecholaminergic neurotransmission in the manifestation of ASD-like behavior, representing a site of potential interventions for amelioration of ASD-like symptoms.
Collapse
Affiliation(s)
- Panagiotis Perdikaris
- Human and Animal Physiology Laboratory, Department of Biology, University of Patras, Rio, 26500 Patras, Greece
| | - Catherine R Dermon
- Human and Animal Physiology Laboratory, Department of Biology, University of Patras, Rio, 26500 Patras, Greece.
| |
Collapse
|
7
|
Monroy-Jaramillo N, Martínez-Magaña JJ, Pérez-Aldana BE, Ortega-Vázquez A, Montalvo-Ortiz J, López-López M. The role of alcohol intake in the pharmacogenetics of treatment with clozapine. Pharmacogenomics 2022; 23:371-392. [PMID: 35311547 DOI: 10.2217/pgs-2022-0006] [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] [Indexed: 12/12/2022] Open
Abstract
Clozapine (CLZ) is an atypical antipsychotic reserved for patients with refractory psychosis, but it is associated with a significant risk of severe adverse reactions (ADRs) that are potentiated with the concomitant use of alcohol. Additionally, pharmacogenetic studies have explored the influence of several genetic variants in CYP450, receptors and transporters involved in the interindividual response to CLZ. Herein, we systematically review the current multiomics knowledge behind the interaction between CLZ and alcohol intake, and how its concomitant use might modulate the pharmacogenetics. CYP1A2*1F, *1C and other alleles not yet discovered could support a precision medicine approach for better therapeutic effects and fewer CLZ ADRs. CLZ monitoring systems should be amended and include alcohol intake to protect patients from severe CLZ ADRs.
Collapse
Affiliation(s)
- Nancy Monroy-Jaramillo
- Department of Genetics, National Institute of Neurology & Neurosurgery, Manuel Velasco Suárez, La Fama, Tlalpan, Mexico City, 14269, Mexico
| | - José Jaime Martínez-Magaña
- Department of Psychiatry, Division of Human Genetics, Yale University School of Medicine, Orange, West Haven, CT 06477, USA
| | - Blanca Estela Pérez-Aldana
- Doctorado en Ciencias Biológicas y de la Salud, Metropolitan Autonomous University, Campus Xochimilco, Villa Quietud, Coyoacán, Mexico City, 04960, Mexico
| | - Alberto Ortega-Vázquez
- Metropolitan Autonomous University, Campus Xochimilco, Villa Quietud, Coyoacán, Mexico City, 04960, Mexico
| | - Janitza Montalvo-Ortiz
- Department of Psychiatry, Division of Human Genetics, Yale University School of Medicine, Orange, West Haven, CT 06477, USA
| | - Marisol López-López
- Metropolitan Autonomous University, Campus Xochimilco, Villa Quietud, Coyoacán, Mexico City, 04960, Mexico
| |
Collapse
|
8
|
Snyder AE, Silberman Y. Corticotropin releasing factor and norepinephrine related circuitry changes in the bed nucleus of the stria terminalis in stress and alcohol and substance use disorders. Neuropharmacology 2021; 201:108814. [PMID: 34624301 PMCID: PMC8578398 DOI: 10.1016/j.neuropharm.2021.108814] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 09/12/2021] [Accepted: 09/24/2021] [Indexed: 12/18/2022]
Abstract
Alcohol Use Disorder (AUD) affects around 14.5 million individuals in the United States, with Substance Use Disorder (SUD) affecting an additional 8.3 million individuals. Relapse is a major barrier to effective long-term treatment of this illness with stress often described as a key trigger for a person with AUD or SUD to relapse during a period of abstinence. Two signaling molecules, norepinephrine (NE) and corticotropin releasing factor (CRF), are released during the stress response, and also play important roles in reward behaviors and the addiction process. Within the addiction literature, one brain region in which there has been increasing research focus in recent years is the bed nucleus of the stria terminalis (BNST). The BNST is a limbic structure with numerous cytoarchitecturally and functionally different subregions that has been implicated in drug-seeking behaviors and stress responses. This review focuses on drug and stress-related neurocircuitry changes in the BNST, particularly within the CRF and NE systems, with an emphasis on differences and similarities between the major dorsal and ventral BNST subregions.
Collapse
Affiliation(s)
- Angela E Snyder
- Penn State College of Medicine, Department of Neural and Behavioral Sciences, USA
| | - Yuval Silberman
- Penn State College of Medicine, Department of Neural and Behavioral Sciences, USA.
| |
Collapse
|
9
|
Gururajan A, Bastiaanssen TFS, Ventura Silva AP, Moloney GM, Cryan JF. The impact of psychosocial defeat stress on the bed nucleus of the stria terminalis transcriptome in adult male mice. Eur J Neurosci 2021; 55:67-77. [PMID: 34904308 DOI: 10.1111/ejn.15567] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 11/07/2021] [Accepted: 12/05/2021] [Indexed: 01/07/2023]
Abstract
The bed nucleus of the stria terminalis (BNST) is a focal point for the convergence of inputs from canonical stress-sensitive structures to fine-tune the response to stress. However, its role in mediating phenotypes of stress resilience or susceptibility is yet to be fully defined. In this study, we carried out unbiased RNA-sequencing to analyse the BNST transcriptomes of adult male mice, which were classified as resilient or susceptible following a 10-day chronic psychosocial defeat stress paradigm. Pairwise comparisons revealed 20 differentially expressed genes in resilience (6) and susceptible (14) mice compared with controls. An in silico validation of our data against an earlier study revealed significant concordance in gene expression profiles associated with resilience to chronic stress. Enrichment analysis revealed that resilience is linked to functions including retinoic acid hydrolase activity, phospholipase inhibitor and tumour necrosis factor (TNF)-receptor activities, whereas susceptibility is linked to alterations in amino acid transporter activity. We also identified differential usage of 134 exons across 103 genes associated with resilience and susceptibility; enrichment analysis for genes with differential exon usage in resilient mice was linked to functions including adrenergic receptor binding mice and oxysterol binding in susceptible mice. Our findings highlight the important and underappreciated role of the BNST in stress resilience and susceptibility and reveal research avenues for follow-up investigations.
Collapse
Affiliation(s)
- Anand Gururajan
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.,APC Microbiome Ireland, University College Cork, Cork, Ireland.,School of Psychology, Faculty of Science, University of Sydney, Sydney, New South Wales, Australia
| | - Thomaz F S Bastiaanssen
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.,APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Ana Paula Ventura Silva
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.,APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Gerard M Moloney
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.,APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - John F Cryan
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.,APC Microbiome Ireland, University College Cork, Cork, Ireland
| |
Collapse
|
10
|
Mather M. Noradrenaline in the aging brain: Promoting cognitive reserve or accelerating Alzheimer's disease? Semin Cell Dev Biol 2021; 116:108-124. [PMID: 34099360 PMCID: PMC8292227 DOI: 10.1016/j.semcdb.2021.05.013] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 05/04/2021] [Accepted: 05/05/2021] [Indexed: 12/19/2022]
Abstract
Many believe that engaging in novel and mentally challenging activities promotes brain health and prevents Alzheimer's disease in later life. However, mental stimulation may also have risks as well as benefits. As neurons release neurotransmitters, they often also release amyloid peptides and tau proteins into the extracellular space. These by-products of neural activity can aggregate into the tau tangle and amyloid plaque signatures of Alzheimer's disease. Over time, more active brain regions accumulate more pathology. Thus, increasing brain activity can have a cost. But the neuromodulator noradrenaline, released during novel and mentally stimulating events, may have some protective effects-as well as some negative effects. Via its inhibitory and excitatory effects on neurons and microglia, noradrenaline sometimes prevents and sometimes accelerates the production and accumulation of amyloid-β and tau in various brain regions. Both α2A- and β-adrenergic receptors influence amyloid-β production and tau hyperphosphorylation. Adrenergic activity also influences clearance of amyloid-β and tau. Furthermore, some findings suggest that Alzheimer's disease increases noradrenergic activity, at least in its early phases. Because older brains clear the by-products of synaptic activity less effectively, increased synaptic activity in the older brain risks accelerating the accumulation of Alzheimer's pathology more than it does in the younger brain.
Collapse
Affiliation(s)
- Mara Mather
- Leonard Davis School of Gerontology, Department of Psychology, & Department of Biomedical Engineering, University of Southern California, 3715 McClintock Ave, Los Angeles, CA 90089, United States.
| |
Collapse
|
11
|
Modulation of the Rat Intergeniculate Leaflet of the Thalamus Network by Norepinephrine. Neuroscience 2021; 469:1-16. [PMID: 34174371 DOI: 10.1016/j.neuroscience.2021.06.027] [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: 04/01/2021] [Revised: 06/11/2021] [Accepted: 06/18/2021] [Indexed: 11/23/2022]
Abstract
Circadian rhythms are regulated by a set of brain structures, one of which is the Intergeniculate Leaflet of the Thalamus (IGL). The most recognised role of the IGL is the integration of a variety of stimuli affecting rhythmicity, such as lighting conditions, received by the eye, or light-independent (non-photic) cues, the information about which is delivered via the activation of the non-specific projections. One of them is the norepinephrinergic system originating in the brainstem Locus Coeruleus (LC). In order to investigate the effect of norepinephrine (NE) on the IGL neurons we have performed ex vivo recordings using the extracellular multi-electrode array technique as well as the intracellular whole-cell patch clamp. Using both agonists and antagonists of specific NE receptor subtypes, we confirmed the presence of functional α1-, α2- and β-adrenergic receptors within the investigated structure, allowing NE to exert multiple types of effects on different IGL neurons, mainly depolarisation of the neurons projecting to the Suprachiasmatic Nuclei - the master circadian pacemaker, and various responses exhibited by the cells creating the connection with the contralateral IGL. Moreover, NE was shown to affect IGL cells both directly and via modulation of the synaptic network, in particular the miniature inhibitory postsynaptic currents. To the best of our knowledge, these are the first studies to confirm the effects of NE on the activity of the IGL network.
Collapse
|
12
|
Lagatta DC, Kuntze LB, Uliana DL, Borges-Assis AB, Resstel LBM. Bed nucleus of the stria terminalis modulates baroreflex cardiac activity: an interaction between alpha-1 receptors and NMDA/nitric oxide pathway. Pflugers Arch 2021; 473:253-271. [PMID: 33140200 DOI: 10.1007/s00424-020-02475-1] [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: 08/07/2020] [Revised: 09/25/2020] [Accepted: 10/06/2020] [Indexed: 10/23/2022]
Abstract
The bed nucleus of the stria terminalis (BNST) is a forebrain structure, involved in the modulation of neuroendocrine, cardiovascular and autonomic responses. One of the responses is baroreflex activity, which consists in a neural mechanism responsible for keeping the blood pressure within a narrow range of variation. It has been reported that blockade of BNST α1-adrenoceptors increased the bradycardic component of baroreflex. In addition, such receptors are able to modulate glutamate release in this structure. Interestingly, BNST NMDA receptor antagonism and neuronal nitric oxide synthase (nNOS) inhibition led to the same effect of the α1-adrenoceptors blockade on baroreflex bradycardic response. Therefore, the hypothesis of the present study is that BNST noradrenergic transmission interacts with NMDA/NO pathway through α1 adrenoceptors to modulate the baroreflex activity. Male Wistar rats had stainless steel guide cannulas bilaterally implanted in the BNST. Subsequently, a catheter was inserted into the femoral artery for cardiovascular recordings, and into the femoral vein for assessing baroreflex activation. Injection of the noradrenaline reuptake inhibitor reboxetine in the BNST did not modify the tachycardic, but significantly decreased the bradycardic component of baroreflex. Administration of an α1, but not an α2 antagonist into the BNST prior to reboxetine prevented this effect. Likewise, previous injection of NMDA/NO pathway blockers inhibited the effect of reboxetine on bradycardic response. In conclusion, it was demonstrated for the first time the existence of an interaction between BNST noradrenergic, glutamatergic and nitrergic neurotransmissions in the modulation of bradycardic baroreflex response.
Collapse
Affiliation(s)
- Davi C Lagatta
- Pharmaceutical Sciences, Food and Nutrition College, Federal University of Mato Grosso do Sul, UFMS, Campo Grande, MS, Brazil
| | - Luciana B Kuntze
- Department of Pharmacology, School of Medicine of Ribeirão Preto, University of São Paulo, Bandeirantes Avenue 3900, Ribeirão Preto, São Paulo, 14049-900, Brazil
| | - Daniela L Uliana
- Department of Pharmacology, School of Medicine of Ribeirão Preto, University of São Paulo, Bandeirantes Avenue 3900, Ribeirão Preto, São Paulo, 14049-900, Brazil
| | - Anna B Borges-Assis
- Department of Pharmacology, School of Medicine of Ribeirão Preto, University of São Paulo, Bandeirantes Avenue 3900, Ribeirão Preto, São Paulo, 14049-900, Brazil
| | - Leonardo B M Resstel
- Department of Pharmacology, School of Medicine of Ribeirão Preto, University of São Paulo, Bandeirantes Avenue 3900, Ribeirão Preto, São Paulo, 14049-900, Brazil.
| |
Collapse
|
13
|
Lustberg D, Tillage RP, Bai Y, Pruitt M, Liles LC, Weinshenker D. Noradrenergic circuits in the forebrain control affective responses to novelty. Psychopharmacology (Berl) 2020; 237:3337-3355. [PMID: 32821984 PMCID: PMC7572912 DOI: 10.1007/s00213-020-05615-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 07/17/2020] [Indexed: 01/02/2023]
Abstract
RATIONALE In rodents, exposure to novel environments elicits initial anxiety-like behavior (neophobia) followed by intense exploration (neophilia) that gradually subsides as the environment becomes familiar. Thus, innate novelty-induced behaviors are useful indices of anxiety and motivation in animal models of psychiatric disease. Noradrenergic neurons are activated by novelty and implicated in exploratory and anxiety-like responses, but the role of norepinephrine (NE) in neophobia has not been clearly delineated. OBJECTIVE We sought to define the role of central NE transmission in neophilic and neophobic behaviors. METHODS We assessed dopamine β-hydroxylase knockout (Dbh -/-) mice lacking NE and their NE-competent (Dbh +/-) littermate controls in neophilic (novelty-induced locomotion; NIL) and neophobic (novelty-suppressed feeding; NSF) behavioral tests with subsequent quantification of brain-wide c-fos induction. We complimented the gene knockout approach with pharmacological interventions. RESULTS Dbh -/- mice exhibited blunted locomotor responses in the NIL task and completely lacked neophobia in the NSF test. Neophobia was rescued in Dbh -/- mice by acute pharmacological restoration of central NE with the synthetic precursor L-3,4-dihydroxyphenylserine (DOPS), and attenuated in control mice by the inhibitory α2-adrenergic autoreceptor agonist guanfacine. Following either NSF or NIL, Dbh -/- mice demonstrated reduced c-fos in the anterior cingulate cortex, medial septum, ventral hippocampus, bed nucleus of the stria terminalis, and basolateral amygdala. CONCLUSION These findings indicate that central NE signaling is required for the expression of both neophilic and neophobic behaviors. Further, we describe a putative noradrenergic novelty network as a potential therapeutic target for treating anxiety and substance abuse disorders.
Collapse
Affiliation(s)
- Daniel Lustberg
- Department of Human Genetics, Emory University, Atlanta, GA, 30322, USA
| | - Rachel P Tillage
- Department of Human Genetics, Emory University, Atlanta, GA, 30322, USA
| | - Yu Bai
- Department of Human Genetics, Emory University, Atlanta, GA, 30322, USA
| | - Molly Pruitt
- University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - L Cameron Liles
- Department of Human Genetics, Emory University, Atlanta, GA, 30322, USA
| | - David Weinshenker
- Department of Human Genetics, Emory University, Atlanta, GA, 30322, USA.
| |
Collapse
|
14
|
Rajbhandari AK, Bakshi VP. Repeated norepinephrine receptor stimulation in the BNST induces sensorimotor gating deficits via corticotropin releasing factor. Neuropharmacology 2020; 172:108090. [PMID: 32360378 DOI: 10.1016/j.neuropharm.2020.108090] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 02/15/2020] [Accepted: 04/02/2020] [Indexed: 11/19/2022]
Abstract
Intense stress precipitates symptoms in disorders such as post-traumatic stress (PTSD) and schizophrenia. Patients with these disorders have dysfunctional sensorimotor gating as indexed by disrupted prepulse inhibition of the startle response (PPI), which refers to decreased startle response when a weak pre-stimulus precedes a startling stimulus. Stress promotes release of norepinephrine (NE) and corticotrophin releasing factor (CRF) within the brain, neurotransmitters that also modulate PPI. We have shown that repeated stress causes sensitization of NE receptors within the basolateral amygdala (BLA) via CRF receptors and promotes long-lasting PPI disruptions and startle abnormalities. The bed nucleus of the stria terminalis (BNST) is another crucial brain region that could be involved in stress-induced alterations in NE and CRF functions to promote PPI changes as this anatomical structure is enriched in CRF and NE receptors that have been shown to regulate each other. We hypothesized that repeated infusions of NE into the BNST would cross-sensitize CRF receptors or vice versa to alter PPI. Separate groups of male Sprague Dawley rats received, CRF (200ng/0.5 μl), NE (20μg/0.5 μl), or vehicle into the BNST, once/day for 3 days and PPI was tested after each infusion. Repeated CRF-or vehicle-treated rats were then challenged with a subthreshold dose of NE (0.3μg/0.5 μl) while repeated NE-treated rats were challenged with CRF (200ng/0.5 μl), and PPI was measured. Surprisingly, initial/repeated CRF or vehicle in the BNST had no effects on PPI. In contrast, initial and repeated NE disrupted PPI. Sub-threshold NE challenge in rats that previously received repeated CRF had no effect on PPI. Interestingly though, intra-BNST challenge dose of CRF significantly disrupted PPI in rats that previously had received repeated NE infusions. Taken together, these results indicate that repeated stress-induced NE release could alter the activity of CRF receptors in the BNST to modulate sensorimotor gating as measured through PPI.
Collapse
Affiliation(s)
- Abha Karki Rajbhandari
- Dept. of Psychiatry and Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI 53719, USA; AKR Is Now at Icahn School of Medicine at Mount Sinai, New York, 10029, USA.
| | - Vaishali P Bakshi
- Dept. of Psychiatry and Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI 53719, USA; AKR Is Now at Icahn School of Medicine at Mount Sinai, New York, 10029, USA
| |
Collapse
|
15
|
Thompson Gray AD, Simonetti J, Adegboye F, Jones CK, Zurawski Z, Hamm HE. Sexual Dimorphism in Stress-induced Hyperthermia in SNAP25Δ3 mice, a mouse model with disabled Gβγ regulation of the exocytotic fusion apparatus. Eur J Neurosci 2020; 52:2815-2826. [PMID: 32449556 DOI: 10.1111/ejn.14836] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 05/14/2020] [Accepted: 05/18/2020] [Indexed: 11/28/2022]
Abstract
Behavioral assays in the mouse can show marked differences between male and female animals of a given genotype. These differences identified in such preclinical studies may have important clinical implications. We recently made a mouse model with impaired presynaptic inhibition through Gβγ-SNARE signaling. Here, we examine the role of sexual dimorphism in the severity of the phenotypes of this model, the SNAP25Δ3 mouse. In males, we already reported that SNAP25Δ3 homozygotes demonstrated phenotypes in motor coordination, nociception, spatial memory and stress processing. We now report that while minimal sexually dimorphic effects were observed for the nociceptive, motor or memory phenotypes, large differences were observed in the stress-induced hyperthermia paradigm, with male SNAP25Δ3 homozygotes exhibiting an increase in body temperature subsequent to handling relative to wild-type littermates, while no such genotype-dependent effect was observed in females. This suggests sexually dimorphic mechanisms of Gβγ-SNARE signaling for stress processing or thermoregulation within the mouse. Second, we examined the effects of heterozygosity with respect to the SNAP25Δ3 mutation. Heterozygote SNAP25Δ3 animals were tested alongside homozygote and wild-type littermates in all of the aforementioned paradigms and displayed phenotypes similar to wild-type animals or an intermediate state. From this, we conclude that the SNAP25Δ3 mutation does not behave in an autosomal dominant manner, but rather displays incomplete dominance for many phenotypes.
Collapse
Affiliation(s)
| | - Justice Simonetti
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Feyisayo Adegboye
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Carrie K Jones
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, USA
| | - Zack Zurawski
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA.,Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, USA
| | - Heidi E Hamm
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
16
|
Papesh MH, Guevara Pinto JD. Spotting rare items makes the brain "blink" harder: Evidence from pupillometry. Atten Percept Psychophys 2019; 81:2635-2647. [PMID: 31222658 PMCID: PMC6858538 DOI: 10.3758/s13414-019-01777-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
In many visual search tasks (e.g., cancer screening, airport baggage inspections), the most serious search targets occur infrequently. As an ironic side effect, when observers finally encounter important objects (e.g., a weapon in baggage), they often fail to notice them, a phenomenon known as the low-prevalence effect (LPE). Although many studies have investigated LPE search errors, we investigated the attentional consequences of successful rare target detection. Using an attentional blink paradigm, we manipulated how often observers encountered the first serial target (T1), then measured its effects on their ability to detect a following target (T2). Across two experiments, we show that the LPE is more than just an inflated miss rate: When observers successfully detected rare targets, they were less likely to spot subsequent targets. Using pupillometry to index locus-coeruleus (LC) mediated attentional engagement, Experiment 2 confirmed that an LC refractory period mediates the attentional blink (`Nieuwenhuis, Gilzenrat, Holmes, & Cohen, 2005, Journal of Experimental Psychology: General, 134[3], 291-307), and that these effects emerge relatively quickly following T1 onset. Moreover, in both behavioral and pupil analyses, we found that detecting low-prevalence targets exacerbates the LC refractory period. Consequences for theories of the LPE are discussed.
Collapse
Affiliation(s)
- Megan H Papesh
- Department of Psychology, Louisiana State University, Baton Rouge, LA, USA.
| | | |
Collapse
|
17
|
Ethanol-induced conditioned place preference and aversion differentially alter plasticity in the bed nucleus of stria terminalis. Neuropsychopharmacology 2019; 44:1843-1854. [PMID: 30795004 PMCID: PMC6785142 DOI: 10.1038/s41386-019-0349-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 02/14/2019] [Accepted: 02/15/2019] [Indexed: 01/11/2023]
Abstract
Contextual cues associated with drugs of abuse, such as ethanol, can trigger craving and drug-seeking behavior. Pavlovian procedures, such as place conditioning, have been widely used to study the rewarding/aversive properties of drugs and the association between environmental cues and drug seeking. Previous research has shown that ethanol as an unconditioned stimulus can induce a strong conditioned place preference (CPP) or aversion (CPA) in rodents. However, the neural mechanisms underlying ethanol-induced reward and aversion have not been thoroughly investigated. The bed nucleus of the stria terminalis (BNST), an integral part of the extended amygdala, is engaged by both rewarding and aversive stimuli and plays a role in ethanol-seeking behavior. Here, we used ex-vivo slice physiology to probe learning-induced synaptic plasticity in the BNST following ethanol-induced CPP and CPA. Male DBA/2 J mice (2-3 months old) were conditioned using previously reported ethanol-induced CPP/CPA procedures. Ethanol-induced CPP was associated with increased neuronal excitability in the ventral BNST (vBNST). Conversely, ethanol-induced CPA resulted in a significant decrease in spontaneous glutamatergic transmission without alterations in GABAergic signaling. Ethanol-CPA also led to a significant increase in the paired-pulse ratio at excitatory synapses, suggestive of a decrease in presynaptic glutamate release. Collectively, these data demonstrate that the vBNST is involved in the modulation of contextual learning associated with both the rewarding and the aversive properties of ethanol in mice.
Collapse
|
18
|
Zurawski Z, Thompson Gray AD, Brady LJ, Page B, Church E, Harris NA, Dohn MR, Yim YY, Hyde K, Mortlock DP, Jones CK, Winder DG, Alford S, Hamm HE. Disabling the Gβγ-SNARE interaction disrupts GPCR-mediated presynaptic inhibition, leading to physiological and behavioral phenotypes. Sci Signal 2019; 12:12/569/eaat8595. [PMID: 30783011 DOI: 10.1126/scisignal.aat8595] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
G protein-coupled receptors (GPCRs) that couple to Gi/o proteins modulate neurotransmission presynaptically by inhibiting exocytosis. Release of Gβγ subunits from activated G proteins decreases the activity of voltage-gated Ca2+ channels (VGCCs), decreasing excitability. A less understood Gβγ-mediated mechanism downstream of Ca2+ entry is the binding of Gβγ to SNARE complexes, which facilitate the fusion of vesicles with the cell plasma membrane in exocytosis. Here, we generated mice expressing a form of the SNARE protein SNAP25 with premature truncation of the C terminus and that were therefore partially deficient in this interaction. SNAP25Δ3 homozygote mice exhibited normal presynaptic inhibition by GABAB receptors, which inhibit VGCCs, but defective presynaptic inhibition by receptors that work directly on the SNARE complex, such as 5-hydroxytryptamine (serotonin) 5-HT1b receptors and adrenergic α2a receptors. Simultaneously stimulating receptors that act through both mechanisms showed synergistic inhibitory effects. SNAP25Δ3 homozygote mice had various behavioral phenotypes, including increased stress-induced hyperthermia, defective spatial learning, impaired gait, and supraspinal nociception. These data suggest that the inhibition of exocytosis by Gi/o-coupled GPCRs through the Gβγ-SNARE interaction is a crucial component of numerous physiological and behavioral processes.
Collapse
Affiliation(s)
- Zack Zurawski
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.,Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | | | - Lillian J Brady
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| | - Brian Page
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Emily Church
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Nicholas A Harris
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | - Michael R Dohn
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| | - Yun Young Yim
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| | - Karren Hyde
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| | - Douglas P Mortlock
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | - Carrie K Jones
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Danny G Winder
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | - Simon Alford
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Heidi E Hamm
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.
| |
Collapse
|
19
|
α 2A-Adrenergic Receptor Activation Decreases Parabrachial Nucleus Excitatory Drive onto BNST CRF Neurons and Reduces Their Activity In Vivo. J Neurosci 2018; 39:472-484. [PMID: 30478032 DOI: 10.1523/jneurosci.1035-18.2018] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 10/18/2018] [Accepted: 11/19/2018] [Indexed: 11/21/2022] Open
Abstract
Stress contributes to numerous psychiatric disorders. Corticotropin releasing factor (CRF) signaling and CRF neurons in the bed nucleus of the stria terminalis (BNST) drive negative affective behaviors, thus agents that decrease activity of these cells may be of therapeutic interest. Here, we show that acute restraint stress increases cFos expression in CRF neurons in the mouse dorsal BNST, consistent with a role for these neurons in stress-related behaviors. We find that activation of α2A-adrenergic receptors (ARs) by the agonist guanfacine reduced cFos expression in these neurons both in stressed and unstressed conditions. Further, we find that α- and β-ARs differentially regulate excitatory drive onto these neurons. Pharmacological and channelrhodopsin-assisted mapping experiments suggest that α2A-ARs specifically reduce excitatory drive from parabrachial nucleus (PBN) afferents onto CRF neurons. Given that the α2A-AR is a Gi-linked GPCR, we assessed the impact of activating the Gi-coupled DREADD hM4Di in the PBN on restraint stress regulation of BNST CRF neurons. CNO activation of PBN hM4Di reduced stress-induced Fos in BNST Crh neurons. Further, using Prkcd as an additional marker of BNST neuronal identity, we uncovered a female-specific upregulation of the coexpression of Prkcd/Crh in BNST neurons following stress, which was prevented by ovariectomy. These findings show that stress activates BNST CRF neurons, and that α2A-AR activation suppresses the in vivo activity of these cells, at least in part by suppressing excitatory drive from PBN inputs onto CRF neurons.SIGNIFICANCE STATEMENT Stress is a major variable contributing to mood disorders. Here, we show that stress increases activation of BNST CRF neurons that drive negative affective behavior. We find that the clinically well tolerated α2A-AR agonist guanfacine reduces activity of these cells in vivo, and reduces excitatory PBN inputs onto these cells ex vivo Additionally, we uncover a novel sex-dependent coexpression of Prkcd with Crh in female BNST neurons after stress, an effect abolished by ovariectomy. These results demonstrate input-specific interactions between norepinephrine and CRF, and point to an action by which guanfacine may reduce negative affective responses.
Collapse
|
20
|
Greenwald MK. Anti-stress neuropharmacological mechanisms and targets for addiction treatment: A translational framework. Neurobiol Stress 2018; 9:84-104. [PMID: 30238023 PMCID: PMC6138948 DOI: 10.1016/j.ynstr.2018.08.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 07/30/2018] [Accepted: 08/10/2018] [Indexed: 12/18/2022] Open
Abstract
Stress-related substance use is a major challenge for treating substance use disorders. This selective review focuses on emerging pharmacotherapies with potential for reducing stress-potentiated seeking and consumption of nicotine, alcohol, marijuana, cocaine, and opioids (i.e., key phenotypes for the most commonly abused substances). I evaluate neuropharmacological mechanisms in experimental models of drug-maintenance and relapse, which translate more readily to individuals presenting for treatment (who have initiated and progressed). An affective/motivational systems model (three dimensions: valence, arousal, control) is mapped onto a systems biology of addiction approach for addressing this problem. Based on quality of evidence to date, promising first-tier neurochemical receptor targets include: noradrenergic (α1 and β antagonist, α2 agonist), kappa-opioid antagonist, nociceptin antagonist, orexin-1 antagonist, and endocannabinoid modulation (e.g., cannabidiol, FAAH inhibition); second-tier candidates may include corticotropin releasing factor-1 antagonists, serotonergic agents (e.g., 5-HT reuptake inhibitors, 5-HT3 antagonists), glutamatergic agents (e.g., mGluR2/3 agonist/positive allosteric modulator, mGluR5 antagonist/negative allosteric modulator), GABA-promoters (e.g., pregabalin, tiagabine), vasopressin 1b antagonist, NK-1 antagonist, and PPAR-γ agonist (e.g., pioglitazone). To address affective/motivational mechanisms of stress-related substance use, it may be advisable to combine agents with actions at complementary targets for greater efficacy but systematic studies are lacking except for interactions with the noradrenergic system. I note clinically-relevant factors that could mediate/moderate the efficacy of anti-stress therapeutics and identify research gaps that should be pursued. Finally, progress in developing anti-stress medications will depend on use of reliable CNS biomarkers to validate exposure-response relationships.
Collapse
Affiliation(s)
- Mark K. Greenwald
- Department of Psychiatry and Behavioral Neurosciences, School of Medicine, Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, 48201, USA
| |
Collapse
|
21
|
Aldosterone-sensitive HSD2 neurons in mice. Brain Struct Funct 2018; 224:387-417. [PMID: 30343334 DOI: 10.1007/s00429-018-1778-y] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 10/03/2018] [Indexed: 02/07/2023]
Abstract
Sodium deficiency elevates aldosterone, which in addition to epithelial tissues acts on the brain to promote dysphoric symptoms and salt intake. Aldosterone boosts the activity of neurons that express 11-beta-hydroxysteroid dehydrogenase type 2 (HSD2), a hallmark of aldosterone-sensitive cells. To better characterize these neurons, we combine immunolabeling and in situ hybridization with fate mapping and Cre-conditional axon tracing in mice. Many cells throughout the brain have a developmental history of Hsd11b2 expression, but in the adult brain one small brainstem region with a leaky blood-brain barrier contains HSD2 neurons. These neurons express Hsd11b2, Nr3c2 (mineralocorticoid receptor), Agtr1a (angiotensin receptor), Slc17a6 (vesicular glutamate transporter 2), Phox2b, and Nxph4; many also express Cartpt or Lmx1b. No HSD2 neurons express cholinergic, monoaminergic, or several other neuropeptidergic markers. Their axons project to the parabrachial complex (PB), where they intermingle with AgRP-immunoreactive axons to form dense terminal fields overlapping FoxP2 neurons in the central lateral subnucleus (PBcL) and pre-locus coeruleus (pLC). Their axons also extend to the forebrain, intermingling with AgRP- and CGRP-immunoreactive axons to form dense terminals surrounding GABAergic neurons in the ventrolateral bed nucleus of the stria terminalis (BSTvL). Sparse axons target the periaqueductal gray, ventral tegmental area, lateral hypothalamic area, paraventricular hypothalamic nucleus, and central nucleus of the amygdala. Dual retrograde tracing revealed that largely separate HSD2 neurons project to pLC/PB or BSTvL. This projection pattern raises the possibility that a subset of HSD2 neurons promotes the dysphoric, anorexic, and anhedonic symptoms of hyperaldosteronism via AgRP-inhibited relay neurons in PB.
Collapse
|
22
|
Farrar MJ, Kolkman KE, Fetcho JR. Features of the structure, development, and activity of the zebrafish noradrenergic system explored in new CRISPR transgenic lines. J Comp Neurol 2018; 526:2493-2508. [PMID: 30070695 DOI: 10.1002/cne.24508] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 07/24/2018] [Accepted: 07/29/2018] [Indexed: 11/11/2022]
Abstract
The noradrenergic (NA) system of vertebrates is implicated in learning, memory, arousal, and neuroinflammatory responses, but is difficult to access experimentally. Small and optically transparent, larval zebrafish offer the prospect of exploration of NA structure and function in an intact animal. We made multiple transgenic zebrafish lines using the CRISPR/Cas9 system to insert fluorescent reporters upstream of slc6a2, the norepinephrine transporter gene. These lines faithfully express reporters in NA cell populations, including the locus coeruleus (LC), which contains only about 14 total neurons. We used the lines in combination with two-photon microscopy to explore the structure and projections of the NA system in the context of the columnar organization of cell types in the zebrafish hindbrain. We found robust alignment of NA projections with glutamatergic neurotransmitter stripes in some hindbrain segments, suggesting orderly relations to neuronal cell types early in life. We also quantified neurite density in the rostral spinal cord in individual larvae with as much as 100% difference in the number of LC neurons, and found no correlation between neuronal number in the LC and projection density in the rostral spinal cord. Finally, using light sheet microscopy, we performed bilateral calcium imaging of the entire LC. We found that large-amplitude calcium responses were evident in all LC neurons and showed bilateral synchrony, whereas small-amplitude events were more likely to show interhemispheric asynchrony, supporting the potential for targeted LC neuromodulation. Our observations and new transgenic lines set the stage for a deeper understanding of the NA system.
Collapse
Affiliation(s)
- Matthew J Farrar
- Department of Neurobiology and Behavior, Cornell University, Ithaca, New York.,Department of Math, Physics and Statistics, Messiah College, Mechanicsburg, Pennsylvania
| | - Kristine E Kolkman
- Department of Neurobiology and Behavior, Cornell University, Ithaca, New York
| | - Joseph R Fetcho
- Department of Neurobiology and Behavior, Cornell University, Ithaca, New York
| |
Collapse
|
23
|
Glutamatergic and gabaergic ventral BNST neurons differ in their physiological properties and responsiveness to noradrenaline. Neuropsychopharmacology 2018; 43:2126-2133. [PMID: 29704000 PMCID: PMC6098041 DOI: 10.1038/s41386-018-0070-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 04/05/2018] [Accepted: 04/10/2018] [Indexed: 02/07/2023]
Abstract
The bed nucleus of the stria terminalis (BNST) regulates defensive responses to threats and its anteroventral portion (BNST-AV) is involved. BNST-AV contains a minority of glutamatergic neurons scattered among a dominant population of GABAergic cells. There is evidence that these two cell types might exert opposite influences, the former promoting and the latter reducing anxiety. Although GABAergic cells greatly outnumber glutamatergic neurons in BNST-AV, in some circumstances the influence of glutamatergic cells appears to predominate. Related to this, BNST-AV receives a very strong noradrenaline (NA) input and negative emotional states are associated with a marked rise of NA concentration in BNST-AV. However, it is currently unclear whether NA differentially alters the excitability of glutamatergic and GABAergic BNST-AV neurons. Thus, to shed light on how BNST-AV regulates negative emotional states, the present study compared the physiological properties and NA responsiveness of glutamatergic and GABAergic BNST-AV neurons using whole-cell recordings in transgenic mice that express a fluorescent reporter in either cell group. We found that glutamatergic cells had a slightly more complex morphology than the GABAergic cells, a higher intrinsic excitability, and a different responsiveness to NA. Indeed, while NA inhibited EPSPs in both cell types through α1 and α2 adrenoreceptors, the EPSP reduction seen in glutamatergic cells had a lower amplitude and a shorter duration than in GABAergic cells. These differences were due to the presence of a β-receptor-mediated EPSP enhancement in the glutamatergic cells. Together, our results suggest that multiple properties contribute to the disproportionate influence of glutamatergic BNST-AV neurons.
Collapse
|
24
|
Dorsal BNST α 2A-Adrenergic Receptors Produce HCN-Dependent Excitatory Actions That Initiate Anxiogenic Behaviors. J Neurosci 2018; 38:8922-8942. [PMID: 30150361 DOI: 10.1523/jneurosci.0963-18.2018] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 08/15/2018] [Accepted: 08/20/2018] [Indexed: 11/21/2022] Open
Abstract
Stress is a precipitating agent in neuropsychiatric disease and initiates relapse to drug-seeking behavior in addicted patients. Targeting the stress system in protracted abstinence from drugs of abuse with anxiolytics may be an effective treatment modality for substance use disorders. α2A-adrenergic receptors (α2A-ARs) in extended amygdala structures play key roles in dampening stress responses. Contrary to early thinking, α2A-ARs are expressed at non-noradrenergic sites in the brain. These non-noradrenergic α2A-ARs play important roles in stress responses, but their cellular mechanisms of action are unclear. In humans, the α2A-AR agonist guanfacine reduces overall craving and uncouples craving from stress, yet minimally affects relapse, potentially due to competing actions in the brain. Here, we show that heteroceptor α2A-ARs postsynaptically enhance dorsal bed nucleus of the stria terminalis (dBNST) neuronal activity in mice of both sexes. This effect is mediated by hyperpolarization-activated cyclic nucleotide-gated cation channels because inhibition of these channels is necessary and sufficient for excitatory actions. Finally, this excitatory action is mimicked by clozapine-N-oxide activation of the Gi-coupled DREADD hM4Di in dBNST neurons and its activation elicits anxiety-like behavior in the elevated plus maze. Together, these data provide a framework for elucidating cell-specific actions of GPCR signaling and provide a potential mechanism whereby competing anxiogenic and anxiolytic actions of guanfacine may affect its clinical utility in the treatment of addiction.SIGNIFICANCE STATEMENT Stress affects the development of neuropsychiatric disorders including anxiety and addiction. Guanfacine is an α2A-adrenergic receptor (α2A-AR) agonist with actions in the bed nucleus of the stria terminalis (BNST) that produces antidepressant actions and uncouples stress from reward-related behaviors. Here, we show that guanfacine increases dorsal BNST neuronal activity through actions at postsynaptic α2A-ARs via a mechanism that involves hyperpolarization-activated cyclic nucleotide gated cation channels. This action is mimicked by activation of the designer receptor hM4Di expressed in the BNST, which also induces anxiety-like behaviors. Together, these data suggest that postsynaptic α2A-ARs in BNST have excitatory actions on BNST neurons and that these actions can be phenocopied by the so-called "inhibitory" DREADDs, suggesting that care must be taken regarding interpretation of data obtained with these tools.
Collapse
|
25
|
Haass-Koffler CL, Swift RM, Leggio L. Noradrenergic targets for the treatment of alcohol use disorder. Psychopharmacology (Berl) 2018; 235:1625-1634. [PMID: 29460163 PMCID: PMC5995154 DOI: 10.1007/s00213-018-4843-6] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 01/28/2018] [Indexed: 12/17/2022]
Abstract
The role of norepinephrine (NE) in the development of alcohol use disorder (AUD) has been studied over the past several decades. However, the NE system has been largely ignored for many years as a potential target for medication development for AUD. More recently, preclinical and clinical studies have demonstrated the potential value of targeting NE signaling for developing new pharmacological treatments for AUD. This review contributes to a special issue of Psychopharmacology focused on promising targets for alcohol addiction. Specifically, this review coalesces preclinical and clinical neuroscience that re-evaluate the noradrenergic system, and in particular the alpha-1 receptor, as a potential target for AUD.
Collapse
Affiliation(s)
- Carolina L Haass-Koffler
- Center for Alcohol and Addiction Studies, Department of Psychiatry and Human Behavior, Brown University, Providence, RI, 02912, USA.
- Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, Brown University, Providence, RI, USA.
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD, USA.
| | - Robert M Swift
- Center for Alcohol and Addiction Studies, Department of Psychiatry and Human Behavior, Brown University, Providence, RI, 02912, USA
- Providence VA Medical Center, Providence, RI, 02912, USA
| | - Lorenzo Leggio
- Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, Brown University, Providence, RI, USA
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
26
|
Yapıcı-Eser H, Dönmez-Demir B, Kılıç K, Eren-Koçak E, Dalkara T. Stress modulates cortical excitability via α-2 adrenergic and glucocorticoid receptors: As assessed by spreading depression. Exp Neurol 2018; 307:45-51. [PMID: 29856967 DOI: 10.1016/j.expneurol.2018.05.024] [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: 04/05/2018] [Revised: 05/15/2018] [Accepted: 05/27/2018] [Indexed: 12/29/2022]
Abstract
An increase in cortical excitability may be one of the factors mediating stress-induced vulnerability to neuropsychiatric disorders. Since stress increases extracellular glutamate and predisposes to migraine with aura attacks, we aimed to study the effect of stress on cortical spreading depression (CSD), the biological substrate of migraine aura and a measure of cortical excitability. CSD was induced by increasing concentrations of KCl applied over the dura with 5-minute intervals and recorded from parieto-occipital cortex to assess the CSD-induction threshold in acutely-stressed, chronically-stressed and naive mice. To study the mechanisms of acute stress-induced decrease in CSD threshold, we systemically administered clonidine, yohimbine, propranolol, CRH1 receptor antagonist NBI27914, mifepristone and spironolactone at doses shown to be effective on stress as well as a central noradrenergic neurotoxin (DSP-4) before acute stress. CSD threshold was significantly lowered by acute and chronic stress as well as central noradrenergic denervation. Clonidine and mifepristone further decreased the CSD threshold below the acute stress-induced levels, whereas yohimbine reversed the acute stress-induced decrease in CSD threshold compared to the saline-injected and stressed control groups. Propranolol, NBI27914 and spironolactone did not modify the effect of acute stress on CSD threshold. Stress increases cortical excitability as illustrated by a decrease in CSD-induction threshold. This action of acute stress is mediated by α2-adrenergic and glucocorticoid receptors. The decrease in CSD threshold may account for the stress-induced susceptibility to migraine. CSD may be used as a tool to study the link between stress-related disorders and cortical excitability.
Collapse
Affiliation(s)
- Hale Yapıcı-Eser
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey; School of Medicine, Department of Psychiatry, Koç University & Research Center for Translational Medicine (KUTTAM), Koç University, İstanbul, Turkey
| | - Buket Dönmez-Demir
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey
| | - Kıvılcım Kılıç
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey; Neurophotonics Center, Boston University, Boston, MA, United States
| | - Emine Eren-Koçak
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey.
| | - Turgay Dalkara
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey.
| |
Collapse
|
27
|
Francesconi W, Szücs A, Berton F, Koob GF, Vendruscolo LF, Sanna PP. Opiate dependence induces cell type-specific plasticity of intrinsic membrane properties in the rat juxtacapsular bed nucleus of stria terminalis (jcBNST). Psychopharmacology (Berl) 2017; 234:3485-3498. [PMID: 28986608 PMCID: PMC5993421 DOI: 10.1007/s00213-017-4732-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 09/05/2017] [Indexed: 01/03/2023]
Abstract
RATIONALE Drugs of abuse can alter circuit dynamics by modifying synaptic efficacy and/or the intrinsic membrane properties of neurons. The juxtacapsular subdivision of the bed nucleus of stria terminalis (jcBNST) has unique connectivity that positions it to integrate cortical and amygdala inputs and provide feed-forward inhibition to the central nucleus of the amygdala (CeA), among other regions. In this study, we investigated changes in the synaptic and intrinsic properties of neurons in the rat jcBNST during protracted withdrawal from morphine dependence using a combination of conventional electrophysiological methods and the dynamic clamp technique. RESULTS A history of opiate dependence induced a form of cell type-specific plasticity characterized by reduced inward rectification associated with more depolarized resting membrane potentials and increased membrane resistance. This cell type also showed a lower rheobase when stimulated with direct current (DC) pulses as well as a decreased firing threshold under simulated synaptic bombardment with the dynamic clamp. Morphine dependence also decreased excitatory postsynaptic potential amplification, suggesting the downregulation of the persistent Na+ current (I NaP). CONCLUSION These findings show that a history of morphine dependence leads to persistent cell type-specific plasticity of the passive membrane properties of a jcBNST neuronal population, leading to an overall increased excitability of such neurons. By altering the activity of extended amygdala circuits where they are embedded, changes in the integration properties of jcBNST neurons may contribute to emotional dysregulation associated with drug dependence and withdrawal.
Collapse
Affiliation(s)
- Walter Francesconi
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA and Department of Anatomy and Cell Biology, School of Medicine, University of Illinois at Chicago, Chicago, IL
| | - Attila Szücs
- BioCircuits Institute, University of California San Diego, La Jolla, CA and MTA-ELTE NAP-B Neuronal Cell Biology Group, Eötvös Lóránd University, Budapest, Hungary
| | - Fulvia Berton
- Dipartimento di Biologia, Universita’ degli Studi di Pisa, Pisa, Italy and Department of Anatomy and Cell Biology, School of Medicine, University of Illinois at Chicago, Chicago, IL
| | - George F. Koob
- Department of Neuroscience, The Scripps Research Institute, La Jolla. Current address: National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, MD
| | - Leandro F. Vendruscolo
- Department of Neuroscience, The Scripps Research Institute, La Jolla. Current address: National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD
| | - Pietro Paolo Sanna
- Department of Immunology and Microbiology and Department ofNeuroscience, The Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|
28
|
Yeganeh F, Nasimi A, Hatam M. Interaction of GABA and norepinephrine in the lateral division of the bed nucleus of the stria terminals in anesthetized rat, correlating single-unit and cardiovascular responses. Neuroscience 2017; 356:255-264. [PMID: 28576724 DOI: 10.1016/j.neuroscience.2017.05.044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 05/18/2017] [Accepted: 05/23/2017] [Indexed: 12/23/2022]
Abstract
The bed nucleus of the stria terminalis (BST) consists of multiple anatomically distinct nuclei. The lateral division, which receives dense noradrenergic innervation, has been implicated in cardiovascular regulation and modulation of responses to stress. This study is performed to identify the cardiovascular and single-unit responses of the lateral BST to norepinephrine (NE), involved adrenoceptors, and possible interaction with GABAergic system of the BST in urethane-anesthetized rats. NE, adrenoreceptor antagonists, and GABAA antagonist were microinjected into the lateral division of BST, while arterial pressure (AP), heart rate (HR), and single-unit responses were simultaneously recorded. NE microinjected into the lateral division of BST produced depressor and bradycardic responses. The decrease in AP and HR to NE was blocked by prazosin, an α1-adrenoreceptor antagonist, but not by yohimbine, an α2 antagonist. Furthermore, injections of the GABAA receptor antagonist, bicuculline methiodide (BMI), into the lateral BST abolished the NE-induced depressor and bradycardic responses. We also observed single-unit responses consisting of excitatory and inhibitory responses correlated with cardiovascular function to the microinjection of NE. In conclusion, these data provide the first evidence that microinjection of NE in the lateral division of BST produces depressor and bradycardic responses in urethane-anesthetized rat. The depressor and bradycardiac response are mediated by local α1- but not α2-adrenoceptors. α1-AR activates the GABAergic system within the BST, which in turn produces depressor and bradycardic responses.
Collapse
Affiliation(s)
- Fahimeh Yeganeh
- Dept. of Physiology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Nasimi
- Dept. of Physiology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Masoumeh Hatam
- Dept. of Physiology, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
29
|
Adami MB, Barretto-de-Souza L, Duarte JO, Almeida J, Crestani CC. Both N-methyl-D-aspartate and non-N-methyl-D-aspartate glutamate receptors in the bed nucleus of the stria terminalis modulate the cardiovascular responses to acute restraint stress in rats. J Psychopharmacol 2017; 31:674-681. [PMID: 28198211 DOI: 10.1177/0269881117691468] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The bed nucleus of the stria terminalis (BNST) is a forebrain structure that has been implicated on cardiovascular responses evoked by emotional stress. However, the local neurochemical mechanisms mediating the BNST control of stress responses are not fully described. In our study we investigated the involvement of glutamatergic neurotransmission within the BNST in cardiovascular changes evoked by acute restraint stress in rats. For this study, we investigated the effects of bilateral microinjections of selective antagonists of either N-methyl-D-aspartate (NMDA) or non-NMDA glutamate receptors into the BNST on the arterial pressure and heart rate increase and the decrease in tail skin temperature induced by acute restraint stress. Microinjection of the selective NMDA glutamate receptor antagonist LY235959 (1 nmol/100 nL) into the BNST decreased the tachycardiac response to restraint stress, without affecting the arterial pressure increase and the drop in skin temperature. Bilateral BNST treatment with the selective non-NMDA glutamate receptor NBQX (1 nmol/100 nL) decreased the heart rate increase and the fall in tail skin temperature, without affecting the blood pressure increase. These findings indicate a facilitatory influence of BNST glutamatergic neurotransmission via coactivation of local NMDA and non-NMDA receptors on the tachycardiac response to stress, whereas control of sympathetic-mediated cutaneous vasoconstriction is selectively mediated by local non-NMDA glutamate receptors.
Collapse
Affiliation(s)
- Mariane B Adami
- 1 Laboratory of Pharmacology, São Paulo State University (UNESP), Araraquara, Brazil
| | - Lucas Barretto-de-Souza
- 1 Laboratory of Pharmacology, São Paulo State University (UNESP), Araraquara, Brazil.,2 Joint Federal University of São Carlos (UFSCar)-UNESP Graduate Program in Physiological Sciences, São Carlos, Brazil
| | - Josiane O Duarte
- 1 Laboratory of Pharmacology, São Paulo State University (UNESP), Araraquara, Brazil
| | - Jeferson Almeida
- 1 Laboratory of Pharmacology, São Paulo State University (UNESP), Araraquara, Brazil.,2 Joint Federal University of São Carlos (UFSCar)-UNESP Graduate Program in Physiological Sciences, São Carlos, Brazil
| | - Carlos C Crestani
- 1 Laboratory of Pharmacology, São Paulo State University (UNESP), Araraquara, Brazil.,2 Joint Federal University of São Carlos (UFSCar)-UNESP Graduate Program in Physiological Sciences, São Carlos, Brazil
| |
Collapse
|
30
|
Fox ME, Rodeberg NT, Wightman RM. Reciprocal Catecholamine Changes during Opiate Exposure and Withdrawal. Neuropsychopharmacology 2017; 42:671-681. [PMID: 27461081 PMCID: PMC5240169 DOI: 10.1038/npp.2016.135] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 06/28/2016] [Accepted: 07/19/2016] [Indexed: 11/09/2022]
Abstract
Dysregulated catecholamine signaling has long been implicated in drug abuse. Although much is known about adaptations following chronic drug administration, little work has investigated how a single drug exposure paired with withdrawal influences catecholamine signaling in vivo. We used fast-scan cyclic voltammetry in freely moving rats to measure real-time catecholamine overflow during acute morphine exposure and naloxone-precipitated withdrawal in two regions associated with the addiction cycle: the dopamine-dense nucleus accumbens (NAc) and norepinephrine-rich ventral bed nucleus of the stria terminalis (vBNST). We compared dopamine transients in the NAc with norepinephrine concentration changes in the vBNST, and correlated release with specific withdrawal-related behaviors. Morphine increased dopamine transients in the NAc, but did not elicit norepinephrine responses in the vBNST. Conversely, dopamine output was decreased during withdrawal, while norepinephrine was released in the vBNST during specific withdrawal symptoms. Both norepinephrine and withdrawal symptoms could be elicited in the absence of morphine by administering naloxone with an α2 antagonist. The data support reciprocal roles for dopamine and norepinephrine signaling during drug exposure and withdrawal. The data also support the allostasis model and show that negative-reinforcement may begin working after a single exposure/withdrawal episode.
Collapse
Affiliation(s)
- Megan E Fox
- Department of Chemistry and Neuroscience Center, University of North Carolina, Chapel Hill, NC, USA
| | - Nathan T Rodeberg
- Department of Chemistry and Neuroscience Center, University of North Carolina, Chapel Hill, NC, USA
| | - R Mark Wightman
- Department of Chemistry and Neuroscience Center, University of North Carolina, Chapel Hill, NC, USA,Department of Chemistry and Neuroscience Center, University of North Carolina, Venable Hall, South Road, Chapel Hill, NC 27599, USA, Tel: +1 919 9621472, Fax: +1 919 962 2388, E-mail:
| |
Collapse
|
31
|
Fox ME, Wightman RM. Contrasting Regulation of Catecholamine Neurotransmission in the Behaving Brain: Pharmacological Insights from an Electrochemical Perspective. Pharmacol Rev 2017; 69:12-32. [PMID: 28267676 PMCID: PMC7558309 DOI: 10.1124/pr.116.012948] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Catecholamine neurotransmission plays a key role in regulating a variety of behavioral and physiologic processes, and its dysregulation is implicated in both neurodegenerative and neuropsychiatric disorders. Over the last four decades, in vivo electrochemistry has enabled the discovery of contrasting catecholamine regulation in the brain. These rapid and spatially resolved measurements have been conducted in brain slices, and in anesthetized and freely behaving animals. In this review, we describe the methods enabling in vivo measurements of dopamine and norepinephrine, and subsequent findings regarding their release and regulation in intact animals. We thereafter discuss key studies in awake animals, demonstrating that these catecholamines are not only differentially regulated, but are released in opposition of each other during appetitive and aversive stimuli.
Collapse
Affiliation(s)
- Megan E Fox
- Department of Chemistry and Neuroscience Center, University of North Carolina, Chapel Hill, North Carolina
| | - R Mark Wightman
- Department of Chemistry and Neuroscience Center, University of North Carolina, Chapel Hill, North Carolina
| |
Collapse
|
32
|
|
33
|
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
|
34
|
Gouveia MK, Miguel TT, Busnardo C, Scopinho AA, Corrêa FM, Nunes-de-Souza RL, Crestani CC. Dissociation in control of physiological and behavioral responses to emotional stress by cholinergic neurotransmission in the bed nucleus of the stria terminalis in rats. Neuropharmacology 2016; 101:379-88. [DOI: 10.1016/j.neuropharm.2015.10.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 10/03/2015] [Accepted: 10/10/2015] [Indexed: 11/25/2022]
|
35
|
Daniel SE, Rainnie DG. Stress Modulation of Opposing Circuits in the Bed Nucleus of the Stria Terminalis. Neuropsychopharmacology 2016; 41:103-25. [PMID: 26096838 PMCID: PMC4677121 DOI: 10.1038/npp.2015.178] [Citation(s) in RCA: 155] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Revised: 05/22/2015] [Accepted: 06/17/2015] [Indexed: 12/11/2022]
Abstract
The anterior bed nucleus of the stria terminalis (BNST) has been recognized as a critical structure in regulating trait anxiety, contextual fear memory, and appetitive behavior, and is known to be sensitive to stress manipulations. As one of the most complex structures in the central nervous system, the intrinsic circuitry of the BNST is largely unknown; however, recent technological developments have allowed researchers to begin to untangle the internal connections of the nucleus. This research has revealed the possibility of two opposing circuits, one anxiolytic and one anxiogenic, within the BNST, the relative strength of which determines the behavioral outcome. The balance of these pathways is critical in maintaining a normal physiological and behavioral state; however, stress and drugs of abuse can differentially affect the opposing circuitry within the nucleus to shift the balance to a pathological state. In this review, we will examine how stress interacts with the neuromodulators, corticotropin-releasing factor, norepinephrine, dopamine, and serotonin to affect the circuitry of the BNST as well as how synaptic plasticity in the BNST is modulated by stress, resulting in long-lasting changes in the circuit and behavioral state.
Collapse
Affiliation(s)
- Sarah E Daniel
- Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Donald G Rainnie
- Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
36
|
Norepinephrine ignites local hotspots of neuronal excitation: How arousal amplifies selectivity in perception and memory. Behav Brain Sci 2015; 39:e200. [PMID: 26126507 DOI: 10.1017/s0140525x15000667] [Citation(s) in RCA: 376] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Emotional arousal enhances perception and memory of high-priority information but impairs processing of other information. Here, we propose that, under arousal, local glutamate levels signal the current strength of a representation and interact with norepinephrine (NE) to enhance high priority representations and out-compete or suppress lower priority representations. In our "glutamate amplifies noradrenergic effects" (GANE) model, high glutamate at the site of prioritized representations increases local NE release from the locus coeruleus (LC) to generate "NE hotspots." At these NE hotspots, local glutamate and NE release are mutually enhancing and amplify activation of prioritized representations. In contrast, arousal-induced LC activity inhibits less active representations via two mechanisms: 1) Where there are hotspots, lateral inhibition is amplified; 2) Where no hotspots emerge, NE levels are only high enough to activate low-threshold inhibitory adrenoreceptors. Thus, LC activation promotes a few hotspots of excitation in the context of widespread suppression, enhancing high priority representations while suppressing the rest. Hotspots also help synchronize oscillations across neural ensembles transmitting high-priority information. Furthermore, brain structures that detect stimulus priority interact with phasic NE release to preferentially route such information through large-scale functional brain networks. A surge of NE before, during, or after encoding enhances synaptic plasticity at NE hotspots, triggering local protein synthesis processes that enhance selective memory consolidation. Together, these noradrenergic mechanisms promote selective attention and memory under arousal. GANE not only reconciles apparently contradictory findings in the emotion-cognition literature but also extends previous influential theories of LC neuromodulation by proposing specific mechanisms for how LC-NE activity increases neural gain.
Collapse
|
37
|
Oliveira LA, Almeida J, Benini R, Crestani CC. CRF1 and CRF2 receptors in the bed nucleus of the stria terminalis modulate the cardiovascular responses to acute restraint stress in rats. Pharmacol Res 2015; 95-96:53-62. [PMID: 25829333 DOI: 10.1016/j.phrs.2015.03.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 03/16/2015] [Accepted: 03/16/2015] [Indexed: 11/19/2022]
Abstract
The corticotropin-releasing factor (CRF) is involved in behavioral and physiological responses to emotional stress through its action in several limbic structures, including the bed nucleus of the stria terminalis (BNST). Nevertheless, the role of CRF1 and CRF2 receptors in the BNST in cardiovascular adjustments during aversive threat is unknown. Therefore, in the present study we investigated the involvement of CRF receptors within the BNST in cardiovascular responses evoked by acute restraint stress in rats. For this, we evaluated the effects of bilateral treatment of the BNST with selective agonists and antagonists of either CRF1 or CRF2 receptors in the arterial pressure and heart rate increase and the decrease in tail skin temperature induced by restraint stress. Microinjection of the selective CRF1 receptor antagonist CP376395 into the BNST reduced the pressor and tachycardiac responses caused by restraint. Conversely, BNST treatment with the selective CRF1 receptor agonist CRF increased restraint-evoked arterial pressure and HR responses and reduced the fall in tail skin temperature response. All effects of CRF were inhibited by local BNST pretreatment with CP376395. The selective CRF2 receptor antagonist antisalvagine-30 reduced the arterial pressure increase and the fall in tail skin temperature. The selective CRF2 receptor agonist urocortin-3 increased restraint-evoked pressor and tachycardiac responses and reduced the drop in cutaneous temperature. All effects of urocortin-3 were abolished by local BNST pretreatment with antisalvagine-30. These findings indicate an involvement of both CRF1 and CRF2 receptors in the BNST in cardiovascular adjustments during emotional stress.
Collapse
Affiliation(s)
- Leandro A Oliveira
- Laboratory of Pharmacology, School of Pharmaceutical Sciences, Univ. Estadual Paulista - UNESP, Araraquara, SP, Brazil; Joint UFSCar-UNESP Graduate Program in Physiological Sciences, São Carlos, SP, Brazil
| | - Jeferson Almeida
- Laboratory of Pharmacology, School of Pharmaceutical Sciences, Univ. Estadual Paulista - UNESP, Araraquara, SP, Brazil; Joint UFSCar-UNESP Graduate Program in Physiological Sciences, São Carlos, SP, Brazil
| | - Ricardo Benini
- Laboratory of Pharmacology, School of Pharmaceutical Sciences, Univ. Estadual Paulista - UNESP, Araraquara, SP, Brazil
| | - Carlos C Crestani
- Laboratory of Pharmacology, School of Pharmaceutical Sciences, Univ. Estadual Paulista - UNESP, Araraquara, SP, Brazil; Joint UFSCar-UNESP Graduate Program in Physiological Sciences, São Carlos, SP, Brazil.
| |
Collapse
|
38
|
McReynolds JR, Vranjkovic O, Thao M, Baker DA, Makky K, Lim Y, Mantsch JR. Beta-2 adrenergic receptors mediate stress-evoked reinstatement of cocaine-induced conditioned place preference and increases in CRF mRNA in the bed nucleus of the stria terminalis in mice. Psychopharmacology (Berl) 2014; 231:3953-63. [PMID: 24696080 PMCID: PMC8647032 DOI: 10.1007/s00213-014-3535-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2014] [Accepted: 03/02/2014] [Indexed: 12/30/2022]
Abstract
RATIONALE Understanding the mechanisms responsible for stress-induced relapse is important for guiding treatment strategies aimed at minimizing the contribution of stress to addiction. Evidence suggests that these mechanisms involve interactions between noradrenergic systems and the neuropeptide corticotropin-releasing factor (CRF). OBJECTIVES The interaction between β-adrenergic receptors (ARs) and CRF as it relates to the reinstatement of cocaine-conditioned reward in response to a stressor was examined in mice. We hypothesized that β2-ARs are required for stress-induced activation of CRF pathways responsible for reinstatement. METHODS Stress-induced relapse was examined based on the re-establishment of cocaine-induced conditioned place preference (CPP; 4 × 15 mg/kg cocaine, i.p.) after extinction using forced swim (6 min at 22 °C) or an injection of the β2-AR agonist, clenbuterol (4 mg/kg, i.p.). The CRF-R1 antagonist antalarmin (10 mg/kg, i.p.) or the β2-AR antagonist ICI-118,551 (1 mg/kg, i.p.) were given 30 min prior to reinstating stimuli. Quantitative PCR was conducted in dissected bed nucleus of the stria terminalis (BNST) and amygdala, putative sources of CRF that contribute to reinstatement, to examine the effects of ICI-118,551 on swim-induced increases in CRF messenger RNA (mRNA) in mice with a cocaine history. RESULTS Pretreatment with ICI-118,551 or antalarmin blocked swim-induced reinstatement of CPP. Reinstatement by clenbuterol was also blocked by antalarmin. ICI-118,551 pretreatment prevented swim-induced increases in CRF mRNA in the BNST. Effects in the amygdala were not observed. CONCLUSIONS These findings indicate that, during stress, norepinephrine, via β2-ARs, either directly or indirectly activates CRF-releasing neurons in the BNST that interface with motivational neurocircuitry to induce reinstatement of cocaine-conditioned reward.
Collapse
Affiliation(s)
- Jayme R. McReynolds
- Corresponding Author: John Mantsch, Ph.D., Department of Biomedical Sciences, Marquette University, , , Telephone Number: (414) 288-2036, Fax Number: (414) 288-6564
| | - Oliver Vranjkovic
- Corresponding Author: John Mantsch, Ph.D., Department of Biomedical Sciences, Marquette University, , , Telephone Number: (414) 288-2036, Fax Number: (414) 288-6564
| | | | | | | | | | | |
Collapse
|
39
|
Vranjkovic O, Gasser PJ, Gerndt CH, Baker DA, Mantsch JR. Stress-induced cocaine seeking requires a beta-2 adrenergic receptor-regulated pathway from the ventral bed nucleus of the stria terminalis that regulates CRF actions in the ventral tegmental area. J Neurosci 2014; 34:12504-14. [PMID: 25209288 PMCID: PMC4160780 DOI: 10.1523/jneurosci.0680-14.2014] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 07/29/2014] [Accepted: 07/31/2014] [Indexed: 11/21/2022] Open
Abstract
The ventral bed nucleus of the stria terminalis (vBNST) has been implicated in stress-induced cocaine use. Here we demonstrate that, in the vBNST, corticotropin releasing factor (CRF) is expressed in neurons that innervate the ventral tegmental area (VTA), a site where the CRF receptor antagonist antalarmin prevents the reinstatement of cocaine seeking by a stressor, intermittent footshock, following intravenous self-administration in rats. The vBNST receives dense noradrenergic innervation and expresses β adrenergic receptors (ARs). Footshock-induced reinstatement was prevented by bilateral intra-vBNST injection of the β-2 AR antagonist, ICI-118,551, but not the β-1 AR antagonist, betaxolol. Moreover, bilateral intra-vBNST injection of the β-2 AR agonist, clenbuterol, but not the β-1 agonist, dobutamine, reinstated cocaine seeking, suggesting that activation of vBNST β-2 AR is both necessary for stress-induced reinstatement and sufficient to induce cocaine seeking. The contribution of a β-2 AR-regulated vBNST-to-VTA pathway that releases CRF was investigated using a disconnection approach. Injection of ICI-118,551 into the vBNST in one hemisphere and antalarmin into the VTA of the contralateral hemisphere prevented footshock-induced reinstatement, whereas ipsilateral manipulations failed to attenuate stress-induced cocaine seeking, suggesting that β-2 AR regulate vBNST efferents that release CRF into the VTA, activating CRF receptors, and promoting cocaine use. Last, reinstatement by clenbuterol delivered bilaterally into the vBNST was prevented by bilateral vBNST pretreatment with antalarmin, indicating that β-2 AR-mediated actions in the vBNST also require local CRF receptor activation. Understanding the processes through which stress induces cocaine seeking should guide the development of new treatments for addiction.
Collapse
Affiliation(s)
- Oliver Vranjkovic
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin 53201
| | - Paul J Gasser
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin 53201
| | - Clayton H Gerndt
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin 53201
| | - David A Baker
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin 53201
| | - John R Mantsch
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin 53201
| |
Collapse
|
40
|
α(2A)-adrenergic receptors filter parabrachial inputs to the bed nucleus of the stria terminalis. J Neurosci 2014; 34:9319-31. [PMID: 25009265 DOI: 10.1523/jneurosci.0822-14.2014] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
α2-adrenergic receptors (AR) within the bed nucleus of the stria terminalis (BNST) reduce stress-reward interactions in rodent models. In addition to their roles as autoreceptors, BNST α(2A)-ARs suppress glutamatergic transmission. One prominent glutamatergic input to the BNST originates from the parabrachial nucleus (PBN) and consists of asymmetric axosomatic synapses containing calcitonin gene-related peptide (CGRP) and vGluT2. Here we provide immunoelectron microscopic data showing that many asymmetric axosomatic synapses in the BNST contain α(2A)-ARs. Further, we examined optically evoked glutamate release ex vivo in BNST from mice with virally delivered channelrhodopsin2 (ChR2) expression in PBN. In BNST from these animals, ChR2 partially colocalized with CGRP, and activation generated EPSCs in dorsal anterolateral BNST neurons that elicited two cell-type-specific outcomes: (1) feedforward inhibition or (2) an EPSP that elicited firing. We found that the α(2A)-AR agonist guanfacine selectively inhibited this PBN input to the BNST, preferentially reducing the excitatory response in ex vivo mouse brain slices. To begin to assess the overall impact of α(2A)-AR control of this PBN input on BNST excitatory transmission, we used a Thy1-COP4 mouse line with little postsynaptic ChR2 expression nor colocalization of ChR2 with CGRP in the BNST. In slices from these mice, we found that guanfacine enhanced, rather than suppressed, optogenetically initiated excitatory drive in BNST. Thus, our study reveals distinct actions of PBN afferents within the BNST and suggests that α(2A)-AR agonists may filter excitatory transmission in the BNST by inhibiting a component of the PBN input while enhancing the actions of other inputs.
Collapse
|
41
|
Excitatory drive onto dopaminergic neurons in the rostral linear nucleus is enhanced by norepinephrine in an α1 adrenergic receptor-dependent manner. Neuropharmacology 2014; 86:116-24. [PMID: 25018040 DOI: 10.1016/j.neuropharm.2014.07.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2014] [Revised: 06/09/2014] [Accepted: 07/01/2014] [Indexed: 12/19/2022]
Abstract
Dopaminergic innervation of the extended amygdala regulates anxiety-like behavior and stress responsivity. A portion of this dopamine input arises from dopamine neurons located in the ventral lateral periaqueductal gray (vlPAG) and rostral (RLi) and caudal linear nuclei of the raphe (CLi). These neurons receive substantial norepinephrine input, which may prime them for involvement in stress responses. Using a mouse line that expresses eGFP under control of the tyrosine hydroxylase promoter, we explored the physiology and responsiveness to norepinephrine of these neurons. We find that RLi dopamine neurons differ from VTA dopamine neurons with respect to membrane resistance, capacitance and the hyperpolarization-activated current, Ih. Further, we found that norepinephrine increased the frequency of spontaneous excitatory postsynaptic currents (sEPSCs) on RLi dopamine neurons. This effect was mediated through the α1 adrenergic receptor (AR), as the actions of norepinephrine were mimicked by the α1-AR agonist methoxamine and blocked by the α1-AR antagonist prazosin. This action of norepinephrine on sEPSCs was transient, as it did not persist in the presence of prazosin. Methoxamine also increased the frequency of miniature EPSCs, indicating that the α1-AR action on glutamatergic transmission likely has a presynaptic mechanism. There was also a modest decrease in sEPSC frequency with the application of the α2-AR agonist UK-14,304. These studies illustrate a potential mechanism through which norepinephrine could recruit the activity of this population of dopaminergic neurons.
Collapse
|
42
|
Medullary norepinephrine neurons modulate local oxygen concentrations in the bed nucleus of the stria terminalis. J Cereb Blood Flow Metab 2014; 34:1128-37. [PMID: 24714037 PMCID: PMC4083375 DOI: 10.1038/jcbfm.2014.60] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Revised: 02/20/2014] [Accepted: 03/03/2014] [Indexed: 11/08/2022]
Abstract
Neurovascular coupling is understood to be the underlying mechanism of functional hyperemia, but the actions of the neurotransmitters involved are not well characterized. Here we investigate the local role of the neurotransmitter norepinephrine in the ventral bed nucleus of the stria terminalis (vBNST) of the anesthetized rat by measuring O₂, which is delivered during functional hyperemia. Extracellular changes in norepinephrine and O₂ were simultaneously monitored using fast-scan cyclic voltammetry. Introduction of norepinephrine by electrical stimulation of the ventral noradrenergic bundle or by iontophoretic ejection induced an initial increase in O₂ levels followed by a brief dip below baseline. Supporting the role of a hyperemic response, the O₂ increases were absent in a brain slice containing the vBNST. Administration of selective pharmacological agents demonstrated that both phases of this response involve β-adrenoceptor activation, where the delayed decrease in O₂ is sensitive to both α- and β-receptor subtypes. Selective lesioning of the locus coeruleus with the neurotoxin DSP-4 confirmed that these responses are caused by the noradrenergic cells originating in the nucleus of the solitary tract and A1 cell groups. Overall, these results support that non-coerulean norepinephrine release can mediate activity-induced O₂ influx in a deep brain region.
Collapse
|
43
|
McReynolds JR, Peña DF, Blacktop JM, Mantsch JR. Neurobiological mechanisms underlying relapse to cocaine use: contributions of CRF and noradrenergic systems and regulation by glucocorticoids. Stress 2014; 17:22-38. [PMID: 24328808 DOI: 10.3109/10253890.2013.872617] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Considering its pervasive and uncontrollable influence in drug addicts, understanding the neurobiological processes through which stress contributes to drug use is a critical goal for addiction researchers and will likely be important for the development of effective medications aimed at relapse prevention. In this paper, we review work from our laboratory and others focused on determining the neurobiological mechanisms that underlie and contribute to stress-induced relapse of cocaine use with an emphasis on the actions of corticotropin-releasing factor in the ventral tegmental area (VTA) and a key pathway from the bed nucleus of the stria terminalis to the VTA that is regulated by norepinephrine and beta adrenergic receptors. Additionally, we discuss work suggesting that the influence of stress in cocaine addiction changes and intensifies with repeated cocaine use in an intake-dependent manner and examine the potential role of glucocorticoid hormones in the underlying drug-induced neuroadaptations. It is our hope that research in this area will inform clinical practice and medication development aimed at minimizing the contribution of stress to the addiction cycle, thereby improving treatment outcomes and reducing the societal costs of addiction.
Collapse
Affiliation(s)
- Jayme R McReynolds
- Department of Biomedical Sciences, Marquette University , Milwaukee, WI , USA
| | | | | | | |
Collapse
|
44
|
Noradrenergic neurotransmission within the bed nucleus of the stria terminalis modulates the retention of immobility in the rat forced swimming test. Behav Pharmacol 2013; 24:214-21. [PMID: 23625378 DOI: 10.1097/fbp.0b013e3283618ae4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The bed nucleus of the stria terminalis (BNST) is a limbic structure that has a direct influence on the autonomic, neuroendocrine, and behavioral responses to stress. It was recently reported that reversible inactivation of synaptic transmission within this structure causes antidepressant-like effects, indicating that activation of the BNST during stressful situations would facilitate the development of behavioral changes related to the neurobiology of depression. Moreover, noradrenergic neurotransmission is abundant in the BNST and has an important role in the regulation of emotional processes related to the stress response. Thus, this study aimed to test the hypothesis that activation of adrenoceptors within the BNST facilitates the development of behavioral consequences of stress. To investigate this hypothesis, male Wistar rats were stressed (forced swimming, 15 min) and 24 h later received intra-BNST injections of vehicle, WB4101, RX821002, CGP20712, or ICI118,551, which are selective α(1), α(2), β(1), and β(2) adrenoceptor antagonists, respectively, 10 min before a 5-min forced swimming test. It was observed that administration of WB4101 (10 and 15 nmol), CGP20712 (5 and 10 nmol), or ICI118,551 (5 nmol) into the BNST reduced the immobility time of rats subjected to forced swimming test, indicating an antidepressant-like effect. These findings suggest that activation of α(1), β(1), and β(2) adrenoceptors in the BNST could be involved in the development of the behavioral consequences of stress.
Collapse
|
45
|
Sterley TL, Howells FM, Russell VA. Evidence for reduced tonic levels of GABA in the hippocampus of an animal model of ADHD, the spontaneously hypertensive rat. Brain Res 2013; 1541:52-60. [PMID: 24161405 DOI: 10.1016/j.brainres.2013.10.023] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2013] [Revised: 10/09/2013] [Accepted: 10/14/2013] [Indexed: 11/25/2022]
Abstract
Recent studies have investigated the role of γ-aminobutyric acid (GABA) in the behavioural symptoms of attention-deficit/hyperactivity disorder (ADHD), specifically in behavioural disinhibition. Spontaneously hypertensive rats (SHR) are widely accepted as an animal model of ADHD, displaying core symptoms of the disorder. Using an in vitro superfusion technique, we have shown that glutamate-stimulated release of radio-actively labelled norepinephrine ([(3)H]NE) from prefrontal cortex and hippocampal slices is greater in SHR than in their normotensive control strain, Wistar-Kyoto rats (WKY), and/or a standard control strain, Sprague-Dawley rats (SD). In the present study, we investigated how the level of extracellular (tonic) GABA affects release of [(3)H]NE in hippocampal slices of male and female SHR, WKY and SD rats, in response to 3 glutamate stimulations (S1, S2, and S3). The hippocampal slices were prelabelled with [(3)H]NE and superfused with buffer containing 0μM, 1μM, 10μM, or 100μM GABA. Three consecutive glutamate stimulations were achieved by exposing slices to 3 pulses of glutamate (1mM), each separated by 10min. Increasing tonic levels of GABA increased basal and stimulated release of [(3)H]NE in all strains. When GABA was omitted from the superfusion buffer used to perfuse SHR hippocampal slices, but present at 100µM in the buffer used to perfuse WKY and SD hippocampal slices, glutamate-stimulated release of [(3)H]NE was similar in all three strains. In these conditions, the decrease in [(3)H]NE release from S1 to S2 and S3 was also similar in all three strains. These findings suggest that extracellular concentrations of GABA may be reduced in SHR hippocampus, in vivo, compared to WKY and SD. An underlying defect in GABA function may be at the root of the dysfunction in catecholamine transmission noted in SHR, and may underlie their ADHD-like behaviours.
Collapse
Affiliation(s)
- Toni-Lee Sterley
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Observatory 7925, South Africa.
| | | | | |
Collapse
|
46
|
Crestani CC, Alves FH, Gomes FV, Resstel LB, Correa FM, Herman JP. Mechanisms in the bed nucleus of the stria terminalis involved in control of autonomic and neuroendocrine functions: a review. Curr Neuropharmacol 2013; 11:141-59. [PMID: 23997750 PMCID: PMC3637669 DOI: 10.2174/1570159x11311020002] [Citation(s) in RCA: 185] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Revised: 11/21/2012] [Accepted: 12/03/2012] [Indexed: 12/22/2022] Open
Abstract
The bed nucleus of the stria terminalis (BNST) is a heterogeneous and complex limbic forebrain structure, which plays an important role in controlling autonomic, neuroendocrine and behavioral responses. The BNST is thought to serve as a key relay connecting limbic forebrain structures to hypothalamic and brainstem regions associated with autonomic and neuroendocrine functions. Its control of physiological and behavioral activity is mediated by local action of numerous neurotransmitters. In the present review we discuss the role of the BNST in control of both autonomic and neuroendocrine function. A description of BNST control of cardiovascular and hypothalamus-pituitary-adrenal axisactivity at rest and during physiological challenges (stress and physical exercise) is presented. Moreover, evidence for modulation of hypothalamic magnocellular neurons activity is also discussed. We attempt to focus on the discussion of BNST neurochemical mechanisms. Therefore, the source and targets of neurochemical inputs to BNST subregions and their role in control of autonomic and neuroendocrine function is discussed in details.
Collapse
Affiliation(s)
- Carlos C Crestani
- Laboratory of Pharmacology, Department of Natural Active Principles and Toxicology, School of Pharmaceutical Sciences, São Paulo State University, UNESP, Araraquara, SP, 14801-902, Brazil
| | | | | | | | | | | |
Collapse
|
47
|
Neurobiological mechanisms that contribute to stress-related cocaine use. Neuropharmacology 2013; 76 Pt B:383-94. [PMID: 23916481 DOI: 10.1016/j.neuropharm.2013.07.021] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Revised: 07/09/2013] [Accepted: 07/12/2013] [Indexed: 12/16/2022]
Abstract
The ability of stressful life events to trigger drug use is particularly problematic for the management of cocaine addiction due to the unpredictable and often uncontrollable nature of stress. For this reason, understanding the neurobiological processes that contribute to stress-related drug use is important for the development of new and more effective treatment strategies aimed at minimizing the role of stress in the addiction cycle. In this review we discuss the neurocircuitry that has been implicated in stress-induced drug use with an emphasis on corticotropin releasing factor actions in the ventral tegmental area (VTA) and an important pathway from the bed nucleus of the stria terminalis to the VTA that is regulated by norepinephrine via actions at beta adrenergic receptors. In addition to the neurobiological mechanisms that underlie stress-induced cocaine seeking, we review findings suggesting that the ability of stressful stimuli to trigger cocaine use emerges and intensifies in an intake-dependent manner with repeated cocaine self-administration. Further, we discuss evidence that the drug-induced neuroadaptations that are necessary for heightened susceptibility to stress-induced drug use are reliant on elevated levels of glucocorticoid hormones at the time of cocaine use. Finally, the potential ability of stress to function as a "stage setter" for drug use - increasing sensitivity to cocaine and drug-associated cues - under conditions where it does not directly trigger cocaine seeking is discussed. As our understanding of the mechanisms through which stress promotes drug use advances, the hope is that so too will the available tools for effectively managing addiction, particularly in cocaine addicts whose drug use is stress-driven. This article is part of a Special Issue entitled 'NIDA 40th Anniversary Issue'.
Collapse
|
48
|
Smith CD, Piasecki CC, Weera M, Olszewicz J, Lonstein JS. Noradrenergic alpha-2 receptor modulators in the ventral bed nucleus of the stria terminalis: effects on anxiety behavior in postpartum and virgin female rats. Behav Neurosci 2013; 127:582-97. [PMID: 23796237 PMCID: PMC3947518 DOI: 10.1037/a0032776] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Emotional hyperreactivity can inhibit maternal responsiveness in female rats and other animals. Maternal behavior in postpartum rats is disrupted by increasing norepinephrine release in the ventral bed nucleus of the stria terminalis (BSTv) with the α2-autoreceptor antagonist, yohimbine, or the more selective α2-autoreceptor antagonist, idazoxan (Smith et al., 2012). Because high noradrenergic activity in the BSTv can also increase anxiety-related behaviors, increased anxiety may underlie the disrupted mothering of dams given yohimbine or idazoxan. To assess this possibility, anxiety-related behaviors in an elevated plus maze were assessed in postpartum rats after administration of yohimbine or idazoxan. It was further assessed if the α2-autoreceptor agonist clonidine (which decreases norepinephrine release) would, conversely, reduce dams' anxiety. Groups of diestrous virgins were also examined. It was found that peripheral or intra-BSTv yohimbine did increase anxiety-related behavior in postpartum females. However, BSTv infusion of idazoxan did not reproduce yohimbine's anxiogenic effects and anxiety was not reduced by peripheral or intra-BSTv clonidine. Because yohimbine is a weak 5HT1A receptor agonist, other groups of females received BSTv infusion of the 5HT1A receptor agonist 8OH-DPAT, but it did not alter their anxiety-related behavior. Lastly, levels of norepinephrine and serotonin in tissue punches from the BSTv did not differ between postpartum and diestrous rats, but serotonin turnover was lower in mothers. These results suggest that the impaired maternal behavior after BSTv infusion of yohimbine or idazoxan cannot both be readily explained by an increase in dams' anxiety, and that BSTv α2-autoreceptor modulation alone has little influence on anxiety-related behaviors in postpartum or diestrous rats.
Collapse
Affiliation(s)
- Carl D. Smith
- Department of Psychology & Neuroscience Program, Michigan State University, 108 Giltner Hall, East Lansing, MI USA 48824
| | - Christopher C. Piasecki
- Department of Psychology & Neuroscience Program, Michigan State University, 108 Giltner Hall, East Lansing, MI USA 48824
| | - Marcus Weera
- Department of Psychology & Neuroscience Program, Michigan State University, 108 Giltner Hall, East Lansing, MI USA 48824
| | - Joshua Olszewicz
- Department of Psychology & Neuroscience Program, Michigan State University, 108 Giltner Hall, East Lansing, MI USA 48824
| | - Joseph S. Lonstein
- Department of Psychology & Neuroscience Program, Michigan State University, 108 Giltner Hall, East Lansing, MI USA 48824
| |
Collapse
|
49
|
Stamatakis AM, Sparta DR, Jennings JH, McElligott ZA, Decot H, Stuber GD. Amygdala and bed nucleus of the stria terminalis circuitry: Implications for addiction-related behaviors. Neuropharmacology 2013; 76 Pt B:320-8. [PMID: 23752096 DOI: 10.1016/j.neuropharm.2013.05.046] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 05/20/2013] [Accepted: 05/22/2013] [Indexed: 02/07/2023]
Abstract
Complex motivated behavioral processes, such as those that can go awry following substance abuse and other neuropsychiatric disorders, are mediated by a distributive network of neurons that reside throughout the brain. Neural circuits within the amygdala regions, such as the basolateral amygdala (BLA), and downstream targets such as the bed nucleus of the stria terminalis (BNST), are critical neuroanatomical structures for orchestrating emotional behavioral responses that may influence motivated actions such as the reinstatement of drug seeking behavior. Here, we review the functional neurocircuitry of the BLA and the BNST, and discuss how these circuits may guide maladaptive behavioral processes such as those seen in addiction. Thus, further study of the functional connectivity within these brain regions and others may provide insight for the development of new treatment strategies for substance use disorders. This article is part of a Special Issue entitled 'NIDA 40th Anniversary Issue'.
Collapse
Affiliation(s)
- Alice M Stamatakis
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Neurobiology Curriculum, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | | | | | | | | | |
Collapse
|
50
|
A corticotropin releasing factor pathway for ethanol regulation of the ventral tegmental area in the bed nucleus of the stria terminalis. J Neurosci 2013; 33:950-60. [PMID: 23325234 DOI: 10.1523/jneurosci.2949-12.2013] [Citation(s) in RCA: 122] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
A growing literature suggests that catecholamines and corticotropin-releasing factor (CRF) interact in a serial manner to activate the bed nucleus of the stria terminalis (BNST) to drive stress- or cue-induced drug- and alcohol-seeking behaviors. Data suggest that these behaviors are driven in part by BNST projections to the ventral tegmental area (VTA). Together, these findings suggest the existence of a CRF-signaling pathway within the BNST that is engaged by catecholamines and regulates the activity of BNST neurons projecting to the VTA. Here we test three aspects of this model to determine: (1) whether catecholamines modify CRF neuron activity in the BNST; (2) whether CRF regulates excitatory drive onto VTA-projecting BNST neurons; and (3) whether this system is altered by ethanol exposure and withdrawal. A CRF neuron fluorescent reporter strategy was used to identify BNST CRF neurons for whole-cell patch-clamp analysis in acutely prepared slices. Using this approach, we found that both dopamine and isoproterenol significantly depolarized BNST CRF neurons. Furthermore, using a fluorescent microsphere-based identification strategy we found that CRF enhances the frequency of spontaneous EPSCs onto VTA-projecting BNST neurons in naive mice. This action of CRF was occluded during acute withdrawal from chronic intermittent ethanol exposure. These findings suggest that dopamine and isoproterenol may enhance CRF release from local BNST sources, leading to enhancement of excitatory neurotransmission on VTA-projecting neurons, and that this pathway is engaged by patterns of alcohol exposure and withdrawal known to drive excessive alcohol intake.
Collapse
|