1
|
Belelli D, Lambert JJ, Wan MLY, Monteiro AR, Nutt DJ, Swinny JD. From bugs to brain: unravelling the GABA signalling networks in the brain-gut-microbiome axis. Brain 2025; 148:1479-1506. [PMID: 39716883 PMCID: PMC12074267 DOI: 10.1093/brain/awae413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 11/21/2024] [Accepted: 12/01/2024] [Indexed: 12/25/2024] Open
Abstract
Convergent data across species paint a compelling picture of the critical role of the gut and its resident microbiota in several brain functions and disorders. The chemicals mediating communication along these sophisticated highways of the brain-gut-microbiome (BGM) axis include both microbiota metabolites and classical neurotransmitters. Amongst the latter, GABA is fundamental to brain function, mediating most neuronal inhibition. Until recently, GABA's role and specific molecular targets in the periphery within the BGM axis had received limited attention. Yet, GABA is produced by neuronal and non-neuronal elements of the BGM, and recently, GABA-modulating bacteria have been identified as key players in GABAergic gut systems, indicating that GABA-mediated signalling is likely to transcend physiological boundaries and species. We review the available evidence to better understand how GABA facilitates the integration of molecularly and functionally disparate systems to bring about overall homeostasis and how GABA perturbations within the BGM axis can give rise to multi-system medical disorders, thereby magnifying the disease burden and the challenges for patient care. Analysis of transcriptomic databases revealed significant overlaps between GABAAR subunits expressed in the human brain and gut. However, in the gut, there are notable expression profiles for a select number of subunits that have received limited attention to date but could be functionally relevant for BGM axis homeostasis. GABAergic signalling, via different receptor subtypes, directly regulates BGM homeostasis by modulating the excitability of neurons within brain centres responsible for gastrointestinal (GI) function in a sex-dependent manner, potentially revealing mechanisms underlying the greater prevalence of GI disturbances in females. Apart from such top-down regulation of the BGM axis, a diverse group of cell types, including enteric neurons, glia, enteroendocrine cells, immune cells and bacteria, integrate peripheral GABA signals to influence brain functions and potentially contribute to brain disorders. We propose several priorities for this field, including the exploitation of available technologies to functionally dissect components of these GABA pathways within the BGM, with a focus on GI and brain-behaviour-disease. Furthermore, in silico ligand-receptor docking analyses using relevant bacterial metabolomic datasets, coupled with advances in knowledge of GABAAR 3D structures, could uncover new ligands with novel therapeutic potential. Finally, targeted design of dietary interventions is imperative to advancing their therapeutic potential to support GABA homeostasis across the BGM axis.
Collapse
Affiliation(s)
- Delia Belelli
- GABA Labs (Research) Ltd., Hemel Hempstead HP2 5HD, UK
- Division of Neuroscience, School of Medicine, Medical Sciences Institute, Dundee University, Dundee DD1 5HL, UK
- School of Medicine, Pharmacy & Biomedical Sciences, University of Portsmouth, Portsmouth PO1 2DT, UK
| | - Jeremy J Lambert
- Division of Neuroscience, School of Medicine, Medical Sciences Institute, Dundee University, Dundee DD1 5HL, UK
| | - Murphy Lam Yim Wan
- School of Medicine, Pharmacy & Biomedical Sciences, University of Portsmouth, Portsmouth PO1 2DT, UK
| | - Ana Rita Monteiro
- School of Medicine, Pharmacy & Biomedical Sciences, University of Portsmouth, Portsmouth PO1 2DT, UK
| | - David J Nutt
- GABA Labs (Research) Ltd., Hemel Hempstead HP2 5HD, UK
- Division of Psychiatry, Department of Brain Sciences, Imperial College London, London W12 0NN, UK
| | - Jerome D Swinny
- School of Medicine, Pharmacy & Biomedical Sciences, University of Portsmouth, Portsmouth PO1 2DT, UK
| |
Collapse
|
2
|
Luskin AT, Li L, Fu X, Martin MM, Barcomb K, Girven KS, Blackburn T, Wells BA, Thai ST, Li EM, Rana AN, Simon RC, Sun L, Gao L, Murry AD, Golden SA, Stuber GD, Ford CP, Gu L, Bruchas MR. Heterogeneous pericoerulear neurons tune arousal and exploratory behaviours. Nature 2025:10.1038/s41586-025-08952-w. [PMID: 40335695 DOI: 10.1038/s41586-025-08952-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 03/27/2025] [Indexed: 05/09/2025]
Abstract
As the primary source of noradrenaline in the brain, the locus coeruleus (LC) regulates arousal, avoidance and stress responses1,2. However, how local neuromodulatory inputs control LC function remains unresolved. Here we identify a population of transcriptionally, spatially and functionally diverse GABAergic (γ-aminobutyric acid-producing) neurons in the LC dendritic field that receive distant inputs and modulate modes of LC firing to control global arousal levels and arousal-related processing and behaviours. We define peri-LC anatomy using viral tracing and combine single-cell RNA sequencing with spatial transcriptomics to molecularly define both LC noradrenaline-producing and peri-LC cell types. We identify several neuronal cell types that underlie peri-LC functional diversity using a series of complementary neural circuit approaches in behaving mice. Our findings indicate that LC and peri-LC neurons are transcriptionally, functionally and anatomically heterogenous neuronal populations that modulate arousal and avoidance states. Defining the molecular, cellular and functional diversity of the LC and peri-LC provides a roadmap for understanding the neurobiological basis of arousal, motivation and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Andrew T Luskin
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA
- Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA, USA
- Graduate Program in Neuroscience, University of Washington, Seattle, WA, USA
- Laboratory of Neural Dynamics and Cognition, The Rockefeller University, New York, NY, USA
| | - Li Li
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA
- Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA, USA
- Seattle Children's Research Institute, Seattle, WA, USA
| | - Xiaonan Fu
- Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Madison M Martin
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA
- Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA, USA
- Graduate Program in Neuroscience, University of Washington, Seattle, WA, USA
| | - Kelsey Barcomb
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Kasey S Girven
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA
- Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA, USA
| | - Taylor Blackburn
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA
- Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA, USA
| | - Bailey A Wells
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA
- Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA, USA
| | - Sarah T Thai
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA
- Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA, USA
| | - Esther M Li
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA
- Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA, USA
- Department of Psychology, University of Washington, Seattle, WA, USA
| | - Akshay N Rana
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA
- Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA, USA
| | - Rhiana C Simon
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA
- Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA, USA
- Graduate Program in Neuroscience, University of Washington, Seattle, WA, USA
| | - Li Sun
- Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- TopoGene Inc., Seattle, WA, USA
| | - Lei Gao
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Alexandria D Murry
- Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA, USA
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| | - Sam A Golden
- Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA, USA
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| | - Garret D Stuber
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA
- Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA, USA
- Department of Pharmacology, University of Washington, Seattle, WA, USA
| | - Christopher P Ford
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Liangcai Gu
- Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Michael R Bruchas
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA.
- Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA, USA.
- Department of Pharmacology, University of Washington, Seattle, WA, USA.
- Department of Bioengineering, University of Washington, Seattle, WA, USA.
| |
Collapse
|
3
|
Pan Y, Zhang Y, Xu Z, Wei Z, Pan R, Hu G, Wang X, Yang L, Wu D, Zhang X, Wen X, Qu S, Li C, Zhu Z, Gao Y, Shi X, Zhu Y, Wu K, Wang D, Liu Y. Transcutaneous auricular vagus nerve stimulation to treat narcolepsy type 1 (TARGET-NT1): A two-arm, randomised, sham-controlled trial. Neurotherapeutics 2025:e00604. [PMID: 40335432 DOI: 10.1016/j.neurot.2025.e00604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2025] [Revised: 04/21/2025] [Accepted: 04/23/2025] [Indexed: 05/09/2025] Open
Abstract
To assess exploratorily the safety and efficacy of transcutaneous auricular vagus nerve stimulation (tVNS) as an adjunctive therapy in improving symptoms in patients with narcolepsy type 1 (NT1). The TARGET-NT1 trial, a two-arm, double-blinded, sham-controlled trial was conducted from April 2022, to June 2024 at Xijing Hospital in Xi'an, China. Participants were randomised to receive tVNS treatment or sham tVNS (stVNS) treatment. Both interventions were performed for two 30-min periods per day with the same stimulation parameters but different stimulation points, for 12 weeks. The primary outcome was the change in mean sleep onset latency of maintenance of wakefulness test (MWT) from baseline to week 12. Secondary outcomes included changes in Narcolepsy Severity Scale (NSS), Epworth Sleepiness Scale (ESS), 14-item Hamilton Anxiety Rating Scale (HAMA-14), 17-item Hamilton Depression Rating Scale (HAMD-17). Among 60 randomised participants (32 men [53.3 %] and 28 [46.7 %]; mean [SD] age, 29.9 [9.9] years), 56 were included in the modified intention-to-treat (mITT) analysis. From baseline to week 12, the difference in mean change in mean sleep onset latency of MWT was 3.09 (95 % CI, 1.00, 5.88; P = 0.0041) as compared with stVNS group. Significant improvements in NSS-EDS (-2.61 [95%CI, -4.07, -1.15; P = 0.0006]), NSS-SP (-1.11 [95%CI, -1.83, -0.38; P = 0.0030]), NSS-HH (-2.71 [95%CI, -3.36, -2.05; P < 0.0001]), NSS- DNS (-0.52 [95%CI, -0.87, -0.17; P = 0.0036]), ESS (-3.03 [95%CI, -4.30, -1.75; P < 0.0001]) and HAMD-17 (-2.50 [95%CI, -4.30, -0.70; P = 0.0069]) were observed in the tVNS group as compared with stVNS group. This exploratory study supported the efficacy and safety of tVNS in patients with NT1 and provided insights into the mechanisms underlying tVNS treatment for NT1. The findings highlight tVNS as a potential non-pharmacological adjunctive therapy for patients with NT1. This trial was registered with the Chinese Clinical Trial Registry, ChiCTR2400094550.
Collapse
Affiliation(s)
- Yuanhang Pan
- Department of Neurology, Xijing Hospital, Air Force Medical University, Xi'an, PR China.
| | - Yingchi Zhang
- Department of Neurology, Xijing Hospital, Air Force Medical University, Xi'an, PR China.
| | - Ziliang Xu
- Department of Radiology, Xijing Hospital, Air Force Medical University, Xi'an, PR China.
| | - Zihan Wei
- Department of Neurosurgery, General Hospital of Southern Theatre Command, Guangzhou, PR China.
| | - Rui Pan
- Department of Pediatrics, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, PR China.
| | - Gengyao Hu
- Department of Neurology, Xijing Hospital, Air Force Medical University, Xi'an, PR China.
| | - Xiaoli Wang
- Department of Neurology, Xijing Hospital, Air Force Medical University, Xi'an, PR China.
| | - Lei Yang
- Department of Neurology, Xijing Hospital, Air Force Medical University, Xi'an, PR China.
| | - Dianwei Wu
- Department of Neurology, Xijing Hospital, Air Force Medical University, Xi'an, PR China.
| | - Xinbo Zhang
- Department of Neurology, Xijing Hospital, Air Force Medical University, Xi'an, PR China.
| | - Xinyu Wen
- Department of Neurology, Xijing Hospital, Air Force Medical University, Xi'an, PR China.
| | - Shuyi Qu
- Department of Neurology, Xijing Hospital, Air Force Medical University, Xi'an, PR China.
| | - Chenwei Li
- Department of Neurology, Xijing Hospital, Air Force Medical University, Xi'an, PR China.
| | - Zhe Zhu
- Department of Neurology, Xijing Hospital, Air Force Medical University, Xi'an, PR China.
| | - Yuwen Gao
- Department of Neurology, Xijing Hospital, Air Force Medical University, Xi'an, PR China.
| | - Xiaodan Shi
- Department of Neurology, Xijing Hospital, Air Force Medical University, Xi'an, PR China.
| | - Yuanqiang Zhu
- Department of Radiology, Xijing Hospital, Air Force Medical University, Xi'an, PR China.
| | - Kejian Wu
- Department of Mathematics and Physics, Basic Medical Science Academy, Air Force Medical University, Xi'an, PR China.
| | - Duolao Wang
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK.
| | - Yonghong Liu
- Department of Neurology, Xijing Hospital, Air Force Medical University, Xi'an, PR China.
| |
Collapse
|
4
|
Rae RJ, Baker NL, Irwin ZT, Shea SD, McMahon LL. Diurnal Modulation of Locus Coeruleus Noradrenergic Neurons in Anesthetized Adult Male Rats. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.06.631545. [PMID: 39829806 PMCID: PMC11741345 DOI: 10.1101/2025.01.06.631545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
The locus coeruleus (LC) is the primary source of noradrenaline (NA) in brain and its activity is essential for learning, memory, stress, arousal, and mood. LC-NA neuron activity varies over the sleep-wake cycle, with higher activity during wakefulness, correlating with increased CSF NA levels. Whether spontaneous and burst firing of LC-NA neurons during active and inactive periods is controlled by mechanisms independent of wakefulness and natural sleep, is currently unknown. Here, using multichannel in vivo electrophysiology under anesthesia, we assessed LC-NA neuron firing in adult male Fisher 344 rats at two different times of day- ZT4- the inactive period (light phase) and ZT16-the active period (dark phase)- independent of contributions from behavioral arousal and natural sleep. In the dark phase, LC-NA neurons exhibit increased average firing rate during baseline compared to the light phase. Using a relatively weak foot shock paradigm, we observed distinct populations of LC-NA neurons with some increasing, and others decreasing, their firing rate compared to baseline. Additionally, while spike frequency during spontaneous and evoked bursts is consistent across the dark-light phase, units recorded during the dark phase have more frequent bursts with a longer duration than those during the light phase. Our findings show that independent of wake state, LC-NA neurons exhibit intrinsic diurnal activity, and that the variability of response to foot shock stimulation demonstrates a physiological heterogeneity of LC-NA neurons that is just beginning to be appreciated. NEW & NOTEWORTHY Multichannel in vivo electrophysiology assesses activity of large populations of NA neurons within an intact LC. Recording activity under anesthesia eliminates influence of behavior and sleep on LC-NA neuron physiology. Our data show that LC-NA neurons have heightened firing and burst activity during the dark phase, suggesting a hardwired diurnal rhythm. Additionally, LC-NA neurons have variable evoked firing highlighting heterogeneity, consistent with a contemporary view that LC physiology is more complex than previously appreciated.
Collapse
|
5
|
Norris MR, Kuo CC, Dunn SS, Kim JR, Becker LJ, Borges G, Thang LV, Parker KE, McCall JG. Mu opioid receptors gate the locus coeruleus pain generator. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.20.562785. [PMID: 37961541 PMCID: PMC10634678 DOI: 10.1101/2023.10.20.562785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
The locus coeruleus (LC) plays a paradoxical role in chronic pain. Although largely known as a potent source of endogenous analgesia, increasing evidence suggests injury can transform the LC into a chronic pain generator. We sought to clarify the role of this system in pain. Here, we show optogenetic inhibition of LC activity is acutely antinociceptive. Following long-term spared nerve injury, the same LC inhibition is analgesic - further supporting its pain generator function. To identify inhibitory substrates that may naturally serve this function, we turned to endogenous LC mu opioid receptors (LC-MOR). These receptors provide powerful LC inhibition and exogenous activation of LC-MOR is antinociceptive. We therefore hypothesized that endogenous LC-MOR-mediated inhibition is critical to how the LC modulates pain. Using cell type-selective conditional knockout and rescue of LC-MOR receptor signaling, we show these receptors bidirectionally regulate thermal and mechanical hyperalgesia - providing a functional gate on the LC pain generator.
Collapse
Affiliation(s)
- Makenzie R. Norris
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA; Department of Pharmaceutical and Administrative Sciences, University of Health Sciences and Pharmacy in St. Louis, St. Louis, MO, USA; Center for Clinical Pharmacology, University of Health Sciences and Pharmacy in St. Louis and Washington University School of Medicine, St. Louis, MO, USA; Washington University Pain Center, Washington University in St. Louis, St. Louis, MO, USA
- Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Chao-Cheng Kuo
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA; Department of Pharmaceutical and Administrative Sciences, University of Health Sciences and Pharmacy in St. Louis, St. Louis, MO, USA; Center for Clinical Pharmacology, University of Health Sciences and Pharmacy in St. Louis and Washington University School of Medicine, St. Louis, MO, USA; Washington University Pain Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Samantha S. Dunn
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA; Department of Pharmaceutical and Administrative Sciences, University of Health Sciences and Pharmacy in St. Louis, St. Louis, MO, USA; Center for Clinical Pharmacology, University of Health Sciences and Pharmacy in St. Louis and Washington University School of Medicine, St. Louis, MO, USA; Washington University Pain Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Jenny R. Kim
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA; Department of Pharmaceutical and Administrative Sciences, University of Health Sciences and Pharmacy in St. Louis, St. Louis, MO, USA; Center for Clinical Pharmacology, University of Health Sciences and Pharmacy in St. Louis and Washington University School of Medicine, St. Louis, MO, USA; Washington University Pain Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Léa J. Becker
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA; Department of Pharmaceutical and Administrative Sciences, University of Health Sciences and Pharmacy in St. Louis, St. Louis, MO, USA; Center for Clinical Pharmacology, University of Health Sciences and Pharmacy in St. Louis and Washington University School of Medicine, St. Louis, MO, USA; Washington University Pain Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Gustavo Borges
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA; Department of Pharmaceutical and Administrative Sciences, University of Health Sciences and Pharmacy in St. Louis, St. Louis, MO, USA; Center for Clinical Pharmacology, University of Health Sciences and Pharmacy in St. Louis and Washington University School of Medicine, St. Louis, MO, USA; Washington University Pain Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Loc V. Thang
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA; Department of Pharmaceutical and Administrative Sciences, University of Health Sciences and Pharmacy in St. Louis, St. Louis, MO, USA; Center for Clinical Pharmacology, University of Health Sciences and Pharmacy in St. Louis and Washington University School of Medicine, St. Louis, MO, USA; Washington University Pain Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Kyle E. Parker
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA; Department of Pharmaceutical and Administrative Sciences, University of Health Sciences and Pharmacy in St. Louis, St. Louis, MO, USA; Center for Clinical Pharmacology, University of Health Sciences and Pharmacy in St. Louis and Washington University School of Medicine, St. Louis, MO, USA; Washington University Pain Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Jordan G. McCall
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA; Department of Pharmaceutical and Administrative Sciences, University of Health Sciences and Pharmacy in St. Louis, St. Louis, MO, USA; Center for Clinical Pharmacology, University of Health Sciences and Pharmacy in St. Louis and Washington University School of Medicine, St. Louis, MO, USA; Washington University Pain Center, Washington University in St. Louis, St. Louis, MO, USA
- Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
6
|
España JC, Yasoda-Mohan A, Vanneste S. The Locus Coeruleus in Chronic Pain. Int J Mol Sci 2024; 25:8636. [PMID: 39201323 PMCID: PMC11354431 DOI: 10.3390/ijms25168636] [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: 06/25/2024] [Revised: 07/30/2024] [Accepted: 08/01/2024] [Indexed: 09/02/2024] Open
Abstract
Pain perception is the consequence of a complex interplay between activation and inhibition. Noradrenergic pain modulation inhibits nociceptive transmission and pain perception. The main source of norepinephrine (NE) in the central nervous system is the Locus Coeruleus (LC), a small but complex cluster of cells in the pons. The aim of this study is to review the literature on the LC-NE inhibitory system, its influence on chronic pain pathways and its frequent comorbidities. The literature research showed that pain perception is the consequence of nociceptive and environmental processing and is modulated by the LC-NE system. If perpetuated in time, nociceptive inputs can generate neuroplastic changes in the central nervous system that reduce the inhibitory effects of the LC-NE complex and facilitate the development of chronic pain and frequent comorbidities, such as anxiety, depression or sleeping disturbances. The exact mechanisms involved in the LC functional shift remain unknown, but there is some evidence that they occur through plastic changes in the medial and lateral pathways and their brain projections. Additionally, there are other influencing factors, like developmental issues, neuroinflammatory glial changes, NE receptor affinity and changes in LC neuronal firing rates.
Collapse
Affiliation(s)
- Jorge Castejón España
- Lab for Clinical and Integrative Neuroscience, Trinity College Institute for Neuroscience, School of Psychology, Trinity College Dublin, D02 PN40 Dublin, Ireland; (J.C.E.); (A.Y.-M.)
- Compass Physio, A83 YW96 Enfield, Ireland
| | - Anusha Yasoda-Mohan
- Lab for Clinical and Integrative Neuroscience, Trinity College Institute for Neuroscience, School of Psychology, Trinity College Dublin, D02 PN40 Dublin, Ireland; (J.C.E.); (A.Y.-M.)
- Global Brain Health Institute, Trinity College Dublin, D02 PN40 Dublin, Ireland
| | - Sven Vanneste
- Lab for Clinical and Integrative Neuroscience, Trinity College Institute for Neuroscience, School of Psychology, Trinity College Dublin, D02 PN40 Dublin, Ireland; (J.C.E.); (A.Y.-M.)
- Global Brain Health Institute, Trinity College Dublin, D02 PN40 Dublin, Ireland
- Brain Research Centre for Advanced, International, Innovative and Interdisciplinary Neuromodulation, 9000 Ghent, Belgium
| |
Collapse
|
7
|
Zouridis IS, Schmors L, Fischer KM, Berens P, Preston-Ferrer P, Burgalossi A. Juxtacellular recordings from identified neurons in the mouse locus coeruleus. Eur J Neurosci 2024; 60:3659-3676. [PMID: 38872397 DOI: 10.1111/ejn.16368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 03/15/2024] [Accepted: 04/11/2024] [Indexed: 06/15/2024]
Abstract
The locus coeruleus (LC) is the primary source of noradrenergic transmission in the mammalian central nervous system. This small pontine nucleus consists of a densely packed nuclear core-which contains the highest density of noradrenergic neurons-embedded within a heterogeneous surround of non-noradrenergic cells. This local heterogeneity, together with the small size of the LC, has made it particularly difficult to infer noradrenergic cell identity based on extracellular sampling of in vivo spiking activity. Moreover, the relatively high cell density, background activity and synchronicity of LC neurons have made spike identification and unit isolation notoriously challenging. In this study, we aimed at bridging these gaps by performing juxtacellular recordings from single identified neurons within the mouse LC complex. We found that noradrenergic neurons (identified by tyrosine hydroxylase, TH, expression; TH-positive) and intermingled putatively non-noradrenergic (TH-negative) cells displayed similar morphologies and responded to foot shock stimuli with excitatory responses; however, on average, TH-positive neurons exhibited more prominent foot shock responses and post-activation firing suppression. The two cell classes also displayed different spontaneous firing rates, spike waveforms and temporal spiking properties. A logistic regression classifier trained on spontaneous electrophysiological features could separate the two cell classes with 76% accuracy. Altogether, our results reveal in vivo electrophysiological correlates of TH-positive neurons, which can be useful for refining current approaches for the classification of LC unit activity.
Collapse
Affiliation(s)
- Ioannis S Zouridis
- Institute of Neurobiology, Eberhard Karls University of Tübingen, Tübingen, Germany
- Werner Reichardt Centre for Integrative Neuroscience, Tübingen, Germany
- Graduate Training Centre of Neuroscience, International Max-Planck Research School (IMPRS), Tübingen, Germany
| | - Lisa Schmors
- Werner Reichardt Centre for Integrative Neuroscience, Tübingen, Germany
- Hertie Institute for AI in Brain Health, University of Tübingen, Tübingen, Germany
| | - Kathrin Maite Fischer
- Institute of Neurobiology, Eberhard Karls University of Tübingen, Tübingen, Germany
- Werner Reichardt Centre for Integrative Neuroscience, Tübingen, Germany
- Graduate Training Centre of Neuroscience, International Max-Planck Research School (IMPRS), Tübingen, Germany
| | - Philipp Berens
- Werner Reichardt Centre for Integrative Neuroscience, Tübingen, Germany
- Hertie Institute for AI in Brain Health, University of Tübingen, Tübingen, Germany
- Tübingen AI Center, University of Tübingen, Tübingen, Germany
| | - Patricia Preston-Ferrer
- Institute of Neurobiology, Eberhard Karls University of Tübingen, Tübingen, Germany
- Werner Reichardt Centre for Integrative Neuroscience, Tübingen, Germany
| | - Andrea Burgalossi
- Institute of Neurobiology, Eberhard Karls University of Tübingen, Tübingen, Germany
- Werner Reichardt Centre for Integrative Neuroscience, Tübingen, Germany
| |
Collapse
|
8
|
Vreven A, Aston-Jones G, Pickering AE, Poe GR, Waterhouse B, Totah NK. In search of the locus coeruleus: guidelines for identifying anatomical boundaries and electrophysiological properties of the blue spot in mice, fish, finches, and beyond. J Neurophysiol 2024; 132:226-239. [PMID: 38842506 PMCID: PMC11383618 DOI: 10.1152/jn.00193.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 06/05/2024] [Indexed: 06/07/2024] Open
Abstract
Our understanding of human brain function can be greatly aided by studying analogous brain structures in other organisms. One brain structure with neurochemical and anatomical homology throughout vertebrate species is the locus coeruleus (LC), a small collection of norepinephrine (NE)-containing neurons in the brainstem that project throughout the central nervous system. The LC is involved in nearly every aspect of brain function, including arousal and learning, which has been extensively examined in rats and nonhuman primates using single-unit recordings. Recent work has expanded into putative LC single-unit electrophysiological recordings in a nonmodel species, the zebra finch. Given the importance of correctly identifying analogous structures as research efforts expand to other vertebrates, we suggest adoption of consensus anatomical and electrophysiological guidelines for identifying LC neurons across species when evaluating brainstem single-unit spiking or calcium imaging. Such consensus criteria will allow for confident cross-species understanding of the roles of the LC in brain function and behavior.
Collapse
Affiliation(s)
- Amelien Vreven
- Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
- Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Gary Aston-Jones
- Brain Health Institute, Rutgers University, Piscataway, New Jersey, United States
| | - Anthony E Pickering
- Anaesthesia, Pain & Critical Care Sciences, School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Gina R Poe
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California, United States
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, California, United States
- Department of Neurobiology, University of California, Los Angeles, California, United States
| | - Barry Waterhouse
- Department of Cell Biology and Neuroscience, Rowan University School of Osteopathic Medicine, Stratford, New Jersey, United States
| | - Nelson K Totah
- Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
- Neuroscience Center, University of Helsinki, Helsinki, Finland
| |
Collapse
|
9
|
Lubejko ST, Livrizzi G, Buczynski SA, Patel J, Yung JC, Yaksh TL, Banghart MR. Inputs to the locus coeruleus from the periaqueductal gray and rostroventral medulla shape opioid-mediated descending pain modulation. SCIENCE ADVANCES 2024; 10:eadj9581. [PMID: 38669335 PMCID: PMC11051679 DOI: 10.1126/sciadv.adj9581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 03/26/2024] [Indexed: 04/28/2024]
Abstract
The supraspinal descending pain modulatory system (DPMS) shapes pain perception via monoaminergic modulation of sensory information in the spinal cord. However, the role and synaptic mechanisms of descending noradrenergic signaling remain unclear. Here, we establish that noradrenergic neurons of the locus coeruleus (LC) are essential for supraspinal opioid antinociception. While much previous work has emphasized the role of descending serotonergic pathways, we find that opioid antinociception is primarily driven by excitatory output from the ventrolateral periaqueductal gray (vlPAG) to the LC. Furthermore, we identify a previously unknown opioid-sensitive inhibitory input from the rostroventromedial medulla (RVM), the suppression of which disinhibits LC neurons to drive spinal noradrenergic antinociception. We describe pain-related activity throughout this circuit and report the presence of prominent bifurcating outputs from the vlPAG to the LC and the RVM. Our findings substantially revise current models of the DPMS and establish a supraspinal antinociceptive pathway that may contribute to multiple forms of descending pain modulation.
Collapse
Affiliation(s)
- Susan T. Lubejko
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
- Neurosciences Graduate Program, University of California, San Diego, La Jolla, CA 92093, USA
| | - Giulia Livrizzi
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
- Biological Sciences Graduate Program, University of California, San Diego, La Jolla, CA 92093, USA
| | - Stanley A. Buczynski
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
- Chemistry and Biochemistry Graduate Program, University of California, San Diego, La Jolla, CA 92093, USA
| | - Janki Patel
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jean C. Yung
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Tony L. Yaksh
- Departments of Anesthesiology and Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Matthew R. Banghart
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
10
|
Grella SL, Donaldson TN. Contextual memory engrams, and the neuromodulatory influence of the locus coeruleus. Front Mol Neurosci 2024; 17:1342622. [PMID: 38375501 PMCID: PMC10875109 DOI: 10.3389/fnmol.2024.1342622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 01/19/2024] [Indexed: 02/21/2024] Open
Abstract
Here, we review the basis of contextual memory at a conceptual and cellular level. We begin with an overview of the philosophical foundations of traversing space, followed by theories covering the material bases of contextual representations in the hippocampus (engrams), exploring functional characteristics of the cells and subfields within. Next, we explore various methodological approaches for investigating contextual memory engrams, emphasizing plasticity mechanisms. This leads us to discuss the role of neuromodulatory inputs in governing these dynamic changes. We then outline a recent hypothesis involving noradrenergic and dopaminergic projections from the locus coeruleus (LC) to different subregions of the hippocampus, in sculpting contextual representations, giving a brief description of the neuroanatomical and physiological properties of the LC. Finally, we examine how activity in the LC influences contextual memory processes through synaptic plasticity mechanisms to alter hippocampal engrams. Overall, we find that phasic activation of the LC plays an important role in promoting new learning and altering mnemonic processes at the behavioral and cellular level through the neuromodulatory influence of NE/DA in the hippocampus. These findings may provide insight into mechanisms of hippocampal remapping and memory updating, memory processes that are potentially dysregulated in certain psychiatric and neurodegenerative disorders.
Collapse
Affiliation(s)
- Stephanie L. Grella
- MNEME Lab, Department of Psychology, Program in Neuroscience, Loyola University Chicago, Chicago, IL, United States
| | - Tia N. Donaldson
- Systems Neuroscience and Behavior Lab, Department of Psychology, The University of New Mexico, Albuquerque, NM, United States
| |
Collapse
|
11
|
Luyo ZNM, Lawrence AB, Stathopoulos TG, Mitrano DA. Localization and neurochemical identity of alpha1-adrenergic receptor-containing elements in the mouse locus coeruleus. J Chem Neuroanat 2023; 133:102343. [PMID: 37777094 PMCID: PMC10842017 DOI: 10.1016/j.jchemneu.2023.102343] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 09/15/2023] [Accepted: 09/25/2023] [Indexed: 10/02/2023]
Abstract
The locus coeruleus (LC) is the major source for norepinephrine (NE) in the brain and projects to areas involved in learning and memory, reward, arousal, attention, and autonomic functions related to stress. There are three types of adrenergic receptors that respond to NE: alpha1-, alpha2-, and beta-adrenergic receptors. Previous behavioral studies have shown the alpha1-adrenergic receptor (α1AR) to be present in the LC, however, with conflicting results. For example, it was shown that α1ARs in the LC are involved in some of the motivational effects of stimulation of the medial forebrain bundle, which was reduced by α1AR antagonist terazosin. Another study showed that during novelty-induced behavioral activation, the α1AR antagonist prazosin reduced c-fos expression in brain regions known to contain motoric α1ARs, except for the LC, where c-fos expression was enhanced. Despite new research delineating more specific connectivity of the neurons in the LC, and some roles of the adrenergic receptors, the α1ARs have not been localized at the subcellular level. Therefore, in order to gain a greater understanding of the aforementioned studies, we used immunohistochemistry at the electron microscopic (EM) level to determine which neuronal or glial elements in the LC express the α1AR. We hypothesized, based on previous work in the ventral periaqueductal gray area, that the α1AR would be found mainly presynaptically in axon terminals, and possibly in glial elements. Single labeling immunohistochemistry at the EM revealed that about 40% of labeled elements that contained the α1AR were glial elements, while approximately 50% of the labeled neuronal elements were axon terminals or small unmyelinated axons in the LC. Double labeling immunohistochemistry found the α1AR expressed in GFAP-labeled astrocytes, in both GABAergic and glutamatergic axon terminals, and in a portion of the α1AR dendrites, colocalized with tyrosine hydroxylase (TH, a marker for noradrenergic neurons). This study sheds light on the neuroanatomical framework underlying the effects of NE and pharmaceuticals acting directly or indirectly on α1ARs in the LC.
Collapse
Affiliation(s)
- Zachary N M Luyo
- Program in Neuroscience, Christopher Newport University, Newport News, VA, USA
| | - Abigail B Lawrence
- Program in Neuroscience, Christopher Newport University, Newport News, VA, USA
| | - Theodore G Stathopoulos
- Department of Molecular Biology & Chemistry, Christopher Newport University, Newport News, VA, USA
| | - Darlene A Mitrano
- Program in Neuroscience, Christopher Newport University, Newport News, VA, USA; Department of Molecular Biology & Chemistry, Christopher Newport University, Newport News, VA, USA.
| |
Collapse
|
12
|
Lubejko ST, Livrizzi G, Patel J, Yung JC, Yaksh TL, Banghart MR. Inputs to the locus coeruleus from the periaqueductal gray and rostroventral medulla shape opioid-mediated descending pain modulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.10.561768. [PMID: 37873091 PMCID: PMC10592708 DOI: 10.1101/2023.10.10.561768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
The supraspinal descending pain modulatory system (DPMS) shapes pain perception via monoaminergic modulation of sensory information in the spinal cord. However, the role and synaptic mechanisms of descending noradrenergic signaling remain unclear. Here, we establish that noradrenergic neurons of the locus coeruleus (LC) are essential for supraspinal opioid antinociception. Unexpectedly, given prior emphasis on descending serotonergic pathways, we find that opioid antinociception is primarily driven by excitatory output from the ventrolateral periaqueductal gray (vlPAG) to the LC. Furthermore, we identify a previously unknown opioid-sensitive inhibitory input from the rostroventromedial medulla (RVM), the suppression of which disinhibits LC neurons to drive spinal noradrenergic antinociception. We also report the presence of prominent bifurcating outputs from the vlPAG to the LC and the RVM. Our findings significantly revise current models of the DPMS and establish a novel supraspinal antinociceptive pathway that may contribute to multiple forms of descending pain modulation.
Collapse
Affiliation(s)
- Susan T. Lubejko
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
- Neurosciences Graduate Program, University of California San Diego, La Jolla, CA 92093, USA
| | - Giulia Livrizzi
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
- Biological Sciences Graduate Program, University of California San Diego, La Jolla, CA 92093, USA
| | - Janki Patel
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Jean C. Yung
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Tony L. Yaksh
- Departments of Anesthesiology and Pharmacology, University of California San Diego, La Jolla, CA, 92093, USA
| | - Matthew R. Banghart
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
13
|
Tanguay E, Bouchard SJ, Lévesque M, De Koninck P, Breton-Provencher V. Shining light on the noradrenergic system. NEUROPHOTONICS 2023; 10:044406. [PMID: 37766924 PMCID: PMC10519836 DOI: 10.1117/1.nph.10.4.044406] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 08/08/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023]
Abstract
Despite decades of research on the noradrenergic system, our understanding of its impact on brain function and behavior remains incomplete. Traditional recording techniques are challenging to implement for investigating in vivo noradrenergic activity, due to the relatively small size and the position in the brain of the locus coeruleus (LC), the primary location for noradrenergic neurons. However, recent advances in optical and fluorescent methods have enabled researchers to study the LC more effectively. Use of genetically encoded calcium indicators to image the activity of noradrenergic neurons and biosensors that monitor noradrenaline release with fluorescence can be an indispensable tool for studying noradrenergic activity. In this review, we examine how these methods are being applied to record the noradrenergic system in the rodent brain during behavior.
Collapse
Affiliation(s)
| | | | - Martin Lévesque
- CERVO Brain Research Centre, Quebec, Quebec, Canada
- Université Laval, Department of Psychiatry and Neuroscience, Faculty of Medicine, Quebec, Quebec, Canada
| | - Paul De Koninck
- CERVO Brain Research Centre, Quebec, Quebec, Canada
- Université Laval, Department of Biochemistry, Microbiology, and Bioinformatics, Faculty of Science and Engineering, Quebec, Quebec, Canada
| | - Vincent Breton-Provencher
- CERVO Brain Research Centre, Quebec, Quebec, Canada
- Université Laval, Department of Psychiatry and Neuroscience, Faculty of Medicine, Quebec, Quebec, Canada
| |
Collapse
|
14
|
Ma HT, Zhang HC, Zuo ZF, Liu YX. Heterogeneous organization of Locus coeruleus: An intrinsic mechanism for functional complexity. Physiol Behav 2023; 268:114231. [PMID: 37172640 DOI: 10.1016/j.physbeh.2023.114231] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/06/2023] [Accepted: 05/09/2023] [Indexed: 05/15/2023]
Abstract
Locus coeruleus (LC) is a small nucleus located deep in the brainstem that contains the majority of central noradrenergic neurons, which provide the primary source of noradrenaline (NA) throughout the entire central nervous system (CNS).The release of neurotransmitter NA is considered to modulate arousal, sensory processing, attention, aversive and adaptive stress responses as well as high-order cognitive function and memory, with the highly ramified axonal arborizations of LC-NA neurons sending wide projections to the targeted brain areas. For over 30 years, LC was thought to be a homogeneous nucleus in structure and function due to the widespread uniform release of NA by LC-NA neurons and simultaneous action in several CNS regions, such as the prefrontal cortex, hippocampus, cerebellum, and spinal cord. However, recent advances in neuroscience tools have revealed that LC is probably not so homogeneous as we previous thought and exhibits heterogeneity in various aspects. Accumulating studies have shown that the functional complexity of LC may be attributed to its heterogeneity in developmental origin, projection patterns, topography distribution, morphology and molecular organization, electrophysiological properties and sex differences. This review will highlight the heterogeneity of LC and its critical role in modulating diverse behavioral outcomes.
Collapse
Affiliation(s)
- Hai-Tao Ma
- Department of Neurobiology, School of Basic Medicine, Jinzhou Medical University, Jinzhou, Liaoning, 121000, China; Department of Neurobiology, School of Basic Medicine, Capital Medical University, Beijing, 100069, China.
| | - Hao-Chen Zhang
- Department of Neurobiology, School of Basic Medicine, Capital Medical University, Beijing, 100069, China
| | - Zhong-Fu Zuo
- Department of Human Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, 121000, China
| | - Ying-Xue Liu
- Department of Human Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, 121000, China.
| |
Collapse
|
15
|
Caramia M, Romanov RA, Syderomenos S, Hevesi Z, Zhao M, Krasniakova M, Xu ZQD, Harkany T, Hökfelt TGM. Neuronal diversity of neuropeptide signaling, including galanin, in the mouse locus coeruleus. Proc Natl Acad Sci U S A 2023; 120:e2222095120. [PMID: 37487094 PMCID: PMC10401028 DOI: 10.1073/pnas.2222095120] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 06/15/2023] [Indexed: 07/26/2023] Open
Abstract
The locus coeruleus (LC) is a small nucleus in the pons from which ascending and descending projections innervate major parts of the central nervous system. Its major transmitter is norepinephrine (NE). This system is evolutionarily conserved, including in humans, and its functions are associated with wakefulness and related to disorders, such as depression. Here, we performed single-cell ribonucleic acid-sequencing (RNA-seq) to subdivide neurons in the LC (24 clusters in total) into 3 NE, 17 glutamate, and 5 γ-aminobutyric acid (GABA) subtypes, and to chart their neuropeptide, cotransmitter, and receptor profiles. We found that NE neurons expressed at least 19 neuropeptide transcripts, notably galanin (Gal) but not Npy, and >30 neuropeptide receptors. Among the galanin receptors, Galr1 was expressed in ~19% of NE neurons, as was also confirmed by in situ hybridization. Unexpectedly, Galr1 was highly expressed in GABA neurons surrounding the NE ensemble. Patch-clamp electrophysiology and cell-type-specific Ca2+-imaging using GCaMP6s revealed that a GalR1 agonist inhibits up to ~35% of NE neurons. This effect is direct and does not rely on feed-forward GABA inhibition. Our results define a role for the galanin system in NE functions, and a conceptual framework for the action of many other peptides and their receptors.
Collapse
Affiliation(s)
- Martino Caramia
- Department of Neuroscience, Biomedicum, Karolinska Institutet, Stockholm17177, Sweden
| | - Roman A. Romanov
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna1090, Austria
| | - Spyridon Syderomenos
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna1090, Austria
| | - Zsofia Hevesi
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna1090, Austria
| | - Ming Zhao
- Department of Neuroscience, Biomedicum, Karolinska Institutet, Stockholm17177, Sweden
| | - Marharyta Krasniakova
- Department of Neuroscience, Biomedicum, Karolinska Institutet, Stockholm17177, Sweden
| | - Zhi-Qing David Xu
- Department of Neurobiology, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute for Brain Disorders, Capital Medical University, Beijing100069, China
- Department of Pathology, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute for Brain Disorders, Capital Medical University, Beijing100069, China
| | - Tibor Harkany
- Department of Neuroscience, Biomedicum, Karolinska Institutet, Stockholm17177, Sweden
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna1090, Austria
| | - Tomas G. M. Hökfelt
- Department of Neuroscience, Biomedicum, Karolinska Institutet, Stockholm17177, Sweden
| |
Collapse
|
16
|
Orlando IF, Shine JM, Robbins TW, Rowe JB, O'Callaghan C. Noradrenergic and cholinergic systems take centre stage in neuropsychiatric diseases of ageing. Neurosci Biobehav Rev 2023; 149:105167. [PMID: 37054802 DOI: 10.1016/j.neubiorev.2023.105167] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 02/28/2023] [Accepted: 03/28/2023] [Indexed: 04/15/2023]
Abstract
Noradrenergic and cholinergic systems are among the most vulnerable brain systems in neuropsychiatric diseases of ageing, including Alzheimer's disease, Parkinson's disease, Lewy body dementia, and progressive supranuclear palsy. As these systems fail, they contribute directly to many of the characteristic cognitive and psychiatric symptoms. However, their contribution to symptoms is not sufficiently understood, and pharmacological interventions targeting noradrenergic and cholinergic systems have met with mixed success. Part of the challenge is the complex neurobiology of these systems, operating across multiple timescales, and with non-linear changes across the adult lifespan and disease course. We address these challenges in a detailed review of the noradrenergic and cholinergic systems, outlining their roles in cognition and behaviour, and how they influence neuropsychiatric symptoms in disease. By bridging across levels of analysis, we highlight opportunities for improving drug therapies and for pursuing personalised medicine strategies.
Collapse
Affiliation(s)
- Isabella F Orlando
- Brain and Mind Centre and School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Australia
| | - James M Shine
- Brain and Mind Centre and School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Australia
| | - Trevor W Robbins
- Behavioural and Clinical Neuroscience Institute and Department of Psychology, University of Cambridge, CB2 3EB, United Kingdom
| | - James B Rowe
- Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust, University of Cambridge, CB2 0SZ, United Kingdom
| | - Claire O'Callaghan
- Brain and Mind Centre and School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Australia.
| |
Collapse
|
17
|
Oh SG, Lee EY, Lee HS. Projections from LIM homeobox 6 (Lhx6) + zona incerta neurons to the cholinergic or monoaminergic nuclei of the rat. Anat Rec (Hoboken) 2023; 306:905-917. [PMID: 36583474 DOI: 10.1002/ar.25143] [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: 07/16/2022] [Revised: 11/03/2022] [Accepted: 12/07/2022] [Indexed: 12/31/2022]
Abstract
A recent report suggested that LIM homeobox 6 (Lhx6) + GABA-releasing neurons of the ventral zona incerta (VZI) promote sleep, particularly paradoxical sleep (PS). While their potential involvement in sleep still needs to be firmly confirmed, little is known about their specific input/output connections with widespread brain regions, including those involved in sleep. Thus, the present study was designed to examine whether Lhx6-expressing neurons (in parallel to intermingled MCH-expressing ones) may send efferent projections to cholinergic and/or monoaminergic nuclei from basal forebrain (BF) to brainstem (BS). Based on the present observations, the proportions of Lhx6+ neuronal projection to the BF and BS cholinergic nuclei over the total number of Lhx6+ VZI cells were approximately 5.9% and 6.9%, respectively. Likewise, the proportions of Lhx6+ neuronal projection to the dorsal raphe and locus coeruleus over the total number of Lhx6+ VZI cells were about 4.3% and 3.9%, respectively. In addition, Lhx6+ cells projecting to the cholinergic or monoaminergic nuclei were scattered along the entire dorsal-to-ventral extent of the VZI. Based on the present as well as our previous observations, it is suggested that Lhx6+ VZI neurons might play an important role in the regulation of PS, partly via the neural network involving the cholinergic as well as monoaminergic nuclei of the rat.
Collapse
Affiliation(s)
- Sung-Gyoon Oh
- Department of Anatomy, School of Medicine, Konkuk University, Seoul, South Korea
| | - Eun-Yeup Lee
- Department of Anatomy, School of Medicine, Konkuk University, Seoul, South Korea
| | - Hyun-Sook Lee
- Department of Anatomy, School of Medicine, Konkuk University, Seoul, South Korea
- Research Institute of Medical Science, School of Medicine, Konkuk University, Seoul, South Korea
| |
Collapse
|
18
|
Kuo CC, Chan H, Hung WC, Chen RF, Yang HW, Min MY. Carbachol increases locus coeruleus activation by targeting noradrenergic neurons, inhibitory interneurons and inhibitory synaptic transmission. Eur J Neurosci 2023; 57:32-53. [PMID: 36382388 DOI: 10.1111/ejn.15866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 10/11/2022] [Accepted: 11/10/2022] [Indexed: 11/18/2022]
Abstract
The locus coeruleus (LC) consists of noradrenergic (NA) neurons and plays an important role in controlling behaviours. Although much of the knowledge regarding LC functions comes from studying behavioural outcomes upon administration of muscarinic acetylcholine receptor (mAChR) agonists into the nucleus, the exact mechanisms remain unclear. Here, we report that the application of carbachol (CCh), an mAChR agonist, increased the spontaneous action potentials (sAPs) of both LC-NA neurons and local inhibitory interneurons (LC I-INs) in acute brain slices by activating M1/M3 mAChRs (m1/3 AChRs). Optogenetic activation of LC I-INs evoked inhibitory postsynaptic currents (IPSCs) in LC-NA neurons that were mediated by γ-aminobutyric acid type A (GABAA ) and glycine receptors, and CCh application decreased the IPSC amplitude through a presynaptic mechanism by activating M4 mAChRs (m4 AChRs). LC-NA neurons also exhibited spontaneous phasic-like activity (sPLA); CCh application increased the incidence of this activity. This effect of CCh application was not observed with blockade of GABAA and glycine receptors, suggesting that the sPLA enhancement occurred likely because of the decreased synaptic transmission of LC I-INs onto LC-NA neurons by the m4 AChR activation and/or increased spiking rate of LC I-INs by the m1/3 AChR activation, which could lead to fatigue of the synaptic transmission. In conclusion, we report that CCh application, while inhibiting their synaptic transmission, increases sAP rates of LC-NA neurons and LC I-INs. Collectively, these effects provide insight into the cellular mechanisms underlying the behaviour modulations following the administration of muscarinic receptor agonists into the LC reported by the previous studies.
Collapse
Affiliation(s)
- Chao-Cheng Kuo
- Department of Life Science, College of Life Science, National Taiwan University, Taipei, Taiwan
| | - Hao Chan
- Department of Life Science, College of Life Science, National Taiwan University, Taipei, Taiwan
| | - Wei-Chen Hung
- Department of Life Science, College of Life Science, National Taiwan University, Taipei, Taiwan
| | - Ruei-Feng Chen
- Department of Life Science, College of Life Science, National Taiwan University, Taipei, Taiwan.,Neurobiology and Cognitive Science Center, National Taiwan University, Taipei, Taiwan
| | - Hsiu-Wen Yang
- Department of Biomedical Sciences, Chung-Shan Medical University, Taichung, Taiwan.,Department of Medical Research, Chung-Shan Medical University Hospital, Taichung, Taiwan
| | - Ming-Yuan Min
- Department of Life Science, College of Life Science, National Taiwan University, Taipei, Taiwan.,Neurobiology and Cognitive Science Center, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
19
|
Dai Z, Liu Y, Nie L, Chen W, Xu X, Li Y, Zhang J, Shen F, Sui N, Liang J. Locus coeruleus input-modulated reactivation of dentate gyrus opioid-withdrawal engrams promotes extinction. Neuropsychopharmacology 2023; 48:327-340. [PMID: 36302846 PMCID: PMC9751301 DOI: 10.1038/s41386-022-01477-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/08/2022] [Accepted: 10/10/2022] [Indexed: 12/26/2022]
Abstract
Extinction training during the reconsolidation window following memory recall is an effective behavioral pattern for promoting the extinction of pathological memory. However, promoted extinction by recall-extinction procedure has not been universally replicated in different studies. One potential reason for this may relate to whether initially acquired memory is successfully activated. Thus, the methods for inducing the memory into an active or plastic condition may contribute to promoting its extinction. The aim of this study is to find and demonstrate a manipulatable neural circuit that engages in the memory recall process and where its activation improves the extinction process through recall-extinction procedure. Here, naloxone-precipitated conditioned place aversion (CPA) in morphine-dependent mice was mainly used as a pathological memory model. We found that the locus coeruleus (LC)-dentate gyrus (DG) circuit was necessary for CPA memory recall and that artificial activation of LC inputs to the DG just prior to initiating a recall-extinction procedure significantly promoted extinction learning. We also found that activating this circuit caused an increase in the ensemble size of DG engram cells activated during the extinction, which was confirmed by a cFos targeted strategy to label cells combined with immunohistochemical and in vivo calcium imaging techniques. Collectively, our data uncover that the recall experience is important for updating the memory during the reconsolidation window; they also suggest a promising neural circuit or target based on the recall-extinction procedure for weakening pathological aversion memory, such as opioid withdrawal memory and fear memory.
Collapse
Affiliation(s)
- Zhonghua Dai
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Ying Liu
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Lina Nie
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Weiqi Chen
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
- Sino-Danish Center for Education and Research, University of Chinese Academy of Sciences, Beijing, China
| | - Xing Xu
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Yonghui Li
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
- Sino-Danish Center for Education and Research, University of Chinese Academy of Sciences, Beijing, China
| | - Jianjun Zhang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Fang Shen
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Nan Sui
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Jing Liang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China.
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China.
- Sino-Danish Center for Education and Research, University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
20
|
Bravo L, Mariscal P, Llorca-Torralba M, López-Cepero JM, Nacher J, Berrocoso E. Altered expression of vesicular glutamate transporter-2 and cleaved caspase-3 in the locus coeruleus of nerve-injured rats. Front Mol Neurosci 2022; 15:918321. [PMID: 35966012 PMCID: PMC9363707 DOI: 10.3389/fnmol.2022.918321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 07/04/2022] [Indexed: 11/15/2022] Open
Abstract
Neuropathic pain is a debilitating chronic condition provoked by a lesion in the nervous system and it induces functional alterations to the noradrenergic locus coeruleus (LC), affecting distinct dimensions of pain, like sensorial hypersensitivity, pain-induced depression, and anxiety. However, the neurobiological changes induced by nerve damage in the LC remain unclear. Here, we analyzed excitatory and inhibitory inputs to the LC, as well as the possible damage that noradrenergic neurons suffer after the induction of neuropathic pain through chronic constriction injury (CCI). Neuropathic pain was induced in male Sprague-Dawley rats, and the expression of the vesicular glutamate transporter 1 or 2 (VGLUT1 or VGLUT2), vesicular GABA transporter (VGAT), and cleaved caspase-3 (CC3) was analyzed by immunofluorescence 7 (CCI7d) or 28 days after the original lesion (CCI28d). While no significant differences in the density of VGLUT1 puncta were evident, CCI7d induced a significant increase in the perisomatic VGLUT2/VGAT ratio relative to Sham-operated and CCI28d animals. By contrast, when the entire region of LC is evaluated, there was a significant reduction in the density of VGLUT2 puncta in CCI28d animals, without changes in VGLUT2/VGAT ratio relative to the CCI7d animals. Additionally, changes in the noradrenergic soma size, and a lower density of mitochondria and lysosomes were evident in CCI28d animals. Interestingly, enhanced expression of the apoptotic marker CC3 was also evident in the CCI28d rats, mainly co-localizing with glial fibrillary acidic protein but not with any neuronal or noradrenergic marker. Overall, short-term pain appears to lead to an increase of markers of excitatory synapses in the perisomatic region of noradrenergic cells in the LC, an effect that is lost after long-term pain, which appears to activate apoptosis.
Collapse
Affiliation(s)
- Lidia Bravo
- Neuropsychopharmacology and Psychobiology Research Group, Department of Neuroscience, University of Cádiz, Cádiz, Spain
- Instituto de Investigación e Innovación Biomédica de Cádiz, INiBICA, Hospital Universitario Puerta del Mar, Cádiz, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
| | - Patricia Mariscal
- Instituto de Investigación e Innovación Biomédica de Cádiz, INiBICA, Hospital Universitario Puerta del Mar, Cádiz, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
- Neuropsychopharmacology and Psychobiology Research Group, Department of Psychology, University of Cádiz, Cádiz, Spain
| | - Meritxell Llorca-Torralba
- Instituto de Investigación e Innovación Biomédica de Cádiz, INiBICA, Hospital Universitario Puerta del Mar, Cádiz, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
- Neuropsychopharmacology and Psychobiology Research Group, Department of Cell Biology and Histology, University of Cádiz, Cádiz, Spain
| | - Jose María López-Cepero
- Neuropsychopharmacology and Psychobiology Research Group, Department of Cell Biology and Histology, University of Cádiz, Cádiz, Spain
| | - Juan Nacher
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
- Neurobiology Unit, Program in Neurosciences and Institute of Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Burjassot, Spain
- Fundación Investigación Hospital Clínico de Valencia, INCLIVA, Valencia, Spain
| | - Esther Berrocoso
- Instituto de Investigación e Innovación Biomédica de Cádiz, INiBICA, Hospital Universitario Puerta del Mar, Cádiz, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
- Neuropsychopharmacology and Psychobiology Research Group, Department of Psychology, University of Cádiz, Cádiz, Spain
| |
Collapse
|
21
|
da Silva Junior CA, Patrone LGA, Biancardi V, Vilela-Costa HH, Marques DA, Cristina-Silva C, da Costa Silva KS, Bícego KC, Szawka RE, Gargaglioni LH. Sexually dimorphic effects of prenatal diazepam exposure on respiratory control and the monoaminergic system of neonate and young rats. Pflugers Arch 2022; 474:1185-1200. [DOI: 10.1007/s00424-022-02730-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/05/2022] [Accepted: 07/08/2022] [Indexed: 11/30/2022]
|
22
|
Kamel LY, Xiong W, Gott BM, Kumar A, Conway CR. Vagus nerve stimulation: An update on a novel treatment for treatment-resistant depression. J Neurol Sci 2022; 434:120171. [DOI: 10.1016/j.jns.2022.120171] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 12/21/2021] [Accepted: 01/21/2022] [Indexed: 12/11/2022]
|
23
|
Borgmann D, Rigoux L, Kuzmanovic B, Edwin Thanarajah S, Münte TF, Fenselau H, Tittgemeyer M. Technical Note: Modulation of fMRI brainstem responses by transcutaneous vagus nerve stimulation. Neuroimage 2021; 244:118566. [PMID: 34509623 DOI: 10.1016/j.neuroimage.2021.118566] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 07/20/2021] [Accepted: 09/07/2021] [Indexed: 01/10/2023] Open
Abstract
Our increasing knowledge about gut-brain interaction is revolutionising the understanding of the links between digestion, mood, health, and even decision making in our everyday lives. In support of this interaction, the vagus nerve is a crucial pathway transmitting diverse gut-derived signals to the brain to monitor of metabolic status, digestive processes, or immune control to adapt behavioural and autonomic responses. Hence, neuromodulation methods targeting the vagus nerve are currently explored as a treatment option in a number of clinical disorders, including diabetes, chronic pain, and depression. The non-invasive variant of vagus nerve stimulation (VNS), transcutaneous auricular VNS (taVNS), has been implicated in both acute and long-lasting effects by modulating afferent vagus nerve target areas in the brain. The physiology of neither of those effects is, however, well understood, and evidence for neuronal response upon taVNS in vagal afferent projection regions in the brainstem and its downstream targets remain to be established. Therefore, to examine time-dependent effects of taVNS on brainstem neuronal responses in healthy human subjects, we applied taVNS during task-free fMRI in a single-blinded crossover design. During fMRI data acquisition, we either stimulated the left earlobe (sham), or the target zone of the auricular branch of the vagus nerve in the outer ear (cymba conchae, verum) for several minutes, both followed by a short 'stimulation OFF' period. Time-dependent effects were assessed by averaging the BOLD response for consecutive 1-minute periods in an ROI-based analysis of the brainstem. We found a significant response to acute taVNS stimulation, relative to the control condition, in downstream targets of vagal afferents, including the nucleus of the solitary tract, the substantia nigra, and the subthalamic nucleus. Most of these brainstem regions remarkably showed increased activity in response to taVNS, and these effect sustained during the post-stimulation period. These data demonstrate that taVNS activates key brainstem regions, and highlight the potential of this approach to modulate vagal afferent signalling. Furthermore, we show that carry-over effects need to be considered when interpreting fMRI data in the context of general vagal neurophysiology and its modulation by taVNS.
Collapse
Affiliation(s)
- Diba Borgmann
- Synaptic Transmission in Energy Homeostasis Group, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany; Translational Neurocircuitry Group, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany; Center for Anatomy II, Neuroanatomy, University Hospital Cologne, Joseph-Stelzmann Str. 9, 50937, Cologne, Germany.
| | - Lionel Rigoux
- Translational Neurocircuitry Group, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany; Translational Neuromodeling Unit, Institute for Biomedical Engineering, Swiss Federal Institute of Technology, Wilfriedstrasse 6, 8032, Zurich, Switzerland
| | - Bojana Kuzmanovic
- Translational Neurocircuitry Group, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany
| | - Sharmili Edwin Thanarajah
- Translational Neurocircuitry Group, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany; Department of Psychiatry, Psychosomatics and Psychotherapy, University Hospital Frankfurt, 60528, Frankfurt, Germany
| | - Thomas F Münte
- Department of Neurology, University of Lübeck, 23538, Lübeck, Germany
| | - Henning Fenselau
- Synaptic Transmission in Energy Homeostasis Group, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany; Cluster of Excellence in Cellular Stress Responses in Aging Associated Diseases (CECAD), University of Cologne, 50931, Cologne, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Strasse 26, 50924, Cologne, Germany
| | - Marc Tittgemeyer
- Translational Neurocircuitry Group, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany; Cluster of Excellence in Cellular Stress Responses in Aging Associated Diseases (CECAD), University of Cologne, 50931, Cologne, Germany
| |
Collapse
|
24
|
Garcia DuBar S, Cosio D, Korthas H, Van Batavia JP, Zderic SA, Sahibzada N, Valentino RJ, Vicini S. Somatostatin Neurons in the Mouse Pontine Nucleus Activate GABA A Receptor Mediated Synaptic Currents in Locus Coeruleus Neurons. Front Synaptic Neurosci 2021; 13:754786. [PMID: 34675794 PMCID: PMC8524133 DOI: 10.3389/fnsyn.2021.754786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 09/10/2021] [Indexed: 11/13/2022] Open
Abstract
The pontine nuclei comprising the locus coeruleus (LC) and Barrington’s nucleus (BRN) amongst others form the neural circuitry(s) that coordinates arousal and voiding behaviors. However, little is known about the synaptic connectivity of neurons within or across these nuclei. These include corticotropin-releasing factor (CRF+) expressing neurons in the BRN that control bladder contraction and somatostatin expressing (SST+) neurons whose role in this region has not been discerned. To determine the synaptic connectivity of these neurons, we employed optogenetic stimulation with recordings from BRN and LC neurons in brain stem slices of channelrhodopsin-2 expressing SST or CRF neurons. Optogenetic stimulation of CRF+ BRN neurons of CrfCre;chr2-yfp mice had little effect on either CRF+ BRN neurons, CRF– BRN neurons, or LC neurons. In contrast, in SstCre;chr2-yfp mice light-activated inhibitory postsynaptic currents (IPSCs) were reliably observed in a majority of LC but not BRN neurons. The GABAA receptor antagonist, bicuculline, completely abolished the light-induced IPSCs. To ascertain if these neurons were part of the neural circuitry that controls the bladder, the trans-synaptic tracer, pseudorabies virus (PRV) was injected into the bladder wall of CrfCre;tdTomato or SstCre;tdTomato mice. At 68–72 h post-viral infection, PRV labeled neurons were present only in the BRN, being preponderant in CRF+ neurons with few SST+ BRN neurons labeled from the bladder. At 76 and 96 h post-virus injection, increased labeling was observed in both BRN and LC neurons. Our results suggest SST+ neurons rather than CRF+ neurons in BRN can regulate the activity of LC neurons.
Collapse
Affiliation(s)
- Selena Garcia DuBar
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, United States
| | - Daniela Cosio
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, United States
| | - Holly Korthas
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC, United States
| | - Jason P Van Batavia
- Division of Urology, The Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Stephen A Zderic
- Division of Urology, The Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Niaz Sahibzada
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, United States.,Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC, United States
| | - Rita J Valentino
- Department of Anesthesiology and Critical Care, The Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Stefano Vicini
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, United States.,Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC, United States
| |
Collapse
|
25
|
McMorris T. The acute exercise-cognition interaction: From the catecholamines hypothesis to an interoception model. Int J Psychophysiol 2021; 170:75-88. [PMID: 34666105 DOI: 10.1016/j.ijpsycho.2021.10.005] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 09/06/2021] [Accepted: 10/11/2021] [Indexed: 02/08/2023]
Abstract
An interoception model for the acute exercise-cognition interaction is presented. During exercise following the norepinephrine threshold, interoceptive feedback induces increased tonic release of extracellular catecholamines, facilitating phasic release hence better cognitive performance of executive functions. When exercise intensity increases to maximum, the nature of task-induced norepinephrine release from the locus coeruleus is dependent on interaction between motivation, perceived effort costs and perceived availability of resources. This is controlled by interaction between the rostral and dorsolateral prefrontal cortices, orbitofrontal cortex, anterior cingulate cortex and anterior insula cortex. If perceived available resources are sufficient to meet predicted effort costs and reward value is high, tonic release from the locus coeruleus is attenuated thus facilitating phasic release, therefore cognition is not inhibited. However, if perceived available resources are insufficient to meet predicted effort costs or reward value is low, tonic release from the locus coeruleus is induced, attenuating phasic release. As a result, cognition is inhibited, although long-term memory and tasks that require switching to new stimuli-response couplings are probably facilitated.
Collapse
Affiliation(s)
- Terry McMorris
- Institute of Sport, University of Chichester, College Lane, Chichester, West Sussex PO19 6PE, United Kingdom; Department of Sport and Exercise Science, Faculty of Science, University of Portsmouth, Guildhall Walk, Portsmouth PO1 2ER, United Kingdom.
| |
Collapse
|
26
|
Broncel A, Bocian R, Kłos-Wojtczak P, Konopacki J. Noradrenergic Profile of Hippocampal Formation Theta Rhythm in Anaesthetized Rats. Neuroscience 2021; 473:13-28. [PMID: 34418519 DOI: 10.1016/j.neuroscience.2021.08.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 08/11/2021] [Accepted: 08/12/2021] [Indexed: 10/20/2022]
Abstract
The present study was undertaken to identify the noradrenergic receptors underlying the production of hippocampal formation (HPC) type 2 theta rhythm. The experiments were performed on urethanized rats wherein type 2 theta is the only rhythm present. In three independent stages of experiments, the effects of noradrenaline (NE) and selective noradrenergic α and β agonists and antagonists were tested. We indicate that the selective activation of three HPC noradrenergic receptors, α1, α2 and β1, induced a similar effect (i.e., inhibition) on type 2 theta rhythm. The remaining HPC β2 and β3 noradrenergic receptors do not seem to be directly involved in the pharmacological mechanism responsible for the suppression of theta rhythm in anaesthetized rats. Obtained results provide evidence for the suppressant effect of exogenous NE on HPC type 2 theta rhythm and show the crucial role of α1, α2 and β1 noradrenergic receptors in the modulation of HPC mechanisms of oscillations and synchrony. This finding is in contrast to the effects of endogenous NE produced by electrical stimulation of the locus coeruleus (LC) and procaine injection into the LC (Broncel et al., 2020).
Collapse
Affiliation(s)
- A Broncel
- Neuromedical, Research Department, Natolin 15, 92-701 Lodz, Poland.
| | - R Bocian
- Department of Neurobiology, Faculty of Biology and Environmental Protection, The University of Lodz, Pomorska St. No 141/143, 90-236 Lodz, Poland.
| | - P Kłos-Wojtczak
- Neuromedical, Research Department, Natolin 15, 92-701 Lodz, Poland.
| | - J Konopacki
- Department of Neurobiology, Faculty of Biology and Environmental Protection, The University of Lodz, Pomorska St. No 141/143, 90-236 Lodz, Poland.
| |
Collapse
|
27
|
Cardenas A, Papadogiannis A, Dimitrov E. The role of medial prefrontal cortex projections to locus ceruleus in mediating the sex differences in behavior in mice with inflammatory pain. FASEB J 2021; 35:e21747. [PMID: 34151467 DOI: 10.1096/fj.202100319rr] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 05/27/2021] [Accepted: 06/07/2021] [Indexed: 11/11/2022]
Abstract
We tested the hypothesis that the cognitive impairment associated with inflammatory pain may result from dysregulation of the top-down control of locus ceruleus's (LC) activity by the medial prefrontal cortex (mPFC). Injection of complete Freund's adjuvant (CFA) served as a model for inflammatory pain. The CFA injection decreased the thermal thresholds in both sexes but only the male mice showed increased anxiety-like behavior and diminished cognition, while the females were not affected. Increased calcium fluorescence, a marker for neuronal activity, was detected by photometry in the mPFC of males but not in females with CFA. Next, while chemogenetic inhibition of the projections from the mPFC to the LC improved the object recognition memory of males with pain, the inhibition of the mPFC to LC pathway in female mice produced anxiolysis and spatial memory deficits. The behavior results prompted us to compare the reciprocal innervation of mPFC and LC between the sexes. We used an anterograde transsynaptic tagging technique, which relies on postsynaptic cre transfer, to assess the innervation of LC by mPFC efferents. The males showed a higher rate of postsynaptic cre transfer into LC neurons from mPFC efferents than the females. And vice versa, a retrograde tracing experiment demonstrated that LC to mPFC projection neurons were more numerous in females when compared to males. In conclusion, we provide evidence that subtle differences in the reciprocal neuronal circuit between the LC and mPFC may contribute to sex differences associated with the adverse cognitive effects of inflammatory pain.
Collapse
Affiliation(s)
- Andrea Cardenas
- Center for the Neurobiology of Stress Resilience and Psychiatric Disorders, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Alexander Papadogiannis
- Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Eugene Dimitrov
- Center for the Neurobiology of Stress Resilience and Psychiatric Disorders, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| |
Collapse
|
28
|
Breton-Provencher V, Drummond GT, Sur M. Locus Coeruleus Norepinephrine in Learned Behavior: Anatomical Modularity and Spatiotemporal Integration in Targets. Front Neural Circuits 2021; 15:638007. [PMID: 34163331 PMCID: PMC8215268 DOI: 10.3389/fncir.2021.638007] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 05/03/2021] [Indexed: 12/16/2022] Open
Abstract
The locus coeruleus (LC), a small brainstem nucleus, is the primary source of the neuromodulator norepinephrine (NE) in the brain. The LC receives input from widespread brain regions, and projects throughout the forebrain, brainstem, cerebellum, and spinal cord. LC neurons release NE to control arousal, but also in the context of a variety of sensory-motor and behavioral functions. Despite its brain-wide effects, much about the role of LC-NE in behavior and the circuits controlling LC activity is unknown. New evidence suggests that the modular input-output organization of the LC could enable transient, task-specific modulation of distinct brain regions. Future work must further assess whether this spatial modularity coincides with functional differences in LC-NE subpopulations acting at specific times, and how such spatiotemporal specificity might influence learned behaviors. Here, we summarize the state of the field and present new ideas on the role of LC-NE in learned behaviors.
Collapse
Affiliation(s)
| | | | - Mriganka Sur
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, United States
| |
Collapse
|
29
|
Bostanciklioğlu M. Unexpected awakenings in severe dementia from case reports to laboratory. Alzheimers Dement 2020; 17:125-136. [PMID: 33064369 DOI: 10.1002/alz.12162] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 06/17/2020] [Accepted: 07/07/2020] [Indexed: 11/12/2022]
Abstract
OBJECTIVE Case report notions of unexpected memory retrieval in patients with severe dementia near to death are starting to alter the central "irreversible" paradigm of dementia and locate dementia as a problem of memory retrieval, not consolidation. We suggest that the most likely central tenet of this paradoxical memory retrieval is the fluctuation of neuromodulators projecting from the brain stem to the medial prefrontal cortex and the hippocampus. The neuromodulation-centric explanation of this phenomenon aims to open the "irreversible" paradigm of dementia up for discussion and suggest a plausible treatment strategy by questioning how the devastating process of death fluctuates memory performance in severe dementia. BACKGROUND Supporting demented patients, who are mostly unresponsive, without making demands or asking a question and regarding them as valuable human beings unexpectedly improve their memory performance around the time of death. NEW LUCIDITY HYPOTHESIS Around the time of death, neurological signs (hyper-arousal and -attention) of demented people point out that neurotransmitter discharges are dramatically changed. Relatively resistant neuromodulator circuits to neurodegeneration can maintain optimal levels of arousal and attention for memory processing. In this way, unexpected episodes of lucidity can be triggered. Also, corticotropin-releasing peptides might increase mental clarity by increasing the excitability of the neuromodulator circuits. The science of memory retrieval is more complicated and nuanced than retrieval observations in case reports, but the rapid development of new techniques holds promise for future understanding of lucidity in severe dementia. MAJOR CHALLENGE FOR THE MODEL There is no an animal or human model to test this hypothesis; however, the similarities between neurological signs (instantaneous cognitive fluctuations) of delirium and paradoxical lucidity could provide a unique window to understand neural events of terminal lucidity on a modified animal model of delirium. Likewise, similarities between unexpected consciousness signs of terminal lucidity and lucid dreaming suggest that lucid dreaming episodes might be considered a human model for terminal lucidity research.
Collapse
|
30
|
Kelberman M, Keilholz S, Weinshenker D. What's That (Blue) Spot on my MRI? Multimodal Neuroimaging of the Locus Coeruleus in Neurodegenerative Disease. Front Neurosci 2020; 14:583421. [PMID: 33122996 PMCID: PMC7573566 DOI: 10.3389/fnins.2020.583421] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 09/16/2020] [Indexed: 01/04/2023] Open
Abstract
The locus coeruleus (LC) has long been underappreciated for its role in the pathophysiology of Alzheimer’s disease (AD), Parkinson’s disease (PD), and other neurodegenerative disorders. While AD and PD are distinct in clinical presentation, both are characterized by prodromal protein aggregation in the LC, late-stage degeneration of the LC, and comorbid conditions indicative of LC dysfunction. Many of these early studies were limited to post-mortem histological techniques due to the LC’s small size and location deep in the brainstem. Thus, there is a growing interest in utilizing in vivo imaging of the LC as a predictor of preclinical neurodegenerative processes and biomarker of disease progression. Simultaneously, neuroimaging in animal models of neurodegenerative disease holds promise for identifying early alterations to LC circuits, but has thus far been underutilized. While still in its infancy, a handful of studies have reported effects of single gene mutations and pathology on LC function in disease using various neuroimaging techniques. Furthermore, combining imaging and optogenetics or chemogenetics allows for interrogation of network connectivity in response to changes in LC activity. The purpose of this article is twofold: (1) to review what magnetic resonance imaging (MRI) and positron emission tomography (PET) have revealed about LC dysfunction in neurodegenerative disease and its potential as a biomarker in humans, and (2) to explore how animal models can be used to test hypotheses derived from clinical data and establish a mechanistic framework to inform LC-focused therapeutic interventions to alleviate symptoms and impede disease progression.
Collapse
Affiliation(s)
- Michael Kelberman
- Department of Human Genetics, Emory University, Atlanta, GA, United States
| | - Shella Keilholz
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Emory University, Atlanta, GA, United States
| | - David Weinshenker
- Department of Human Genetics, Emory University, Atlanta, GA, United States
| |
Collapse
|
31
|
|
32
|
Kuo CC, Hsieh JC, Tsai HC, Kuo YS, Yau HJ, Chen CC, Chen RF, Yang HW, Min MY. Inhibitory interneurons regulate phasic activity of noradrenergic neurons in the mouse locus coeruleus and functional implications. J Physiol 2020; 598:4003-4029. [PMID: 32598024 DOI: 10.1113/jp279557] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 06/25/2020] [Indexed: 01/16/2023] Open
Abstract
KEY POINTS The locus coeruleus (LC) contains noradrenergic (NA) neurons that respond to novel stimuli in the environment with phasic activation to initiate an orienting response; phasic LC activation is also triggered by stimuli, representing the outcome of task-related decision processes, to facilitate ensuing behaviours and help optimize task performance. Here, we report that LC-NA neurons exhibit bursts of action potentials in vitro resembling phasic LC activation in vivo, and the activity is gated by inhibitory interneurons (I-INs) located in the peri-LC. We also observe that inhibition of peri-LC I-INs enhances prepulse inhibition and axons from cortical areas that play important roles in evaluating the cost/reward of a stimulus synapse on both peri-LC I-INs and LC-NA neurons. The results help us understand the cellular mechanisms underlying the generation and regulation of phasic LC activation with a focus on the role of peri-LC I-INs. ABSTRACT Noradrenergic (NA) neurons in the locus coeruleus (LC) have global axonal projection to the brain. These neurons discharge action potentials phasically in response to either novel stimuli in the environment to initiate an orienting behaviour or stimuli representing the outcome of task-related decision processes to facilitate ensuing behaviours and help optimize task performance. Nevertheless, the cellular mechanisms underlying the generation and regulation of phasic LC activation remain unknown. We report here that LC-NA neurons recorded in brain slices exhibit bursts of action potentials that resembled the phasic activation-pause profile observed in animals. The activity was referred to as phasic-like activity (PLA) and was suppressed and enhanced by blocking excitatory and inhibitory synaptic transmissions, respectively. These results suggest the existence of a local circuit to drive PLA, and the activity could be regulated by the excitatory-inhibitory balance of the circuit. In support of this notion, we located a population of inhibitory interneurons (I-INs) in the medial part of the peri-LC that exerted feedforward inhibition of LC-NA neurons through GABAergic and glycinergic transmissions. Selective inhibition of peri-LC I-INs with chemogenetic methods could enhance PLA in brain slices and increase prepulse inhibition in animals. Moreover, axons from the orbitofrontal and prelimbic cortices, which play important roles in evaluating the cost/reward of a stimulus, synapse on both peri-LC I-INs and LC-NA neurons. These observations demonstrate functional roles of peri-LC I-INs in integrating inputs of the frontal cortex onto LC-NA neurons and gating the phasic LC output.
Collapse
Affiliation(s)
- Chao-Cheng Kuo
- Department of Life Science, College of Life Science, National Taiwan University, Taipei, 10617, Taiwan
| | - Jung-Chien Hsieh
- Department of Life Science, College of Life Science, National Taiwan University, Taipei, 10617, Taiwan
| | - Hsing-Chun Tsai
- Department of Life Science, College of Life Science, National Taiwan University, Taipei, 10617, Taiwan
| | - Yu-Shan Kuo
- Department of Life Science, College of Life Science, National Taiwan University, Taipei, 10617, Taiwan.,Departments of Biomedical Sciences and Medical Research, Chung-Shan Medical University and Chung-Shan Medical University Hospital, Taichung, 40201, Taiwan
| | - Hau-Jie Yau
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Chih-Cheng Chen
- Institute of Biomedical Science, Academia Sinica, Taipei, 11529, Taiwan
| | - Ruei-Feng Chen
- Department of Life Science, College of Life Science, National Taiwan University, Taipei, 10617, Taiwan
| | - Hsiu-Wen Yang
- Departments of Biomedical Sciences and Medical Research, Chung-Shan Medical University and Chung-Shan Medical University Hospital, Taichung, 40201, Taiwan
| | - Ming-Yuan Min
- Department of Life Science, College of Life Science, National Taiwan University, Taipei, 10617, Taiwan
| |
Collapse
|
33
|
Johnson CM, Cui N, Xing H, Wu Y, Jiang C. The antitussive cloperastine improves breathing abnormalities in a Rett Syndrome mouse model by blocking presynaptic GIRK channels and enhancing GABA release. Neuropharmacology 2020; 176:108214. [PMID: 32622786 DOI: 10.1016/j.neuropharm.2020.108214] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 06/12/2020] [Accepted: 06/14/2020] [Indexed: 12/21/2022]
Abstract
Rett Syndrome (RTT) is an X-linked neurodevelopmental disorder caused mainly by mutations in the MECP2 gene. One of the major RTT features is breathing dysfunction characterized by periodic hypo- and hyperventilation. The breathing disorders are associated with increased brainstem neuronal excitability, which can be alleviated with GABA agonists. Since neuronal hypoexcitability occurs in the forebrain of RTT models, it is necessary to find pharmacological agents with a relative preference to brainstem neurons. Here we show evidence for the improvement of breathing disorders of Mecp2-disrupted mice with the brainstem-acting drug cloperastine (CPS) and its likely neuronal targets. CPS is an over-the-counter cough medicine that has an inhibitory effect on brainstem neuronal networks. In Mecp2-disrupted mice, CPS (30 mg/kg, i.p.) decreased the occurrence of apneas/h and breath frequency variation. GIRK currents expressed in HEK cells were inhibited by CPS with IC50 1 μM. Whole-cell patch clamp recordings in locus coeruleus (LC) and dorsal tegmental nucleus (DTN) neurons revealed an overall inhibitory effect of CPS (10 μM) on neuronal firing activity. Such an effect was reversed by the GABAA receptor antagonist bicuculline (20 μM). Voltage clamp studies showed that CPS increased GABAergic sIPSCs in LC cells, which was blocked by the GABAB receptor antagonist phaclofen. Functional GABAergic connections of DTN neurons with LC cells were shown. These results suggest that CPS improves breathing dysfunction in Mecp2-null mice by blocking GIRK channels in synaptic terminals and enhancing GABA release.
Collapse
Affiliation(s)
- Christopher M Johnson
- Department of Biology, Georgia State University, 100 Piedmont Avenue, Atlanta, GA, 30303, USA
| | - Ningren Cui
- Department of Biology, Georgia State University, 100 Piedmont Avenue, Atlanta, GA, 30303, USA
| | - Hao Xing
- Department of Biology, Georgia State University, 100 Piedmont Avenue, Atlanta, GA, 30303, USA
| | - Yang Wu
- Department of Biology, Georgia State University, 100 Piedmont Avenue, Atlanta, GA, 30303, USA
| | - Chun Jiang
- Department of Biology, Georgia State University, 100 Piedmont Avenue, Atlanta, GA, 30303, USA.
| |
Collapse
|
34
|
Paredes-Rodriguez E, Vegas-Suarez S, Morera-Herreras T, De Deurwaerdere P, Miguelez C. The Noradrenergic System in Parkinson's Disease. Front Pharmacol 2020; 11:435. [PMID: 32322208 PMCID: PMC7157437 DOI: 10.3389/fphar.2020.00435] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 03/20/2020] [Indexed: 12/16/2022] Open
Abstract
Nowadays it is well accepted that in Parkinson’s disease (PD), the neurodegenerative process occurs in stages and that damage to other areas precedes the neuronal loss in the substantia nigra pars compacta, which is considered a pathophysiological hallmark of PD. This heterogeneous and progressive neurodegeneration may explain the diverse symptomatology of the disease, including motor and non-motor alterations. In PD, one of the first areas undergoing degeneration is the locus coeruleus (LC). This noradrenergic nucleus provides extensive innervation throughout the brain and plays a fundamental neuromodulator role, participating in stress responses, emotional memory, and control of motor, sensory, and autonomic functions. Early in the disease, LC neurons suffer modifications that can condition the effectiveness of pharmacological treatments, and importantly, can lead to the appearance of common non-motor symptomatology. The noradrenergic system also exerts anti-inflammatory and neuroprotective effect on the dopaminergic degeneration and noradrenergic damage can consequently condition the progress of the disease. From the pharmacological point of view, it is also important to understand how the noradrenergic system performs in PD, since noradrenergic medication is often used in these patients, and drug interactions can take place when combining them with the gold standard drug therapy in PD, L-3,4-dihydroxyphenylalanine (L-DOPA). This review provides an overview about the functional status of the noradrenergic system in PD and its contribution to the efficacy of pharmacological-based treatments. Based on preclinical and clinical publications, a special attention will be dedicated to the most prevalent non-motor symptoms of the disease.
Collapse
Affiliation(s)
- Elena Paredes-Rodriguez
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain.,Autonomic and Movement Disorders Unit, Neurodegenerative Diseases, Biocruces Health Research Institute, Barakaldo, Spain
| | - Sergio Vegas-Suarez
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain.,Autonomic and Movement Disorders Unit, Neurodegenerative Diseases, Biocruces Health Research Institute, Barakaldo, Spain
| | - Teresa Morera-Herreras
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain.,Autonomic and Movement Disorders Unit, Neurodegenerative Diseases, Biocruces Health Research Institute, Barakaldo, Spain
| | - Philippe De Deurwaerdere
- Centre National de la Recherche scientifique, Institut des Neurosciences Cognitives et Intégratives d'Aquitaine (INCIA UMR 5287), Bordeaux, France
| | - Cristina Miguelez
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain.,Autonomic and Movement Disorders Unit, Neurodegenerative Diseases, Biocruces Health Research Institute, Barakaldo, Spain
| |
Collapse
|
35
|
Torralva R, Janowsky A. Noradrenergic Mechanisms in Fentanyl-Mediated Rapid Death Explain Failure of Naloxone in the Opioid Crisis. J Pharmacol Exp Ther 2019; 371:453-475. [PMID: 31492824 PMCID: PMC6863461 DOI: 10.1124/jpet.119.258566] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 09/03/2019] [Indexed: 12/25/2022] Open
Abstract
In December 2018, the Centers for Disease Control declared fentanyl the deadliest drug in America. Opioid overdose is the single greatest cause of death in the United States adult population (ages 18-50), and fentanyl and its analogs [fentanyl/fentanyl analogs (F/FAs)] are currently involved in >50% of these deaths. Anesthesiologists in the United States were introduced to fentanyl in the early 1970s when it revolutionized surgical anesthesia by combining profound analgesia with hemodynamic stability. However, they quickly had to master its unique side effect. F/FAs can produce profound rigidity in the diaphragm, chest wall and upper airway within an extremely narrow dosing range. This clinical effect was called wooden chest syndrome (WCS) by anesthesiologists and is not commonly known outside of anesthesiology or to clinicians or researchers in addiction research/medicine. WCS is almost routinely fatal without expert airway management. This review provides relevant clinical human pharmacology and animal data demonstrating that the significant increase in the number of F/FA-induced deaths may involve α-adrenergic and cholinergic receptor-mediated mechanical failure of the respiratory and cardiovascular systems with rapid development of rigidity and airway closure. Although morphine and its prodrug, heroin, can cause mild rigidity in abdominal muscles at high doses, neither presents with the distinct and rapid respiratory failure seen with F/FA-induced WCS, separating F/FA overdose from the slower onset of respiratory depression caused by morphine-derived alkaloids. This distinction has significant consequences for the design and implementation of new pharmacologic strategies to effectively prevent F/FA-induced death. SIGNIFICANCE STATEMENT: Deaths from fentanyl and F/FAs are increasing in spite of availability and awareness of the opioid reversal drug naloxone. This article reviews literature suggesting that naloxone may be ineffective against centrally mediated noradrenergic and cholinergic effects of F/FAs, which clinically manifest as severe muscle rigidity and airway compromise (e.g., wooden chest syndrome) that is rapid and distinct from respiratory depression seen with morphine-derived alkaloids. A physiologic model is proposed and implications for new drug development and treatment are discussed.
Collapse
Affiliation(s)
- Randy Torralva
- CODA Inc., Research Department, Portland, Oregon (R.T.); Research Service, VA Portland Health Care System, Portland, Oregon (R.T., A.J.); and Department of Psychiatry, Oregon Health & Science University, Portland, Oregon (R.T., A.J.)
| | - Aaron Janowsky
- CODA Inc., Research Department, Portland, Oregon (R.T.); Research Service, VA Portland Health Care System, Portland, Oregon (R.T., A.J.); and Department of Psychiatry, Oregon Health & Science University, Portland, Oregon (R.T., A.J.)
| |
Collapse
|
36
|
Neuroprotective effect of exogenous melatonin on the noradrenergic neurons of adult male rats’ locus coeruleus nucleus following REM sleep deprivation. J Chem Neuroanat 2019; 100:101656. [DOI: 10.1016/j.jchemneu.2019.101656] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 06/13/2019] [Accepted: 06/15/2019] [Indexed: 12/17/2022]
|
37
|
Neuromodulation in circuits of aversive emotional learning. Nat Neurosci 2019; 22:1586-1597. [PMID: 31551602 DOI: 10.1038/s41593-019-0503-3] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 08/21/2019] [Indexed: 02/07/2023]
Abstract
Emotional learning and memory are functionally and dysfunctionally regulated by the neuromodulatory state of the brain. While the role of excitatory and inhibitory neural circuits mediating emotional learning and its control have been the focus of much research, we are only now beginning to understand the more diffuse role of neuromodulation in these processes. Recent experimental studies of the acetylcholine, noradrenaline and dopamine systems in fear learning and extinction of fear responding provide surprising answers to key questions in neuromodulation. One area of research has revealed how modular organization, coupled with context-dependent coding modes, allows for flexible brain-wide or targeted neuromodulation. Other work has shown how these neuromodulators act in downstream targets to enhance signal-to-noise ratios and gain, as well as to bind distributed circuits through neuronal oscillations. These studies elucidate how different neuromodulatory systems regulate aversive emotional processing and reveal fundamental principles of neuromodulatory function.
Collapse
|
38
|
Sex differences in breathing. Comp Biochem Physiol A Mol Integr Physiol 2019; 238:110543. [PMID: 31445081 DOI: 10.1016/j.cbpa.2019.110543] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 08/08/2019] [Accepted: 08/09/2019] [Indexed: 01/15/2023]
Abstract
Breathing is a vital behavior that ensures both the adequate supply of oxygen and the elimination of CO2, and it is influenced by many factors. Despite that most of the studies in respiratory physiology rely heavily on male subjects, there is much evidence to suggest that sex is an important factor in the respiratory control system, including the susceptibility for some diseases. These different respiratory responses in males and females may be related to the actions of sex hormones, especially in adulthood. These hormones affect neuromodulatory systems that influence the central medullary rhythm/pontine pattern generator and integrator, sensory inputs to the integrator and motor output to the respiratory muscles. In this article, we will first review the sex dependence on the prevalence of some respiratory-related diseases. Then, we will discuss the role of sex and gonadal hormones in respiratory control under resting conditions and during respiratory challenges, such as hypoxia and hypercapnia, and whether hormonal fluctuations during the estrous/menstrual cycle affect breathing control. We will then discuss the role of the locus coeruleus, a sexually dimorphic CO2/pH-chemosensitive nucleus, on breathing regulation in males and females. Next, we will highlight the studies that exist regarding sex differences in respiratory control during development. Finally, the few existing studies regarding the influence of sex on breathing control in non-mammalian vertebrates will be discussed.
Collapse
|
39
|
Totah NK, Logothetis NK, Eschenko O. Noradrenergic ensemble-based modulation of cognition over multiple timescales. Brain Res 2019; 1709:50-66. [DOI: 10.1016/j.brainres.2018.12.031] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 12/11/2018] [Accepted: 12/21/2018] [Indexed: 11/30/2022]
|
40
|
Breton-Provencher V, Sur M. Active control of arousal by a locus coeruleus GABAergic circuit. Nat Neurosci 2019; 22:218-228. [PMID: 30643295 PMCID: PMC6385895 DOI: 10.1038/s41593-018-0305-z] [Citation(s) in RCA: 208] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 11/26/2018] [Indexed: 12/22/2022]
Abstract
Arousal responses linked to locus coeruleus noradrenergic (LC-NA) activity affect cognition. However, the mechanisms that control modes of LC-NA activity remain unknown. Here, we reveal a local population of GABAergic neurons (LC-GABA) capable of modulating LC-NA activity and arousal. Retrograde tracing shows that inputs to LC-GABA and LC-NA neurons arise from similar regions, though a few regions provide differential inputs to one subtype over the other. Recordings in the locus coeruleus demonstrate two modes of LC-GABA responses whereby spiking is either correlated or broadly anticorrelated with LC-NA responses, reflecting anatomically similar and functionally coincident inputs, or differential and non-coincident inputs, to LC-NA and LC-GABA neurons. Coincident inputs control the gain of LC-NA-mediated arousal responses, whereas non-coincident inputs, such as from the prefrontal cortex to the locus coeruleus, alter global arousal levels. These findings demonstrate distinct modes by which an inhibitory locus coeruleus circuit regulates arousal in the brain.
Collapse
Affiliation(s)
- Vincent Breton-Provencher
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Mriganka Sur
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
41
|
Campos-Lira E, Kelly L, Seifi M, Jackson T, Giesecke T, Mutig K, Koshimizu TAA, Hernandez VS, Zhang L, Swinny JD. Dynamic Modulation of Mouse Locus Coeruleus Neurons by Vasopressin 1a and 1b Receptors. Front Neurosci 2018; 12:919. [PMID: 30618551 PMCID: PMC6295453 DOI: 10.3389/fnins.2018.00919] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 11/22/2018] [Indexed: 11/29/2022] Open
Abstract
The locus coeruleus (LC) is a brainstem nucleus distinguished by its supply of noradrenaline throughout the central nervous system. Apart from modulating a range of brain functions, such as arousal, cognition and the stress response, LC neuronal excitability also corresponds to the activity of various peripheral systems, such as pelvic viscera and the cardiovascular system. Neurochemically diverse inputs set the tone for LC neuronal activity, which in turn modulates these adaptive physiological and behavioral responses essential for survival. One such LC afferent system which is poorly understood contains the neurohormone arginine-vasopressin (AVP). Here we provide the first demonstration of the molecular and functional characteristics of the LC-AVP system, by characterizing its receptor-specific modulation of identified LC neurons and plasticity in response to stress. High resolution confocal microscopy revealed that immunoreactivity for the AVP receptor 1b (V1b) was located on plasma membranes of noradrenergic and non-noradrenergic LC neurons. In contrast, immunoreactivity for the V1a receptor was exclusively located on LC noradrenergic neurons. No specific signal, either at the mRNA or protein level, was detected for the V2 receptor in the LC. Clusters immunoreactive for V1a-b were located in proximity to profiles immunoreactive for GABAergic and glutamatergic synaptic marker proteins. AVP immunopositive varicosities were also located adjacent to labeling for such synaptic markers. Whole-cell patch clamp electrophysiology revealed that the pharmacological activation of V1b receptors significantly increased the spontaneous activity of 45% (9/20) of recorded noradrenergic neurons, with the remaining 55% (11/20) of cells exhibiting a significant decrease in their basal firing patterns. Blockade of V1a and V1b receptors on their own significantly altered LC neuronal excitability in a similar heterogeneous manner, demonstrating that endogenous AVP sets the basal LC neuronal firing rates. Finally, exposing animals to acute stress increased V1b, but not V1a receptor expression, whilst decreasing AVP immunoreactivity. This study reveals the AVP-V1a-b system as a considerable component of the LC molecular architecture and regulator of LC activity. Since AVP primarily functions as a regulator of homeostasis, the data suggest a novel pathway by modulating the functioning of a brain region that is integral to mediating adaptive responses.
Collapse
Affiliation(s)
- Elba Campos-Lira
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Louise Kelly
- Institute for Biomedical and Biomolecular Sciences, School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom
| | - Mohsen Seifi
- Institute for Biomedical and Biomolecular Sciences, School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom
| | - Torquil Jackson
- Institute for Biomedical and Biomolecular Sciences, School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom
| | - Torsten Giesecke
- Department of Anatomy, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Kerim Mutig
- Department of Anatomy, Charité - Universitätsmedizin Berlin, Berlin, Germany.,I.M. Sechenov First Moscow State Medical University of the Ministry of Healthcare of the Russian Federation (Sechenovskiy University), Moscow, Russia
| | - Taka-Aki A Koshimizu
- Division of Molecular Pharmacology, Department of Pharmacology, Jichi Medical University, Shimotsuke, Japan
| | - Vito S Hernandez
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Limei Zhang
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Jerome D Swinny
- Institute for Biomedical and Biomolecular Sciences, School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom
| |
Collapse
|
42
|
Foote SL, Berridge CW. New developments and future directions in understanding locus coeruleus - Norepinephrine (LC-NE) function. Brain Res 2018; 1709:81-84. [PMID: 30267649 DOI: 10.1016/j.brainres.2018.09.033] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 09/24/2018] [Indexed: 12/29/2022]
Abstract
In this commentary we utilize recent observations regarding the organization and actions of the locus coeruleus-noradrenergic system to identify major issues in need of further study to more fully understand the behavioral actions of this major neurotransmitter system.
Collapse
Affiliation(s)
- Stephen L Foote
- Psychology Department, University of Wisconsin-Madison, Madison, WI 53706, United States
| | - Craig W Berridge
- Psychology Department, University of Wisconsin-Madison, Madison, WI 53706, United States.
| |
Collapse
|
43
|
The Mechanism of Action of Vagus Nerve Stimulation in Treatment-Resistant Depression: Current Conceptualizations. Psychiatr Clin North Am 2018; 41:395-407. [PMID: 30098653 DOI: 10.1016/j.psc.2018.04.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Stimulation of the left cervical vagus nerve, or vagus nerve stimulation (VNS), brings about an antidepressant response in a subset of treatment-resistant depression (TRD) patients. How this occurs is poorly understood; however, knowledge of the neuroanatomic vagal pathways, in conjunction with functional brain imaging studies, suggests several brain regions associated with mood regulation are critical: brainstem nuclei (locus coeruleus, dorsal raphe, and ventral tegmental area), thalamus, and insular and prefrontal cortex. Furthermore, animal studies suggest that VNS enhances neuroplasticity and changes in neuronal firing patterns. Continued study to better understand the mechanism of action of VNS in TRD is warranted.
Collapse
|
44
|
Kohtz AS, Lin B, Smith ME, Aston-Jones G. Attenuated cocaine-seeking after oxytocin administration in male and female rats. Psychopharmacology (Berl) 2018; 235:2051-2063. [PMID: 29671014 PMCID: PMC6015788 DOI: 10.1007/s00213-018-4902-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 04/09/2018] [Indexed: 12/20/2022]
Abstract
RATIONALE Initial drug abstinence (modeled here as Extinction Day 1, ED1) is a critical time point in the progression of addiction that is strongly influenced by stress and sex. ED1 induces corticosterone release in both sexes, and cocaine-seeking during ED1 can be mitigated by corticotrophin-releasing factor (CRF) antagonists more effectively in female rats. Oxytocin (OXT) is a neuropeptide that has several biological functions, including regulation of stress pathways. METHODS To investigate a relationship between OXT, sex, and cocaine-seeking, we examined Fos on ED1 in OXT neurons of paraventricular (PVN) and supraoptic nuclei (SON) compared to homecage (cocaine experienced) or naïve male and female rats. We also administered OXT 30 min prior to ED1 testing or cued reinstatement testing. RESULTS OXT neurons had decreased activity (as reflected by Fos protein) in PVN and SON on withdrawal day 1 (homecage) compared to naïve rats. Fos in OXT neurons was further decreased on ED1, compared to homecage controls, in both males and females even though in SON, cocaine exposure increased the number of OXT-expressing neurons. In addition, systemically administered OXT reduced cocaine-seeking during ED1 and cue-induced reinstatement of cocaine-seeking but delayed extinction, similarly among male and female rats. CONCLUSIONS These data indicate that OXT neurons in PVN and SON may be involved in cocaine-seeking during ED1 and support OXT as a possible therapeutic to decrease cocaine-seeking during initial abstinence and in response to cocaine-associated cues.
Collapse
Affiliation(s)
- Amy S. Kohtz
- Brain Health Institute, Rutgers University, Piscataway, NJ 08854,Department of Pharmacology and Toxicology, Rutgers University, Piscataway, NJ 08854
| | - Belle Lin
- Brain Health Institute, Rutgers University, Piscataway, NJ 08854
| | | | - Gary Aston-Jones
- Brain Health Institute, Rutgers University, RWJMS Research Building Rm. 259, 683 Hoes Lane West, Piscataway, NJ, 08854, USA.
| |
Collapse
|
45
|
Melnychuk MC, Dockree PM, O'Connell RG, Murphy PR, Balsters JH, Robertson IH. Coupling of respiration and attention via the locus coeruleus: Effects of meditation and pranayama. Psychophysiology 2018; 55:e13091. [PMID: 29682753 DOI: 10.1111/psyp.13091] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 03/06/2018] [Accepted: 03/13/2018] [Indexed: 01/26/2023]
Abstract
The locus coeruleus (LC) has established functions in both attention and respiration. Good attentional performance requires optimal levels of tonic LC activity, and must be matched to task consistently. LC neurons are chemosensitive, causing respiratory phrenic nerve firing to increase frequency with higher CO2 levels, and as CO2 level varies with the phase of respiration, tonic LC activity should exhibit fluctuations at respiratory frequency. Top-down modulation of tonic LC activity from brain areas involved in attentional regulation, intended to optimize LC firing to suit task requirements, may have respiratory consequences as well, as increases in LC activity influence phrenic nerve firing. We hypothesize that, due to the physiological and functional overlaps of attentional and respiratory functions of the LC, this small neuromodulatory nucleus is ideally situated to act as a mechanism of synchronization between respiratory and attentional systems, giving rise to a low-amplitude oscillation that enables attentional flexibility, but may also contribute to unintended destabilization of attention. Meditative and pranayama practices result in attentional, emotional, and physiological enhancements that may be partially due to the LC's pivotal role as the nexus in this coupled system. We present original findings of synchronization between respiration and LC activity (via fMRI and pupil dilation) and provide evidence of a relationship between respiratory phase modulation and attentional performance. We also present a mathematical dynamical systems model of respiratory-LC-attentional coupling, review candidate neurophysiological mechanisms of changes in coupling dynamics, and discuss implications for attentional theory, meditation, and pranayama, and possible therapeutic applications.
Collapse
Affiliation(s)
| | - Paul M Dockree
- Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | | | - Peter R Murphy
- Department of Neurophysiology and Pathophysiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Joshua H Balsters
- Department of Psychology, Royal Holloway University of London, Egham, United Kingdom
| | - Ian H Robertson
- Institute of Neuroscience and Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
46
|
Giustino TF, Maren S. Noradrenergic Modulation of Fear Conditioning and Extinction. Front Behav Neurosci 2018; 12:43. [PMID: 29593511 PMCID: PMC5859179 DOI: 10.3389/fnbeh.2018.00043] [Citation(s) in RCA: 126] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 02/26/2018] [Indexed: 12/12/2022] Open
Abstract
The locus coeruleus norepinephrine (LC-NE) system plays a broad role in learning and memory. Here we begin with an overview of the LC-NE system. We then consider how both direct and indirect manipulations of the LC-NE system affect cued and contextual aversive learning and memory. We propose that NE dynamically modulates Pavlovian conditioning and extinction, either promoting or impairing learning aversive processes under different levels of behavioral arousal. We suggest that under high levels of stress (e.g., during/soon after fear conditioning) the locus coeruleus (LC) promotes cued fear learning by enhancing amygdala function while simultaneously blunting prefrontal function. Under low levels of arousal, the LC promotes PFC function to promote downstream inhibition of the amygdala and foster the extinction of cued fear. Thus, LC-NE action on the medial prefrontal cortex (mPFC) might be described by an inverted-U function such that it can either enhance or hinder learning depending on arousal states. In addition, LC-NE seems to be particularly important for the acquisition, consolidation and extinction of contextual fear memories. This may be due to dense adrenoceptor expression in the hippocampus (HPC) which encodes contextual information, and the ability of NE to regulate long-term potentiation (LTP). Moreover, recent work reveals that the diversity of LC-NE functions in aversive learning and memory are mediated by functionally heterogeneous populations of LC neurons that are defined by their projection targets. Hence, LC-NE function in learning and memory is determined by projection-specific neuromodulation that accompanies various states of behavioral arousal.
Collapse
Affiliation(s)
- Thomas F Giustino
- Department of Psychological and Brain Sciences, Texas A&M Institute for Neuroscience, Texas A&M University, College Station, TX, United States
| | - Stephen Maren
- Department of Psychological and Brain Sciences, Texas A&M Institute for Neuroscience, Texas A&M University, College Station, TX, United States
| |
Collapse
|
47
|
Vazey EM, den Hartog CR, Moorman DE. Central Noradrenergic Interactions with Alcohol and Regulation of Alcohol-Related Behaviors. Handb Exp Pharmacol 2018; 248:239-260. [PMID: 29687164 DOI: 10.1007/164_2018_108] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Alcohol use disorder (AUD) results from disruption of a number of neural systems underlying motivation, emotion, and cognition. Patients with AUD exhibit not only elevated motivation for alcohol but heightened stress and anxiety, and disruptions in cognitive domains such as decision-making. One system at the intersection of these functions is the central norepinephrine (NE) system. This catecholaminergic neuromodulator, produced by several brainstem nuclei, plays profound roles in a wide range of behaviors and functions, including arousal, attention, and other aspects of cognition, motivation, emotional regulation, and control over basic physiological processes. It has been known for some time that NE has an impact on alcohol seeking and use, but the mechanisms of its influence are still being revealed. This chapter will discuss the influence of NE neuron activation and NE release at alcohol-relevant targets on behaviors and disruptions underlying alcohol motivation and AUD. Potential NE-based pharmacotherapies for AUD treatment will also be discussed. Given the basic properties of NE function, the strong relationship between NE and alcohol use, and the effectiveness of current NE-related treatments, the studies presented here indicate an encouraging direction for the development of precise and efficacious future therapies for AUD.
Collapse
Affiliation(s)
- Elena M Vazey
- Department of Biology & Neuroscience and Behavior Graduate Program, University of Massachusetts Amherst, Amherst, MA, USA.
| | - Carolina R den Hartog
- Department of Biology & Neuroscience and Behavior Graduate Program, University of Massachusetts Amherst, Amherst, MA, USA
| | - David E Moorman
- Department of Psychological and Brain Sciences & Neuroscience and Behavior Graduate Program, University of Massachusetts Amherst, Amherst, MA, USA
| |
Collapse
|
48
|
Korte SM, Prins J, Van den Bergh FS, Oosting RS, Dupree R, Korte-Bouws GA, Westphal KG, Olivier B, Denys DA, Garland A, Güntürkün O. The 5-HT1A/1B-receptor agonist eltoprazine increases both catecholamine release in the prefrontal cortex and dopamine release in the nucleus accumbens and decreases motivation for reward and “waiting” impulsivity, but increases “stopping” impulsivity. Eur J Pharmacol 2017; 794:257-269. [DOI: 10.1016/j.ejphar.2016.11.024] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 11/03/2016] [Accepted: 11/16/2016] [Indexed: 10/20/2022]
|
49
|
Kaushal R, Taylor BK, Jamal AB, Zhang L, Ma F, Donahue R, Westlund KN. GABA-A receptor activity in the noradrenergic locus coeruleus drives trigeminal neuropathic pain in the rat; contribution of NAα1 receptors in the medial prefrontal cortex. Neuroscience 2016; 334:148-159. [PMID: 27520081 DOI: 10.1016/j.neuroscience.2016.08.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 07/20/2016] [Accepted: 08/03/2016] [Indexed: 12/24/2022]
Abstract
Trigeminal neuropathic pain is described as constant excruciating facial pain. The study goal was to investigate the role of nucleus locus coeruleus (LC) in a model of chronic orofacial neuropathic pain (CCI-ION). The study examines LC's relationship to both the medullary dorsal horn receiving trigeminal nerve sensory innervation and the medial prefrontal cortex (mPFC). LC is a major source of CNS noradrenaline (NA) and a primary nucleus involved in pain modulation. Although descending inhibition of acute pain by LC is well established, contribution of the LC to facilitation of chronic neuropathic pain is also reported. In the present study, a rat orofacial pain model of trigeminal neuropathy was induced by chronic constrictive injury of the infraorbital nerve (CCI-ION). Orofacial neuropathic pain was indicated by development of whisker pad mechanical hypersensitivity. Hypersensitivity was alleviated by selective elimination of NA neurons, including LC (A6 cell group), with the neurotoxin anti-dopamine-β-hydroxylase saporin (anti-DβH-saporin) microinjected either intracerebroventricularly (i.c.v.) or into trigeminal spinal nucleus caudalis (spVc). The GABAA receptor antagonist, bicuculline, administered directly into LC (week 8) inhibited hypersensitivity. This indicates a valence shift in which increased GABAA signaling ongoing in LC after trigeminal nerve injury paradoxically produces excitatory facilitation of the chronic pain state. Microinjection of NAα1 receptor antagonist, benoxathian, into mPFC attenuated whisker pad hypersensitivity, while NAα2 receptor antagonist, idazoxan, was ineffective. Thus, GABAA-mediated activation of NA neurons during CCI-ION can facilitate hypersensitivity through NAα1 receptors in the mPFC. These data indicate LC is a chronic pain generator.
Collapse
Affiliation(s)
- R Kaushal
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY 40536-0298, United States
| | - B K Taylor
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY 40536-0298, United States
| | - A B Jamal
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY 40536-0298, United States
| | - L Zhang
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY 40536-0298, United States
| | - F Ma
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY 40536-0298, United States
| | - R Donahue
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY 40536-0298, United States
| | - K N Westlund
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY 40536-0298, United States.
| |
Collapse
|
50
|
Aston-Jones G, Waterhouse B. Locus coeruleus: From global projection system to adaptive regulation of behavior. Brain Res 2016; 1645:75-8. [PMID: 26969408 DOI: 10.1016/j.brainres.2016.03.001] [Citation(s) in RCA: 175] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 02/29/2016] [Accepted: 03/02/2016] [Indexed: 01/04/2023]
Abstract
The brainstem nucleus locus coeruleus (LC) is a major source of norepinephrine (NE) projections throughout the CNS. This important property was masked in very early studies by the inability to visualize endogenous monoamines. The development of monoamine histofluorescence methods by Swedish scientists led to a plethora of studies, including a paper published in Brain Research by Loizou in 1969. That paper was highly cited (making it a focal point for the 50th anniversary issue of this journal), and helped to spark a large and continuing set of investigations to further refine our understating of the LC-NE system and its contribution to brain function and behavior. This paper very briefly reviews the ensuing advances in anatomical, physiological and behavioral aspects of the LC-NE system. Although its projections are ubiquitously present throughout the CNS, recent studies find surprising specificity within the organizational and operational domains of LC neurons. These and other findings lead us to expect that future work will unmask additional features of the LC-NE system and its roles in normative and pathological brain and behavioral processes. This article is part of a Special Issue entitled SI:50th Anniversary Issue.
Collapse
Affiliation(s)
- G Aston-Jones
- Brain Health Institute, Rutgers University/Rutgers Biomedical and Health Sciences, Piscataway, NJ 08854, USA.
| | - B Waterhouse
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA.
| |
Collapse
|