51
|
Feinstein JS, Gould D, Khalsa SS. Amygdala-driven apnea and the chemoreceptive origin of anxiety. Biol Psychol 2022; 170:108305. [PMID: 35271957 DOI: 10.1016/j.biopsycho.2022.108305] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 02/09/2022] [Accepted: 03/03/2022] [Indexed: 12/13/2022]
Abstract
Although the amygdala plays an important part in the pathogenesis of anxiety and generation of exteroceptive fear, recent discoveries have challenged the directionality of this brain-behavior relationship with respect to interoceptive fear. Here we highlight several paradoxical findings including: (1) amygdala lesion patients who experience excessive fear and panic following inhalation of carbon dioxide (CO2), (2) clinically anxious patients who have significantly smaller (rather than larger) amygdalae and a pronounced hypersensitivity toward CO2, and (3) epilepsy patients who exhibit apnea immediately following stimulation of their amygdala yet have no awareness that their breathing has stopped. The above findings elucidate an entirely novel role for the amygdala in the induction of apnea and inhibition of CO2-induced fear. Such a role is plausible given the strong inhibitory connections linking the central nucleus of the amygdala with respiratory and chemoreceptive centers in the brainstem. Based on this anatomical arrangement, we propose a model of Apnea-induced Anxiety (AiA) which predicts that recurring episodes of apnea are being unconsciously elicited by amygdala activation, resulting in transient spikes in CO2 that provoke fear and anxiety, and lead to characteristic patterns of escape and avoidance behavior in patients spanning the spectrum of anxiety. If this new conception of AiA proves to be true, and activation of the amygdala can repeatedly trigger states of apnea outside of one's awareness, then it remains possible that the chronicity of anxiety disorders is being interoceptively driven by a chemoreceptive system struggling to maintain homeostasis in the midst of these breathless states.
Collapse
Affiliation(s)
- Justin S Feinstein
- Laureate Institute for Brain Research, Tulsa, Oklahoma, USA, 74136; University of Tulsa, Oxley College of Health Sciences, Tulsa, Oklahoma, USA, 74104; University of Iowa, Department of Neurology, Iowa City, Iowa, USA, 52242.
| | - Dylan Gould
- Laureate Institute for Brain Research, Tulsa, Oklahoma, USA, 74136
| | - Sahib S Khalsa
- Laureate Institute for Brain Research, Tulsa, Oklahoma, USA, 74136; University of Tulsa, Oxley College of Health Sciences, Tulsa, Oklahoma, USA, 74104
| |
Collapse
|
52
|
Subfornical organ interleukin 1 receptor: A novel regulator of spontaneous and conditioned fear associated behaviors in mice. Brain Behav Immun 2022; 101:304-317. [PMID: 35032573 PMCID: PMC9836229 DOI: 10.1016/j.bbi.2022.01.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 12/25/2021] [Accepted: 01/07/2022] [Indexed: 01/14/2023] Open
Abstract
Impaired threat responding and fear regulation is a hallmark of psychiatric conditions such as post-traumatic stress disorder (PTSD) and Panic Disorder (PD). Most studies have focused on external psychogenic threats to study fear, however, accumulating evidence suggests a primary role of homeostatic perturbations and interoception in regulating emotional behaviors. Heightened reactivity to interoceptive threat carbon dioxide (CO2) inhalation associates with increased risk for developing PD and PTSD, however, contributory mechanisms and molecular targets are not well understood. Previous studies from our group suggested a potential role of interleukin 1 receptor (IL-1R1) signaling within BBB-devoid sensory circumventricular organ, the subfornical organ (SFO) in CO2-evoked fear. However, the necessity of SFO-IL-1R1 in regulating CO2-associated spontaneous fear as well as, long-term fear potentiation relevant to PD/PTSD has not been investigated. The current study tested male mice with SFO-targeted microinfusion of the IL-1R1 antagonist (IL-1RA) or vehicle in a recently developed CO2-startle-fear conditioning-extinction paradigm. Consistent with our hypothesis, SFO IL-1RA treatment elicited significant attenuation of freezing and increased rearing during CO2 inhalation suggesting SFO-IL1R1 regulation of spontaneous fear to CO2. Intriguingly, SFO IL-1RA treatment normalized CO2-associated potentiation of conditioned fear and impaired extinction a week later suggesting modulation of long-term fear by SFO-IL-1R1 signaling. Post behavior FosB mapping revealed recruitment of prefrontal cortex-amygdala-periaqueductal gray (PAG) areas in SFO-IL-1RA mediated effects. Additionally, we localized cellular IL-1R1 expression within the SFO to blood vessel endothelial cells and observed CO2-induced alterations in IL-1β/IL-1R1 expression in peripheral mononuclear cells and SFO. Lastly, CO2-evoked microglial activation was attenuated in SFO-IL-1RA treated mice. These observations suggest a peripheral monocyte-endothelial-microglia interplay in SFO-IL-1R1 modulation of CO2-associated spontaneous fear and delayed fear memory. Collectively, our data highlight a novel, "bottom-up" neuroimmune mechanism that integrates interoceptive and exteroceptive threat processing of relevance to fear-related pathologies.
Collapse
|
53
|
Koffman EE, Kruse CM, Singh K, Naghavi FS, Curtis MA, Egbo J, Houdi M, Lin B, Lu H, Debiec J, Du J. Acid-sensing ion channel 1a regulates the specificity of reconsolidation of conditioned threat responses. JCI Insight 2022; 7:155341. [PMID: 35025766 PMCID: PMC8876458 DOI: 10.1172/jci.insight.155341] [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: 09/27/2021] [Accepted: 01/12/2022] [Indexed: 11/26/2022] Open
Abstract
Recent research on altering threat memory has focused on a reconsolidation window. During reconsolidation, threat memories are retrieved and become labile. Reconsolidation of distinct threat memories is synapse dependent, whereas the underlying regulatory mechanism of the specificity of reconsolidation is poorly understood. We designed a unique behavioral paradigm in which a distinct threat memory can be retrieved through the associated conditioned stimulus. In addition, we proposed a regulatory mechanism by which the activation of acid-sensing ion channels (ASICs) strengthens the distinct memory trace associated with the memory reconsolidation to determine its specificity. The activation of ASICs by CO2 inhalation, when paired with memory retrieval, triggers the reactivation of the distinct memory trace, resulting in greater memory lability. ASICs potentiate the memory trace by altering the amygdala-dependent synaptic transmission and plasticity at selectively targeted synapses. Our results suggest that inhaling CO2 during the retrieval event increases the lability of a threat memory through a synapse-specific reconsolidation process.
Collapse
Affiliation(s)
- Erin E Koffman
- Department of Biological Sciences, The University of Toledo, Toledo, United States of America
| | - Charles M Kruse
- Department of Biological Sciences, The University of Toledo, Toledo, United States of America
| | - Kritika Singh
- Department of Biological Sciences, The University of Toledo, Toledo, United States of America
| | - Farzaneh Sadat Naghavi
- Department of Biological Sciences, The University of Toledo, Toledo, United States of America
| | - Melissa A Curtis
- Department of Biological Sciences, The University of Toledo, Toledo, United States of America
| | - Jennifer Egbo
- Department of Biological Sciences, The University of Toledo, Toledo, United States of America
| | - Mark Houdi
- Department of Biological Sciences, The University of Toledo, Toledo, United States of America
| | - Boren Lin
- Department of Biological Sciences, The University of Toledo, Toledo, United States of America
| | - Hui Lu
- Department of Pharmacology and Physiology, George Washington University, Washington, DC, United States of America
| | - Jacek Debiec
- Department of Psychiatry, The University of Michigan Medical School, Ann Arbor, United States of America
| | - Jianyang Du
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, United States of America
| |
Collapse
|
54
|
Guyenet PG, Stornetta RL. Rostral ventrolateral medulla, retropontine region and autonomic regulations. Auton Neurosci 2021; 237:102922. [PMID: 34814098 DOI: 10.1016/j.autneu.2021.102922] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 11/08/2021] [Accepted: 11/15/2021] [Indexed: 12/17/2022]
Abstract
The rostral half of the ventrolateral medulla (RVLM) and adjacent ventrolateral retropontine region (henceforth RVLMRP) have been divided into various sectors by neuroscientists interested in breathing or autonomic regulations. The RVLMRP regulates respiration, glycemia, vigilance and inflammation, in addition to blood pressure. It contains interoceptors that respond to acidification, hypoxia and intracranial pressure and its rostral end contains the retrotrapezoid nucleus (RTN) which is the main central respiratory chemoreceptor. Acid detection by the RTN is an intrinsic property of the principal neurons that is enhanced by paracrine influences from surrounding astrocytes and CO2-dependent vascular constriction. RTN mediates the hypercapnic ventilatory response via complex projections to the respiratory pattern generator (CPG). The RVLM contributes to autonomic response patterns via differential recruitment of several subtypes of adrenergic (C1) and non-adrenergic neurons that directly innervate sympathetic and parasympathetic preganglionic neurons. The RVLM also innervates many brainstem and hypothalamic nuclei that contribute, albeit less directly, to autonomic responses. All lower brainstem noradrenergic clusters including the locus coeruleus are among these targets. Sympathetic tone to the circulatory system is regulated by subsets of presympathetic RVLM neurons whose activity is continuously restrained by the baroreceptors and modulated by the respiratory CPG. The inhibitory input from baroreceptors and the excitatory input from the respiratory CPG originate from neurons located in or close to the rhythm generating region of the respiratory CPG (preBötzinger complex).
Collapse
Affiliation(s)
- Patrice G Guyenet
- University of Virginia School of Medicine, Department of Pharmacology, 1340 Jefferson Park Avenue, Charlottesville, VA 22908-0735, USA.
| | - Ruth L Stornetta
- University of Virginia School of Medicine, Department of Pharmacology, 1340 Jefferson Park Avenue, Charlottesville, VA 22908-0735, USA.
| |
Collapse
|
55
|
John S, K G G, Krishna AP, Mishra R. Neurotherapeutic implications of sense and respond strategies generated by astrocytes and astrocytic tumours to combat pH mechanical stress. Neuropathol Appl Neurobiol 2021; 48:e12774. [PMID: 34811795 PMCID: PMC9300154 DOI: 10.1111/nan.12774] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 09/24/2021] [Accepted: 11/14/2021] [Indexed: 02/04/2023]
Abstract
Aims Astrocytes adapt to acute acid stress. Intriguingly, cancer cells with astrocytic differentiation thrive even better in an acidic microenvironment. How changes in extracellular pH (pHe) are sensed and measured by the cell surface assemblies that first intercept the acid stress, and how this information is relayed downstream for an appropriate survival response remains largely uncharacterized. Methods In vitro cell‐based studies were combined with an in vivo animal model to delineate the machinery involved in pH microenvironment sensing and generation of mechanoadaptive responses in normal and neoplastic astrocytes. The data was further validated on patient samples from acidosis driven ischaemia and astrocytic tumour tissues. Results We demonstrate that low pHe is perceived and interpreted by cells as mechanical stress. GM3 acts as a lipid‐based pH sensor, and in low pHe, its highly protonated state generates plasma membrane deformation stress which activates the IRE1‐sXBP1‐SREBP2‐ACSS2 response axis for cholesterol biosynthesis and surface trafficking. Enhanced surface cholesterol provides mechanical tenacity and prevents acid‐mediated membrane hydrolysis, which would otherwise result in cell leakage and death. Conclusions In summary, activating these lipids or the associated downstream machinery in acidosis‐related neurodegeneration may prevent disease progression, while specifically suppressing this key mechanical ‘sense‐respond’ axis should effectively target astrocytic tumour growth.
Collapse
Affiliation(s)
- Sebastian John
- Brain and Cerebrovascular Mechanobiology Research, Laboratory of Translational Mechanobiology, Department of Neurobiology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India.,Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| | - Gayathri K G
- Brain and Cerebrovascular Mechanobiology Research, Laboratory of Translational Mechanobiology, Department of Neurobiology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India.,Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| | - Aswani P Krishna
- Brain and Cerebrovascular Mechanobiology Research, Laboratory of Translational Mechanobiology, Department of Neurobiology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - Rashmi Mishra
- Brain and Cerebrovascular Mechanobiology Research, Laboratory of Translational Mechanobiology, Department of Neurobiology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India.,Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| |
Collapse
|
56
|
Taugher RJ, Wunsch AM, Wang GZ, Chan AC, Dlouhy BJ, Wemmie JA. Post-acquisition CO 2 Inhalation Enhances Fear Memory and Depends on ASIC1A. Front Behav Neurosci 2021; 15:767426. [PMID: 34776896 PMCID: PMC8585996 DOI: 10.3389/fnbeh.2021.767426] [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: 09/01/2021] [Accepted: 10/04/2021] [Indexed: 11/25/2022] Open
Abstract
A growing body of evidence suggests that memories of fearful events may be altered after initial acquisition or learning. Although much of this work has been done in rodents using Pavlovian fear conditioning, it may have important implications for fear memories in humans such as in post-traumatic stress disorder (PTSD). A recent study suggested that cued fear memories, made labile by memory retrieval, were made additionally labile and thus more vulnerable to subsequent modification when mice inhaled 10% carbon dioxide (CO2) during retrieval. In light of this finding, we hypothesized that 10% CO2 inhalation soon after fear acquisition might affect memory recall 24 h later. We found that both cue and context fear memory were increased by CO2 exposure after fear acquisition. The effect of CO2 was time-dependent, as CO2 inhalation administered 1 or 4 h after cued fear acquisition increased fear memory, whereas CO2 inhalation 4 h before or 24 h after cued fear acquisition did not increase fear memory. The ability of CO2 exposure following acquisition to enhance fear memory was not a general consequence of stress, as restraining mice after acquisition did not alter cued fear memory. The memory-enhancing action of CO2 may be relatively specific to fear conditioning as novel object recognition was impaired by post-training CO2 inhalation. To explore the molecular underpinnings of these effects, we tested if they depended on the acid-sensing ion channel-1a (ASIC1A), a proton-gated cation channel that mediates other effects of CO2, likely via its ability to sense acidosis induced during CO2 inhalation. We found that CO2 inhalation did not alter cued or context fear memory in Asic1a–/– mice, suggesting that this phenomenon critically depends on ASIC1A. These results suggest that brain acidosis around the time of a traumatic event may enhance memory of the trauma, and may thus constitute an important risk factor for developing PTSD. Moreover, preventing peritraumatic acidosis might reduce risk of PTSD.
Collapse
Affiliation(s)
- Rebecca J Taugher
- Department of Psychiatry, University of Iowa, Iowa City, IA, United States.,Department of Veterans Affairs Medical Center, Iowa City, IA, United States
| | - Amanda M Wunsch
- Department of Psychiatry, University of Iowa, Iowa City, IA, United States.,Department of Veterans Affairs Medical Center, Iowa City, IA, United States
| | - Grace Z Wang
- Department of Psychiatry, University of Iowa, Iowa City, IA, United States.,Department of Veterans Affairs Medical Center, Iowa City, IA, United States
| | - Aubrey C Chan
- Department of Psychiatry, University of Iowa, Iowa City, IA, United States.,Department of Veterans Affairs Medical Center, Iowa City, IA, United States.,Pappajohn Biomedical Institute, University of Iowa, Iowa City, IA, United States.,Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, United States.,Department of Internal Medicine, University of Iowa, Iowa City, IA, United States
| | - Brian J Dlouhy
- Pappajohn Biomedical Institute, University of Iowa, Iowa City, IA, United States.,Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, United States.,Department of Neurosurgery, University of Iowa, Iowa City, IA, United States
| | - John A Wemmie
- Department of Psychiatry, University of Iowa, Iowa City, IA, United States.,Department of Veterans Affairs Medical Center, Iowa City, IA, United States.,Pappajohn Biomedical Institute, University of Iowa, Iowa City, IA, United States.,Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, United States.,Department of Neurosurgery, University of Iowa, Iowa City, IA, United States.,Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, United States.,Roy J. Carver Chair of Psychiatry and Neuroscience, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
57
|
Sfera A, Osorio C, Rahman L, Zapata-Martín del Campo CM, Maldonado JC, Jafri N, Cummings MA, Maurer S, Kozlakidis Z. PTSD as an Endothelial Disease: Insights From COVID-19. Front Cell Neurosci 2021; 15:770387. [PMID: 34776871 PMCID: PMC8586713 DOI: 10.3389/fncel.2021.770387] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 10/11/2021] [Indexed: 12/15/2022] Open
Abstract
SARS-CoV-2 virus, the etiologic agent of COVID-19, has affected almost every aspect of human life, precipitating stress-related pathology in vulnerable individuals. As the prevalence rate of posttraumatic stress disorder in pandemic survivors exceeds that of the general and special populations, the virus may predispose to this disorder by directly interfering with the stress-processing pathways. The SARS-CoV-2 interactome has identified several antigens that may disrupt the blood-brain-barrier by inducing premature senescence in many cell types, including the cerebral endothelial cells. This enables the stress molecules, including angiotensin II, endothelin-1 and plasminogen activator inhibitor 1, to aberrantly activate the amygdala, hippocampus, and medial prefrontal cortex, increasing the vulnerability to stress related disorders. This is supported by observing the beneficial effects of angiotensin receptor blockers and angiotensin converting enzyme inhibitors in both posttraumatic stress disorder and SARS-CoV-2 critical illness. In this narrative review, we take a closer look at the virus-host dialog and its impact on the renin-angiotensin system, mitochondrial fitness, and brain-derived neurotrophic factor. We discuss the role of furin cleaving site, the fibrinolytic system, and Sigma-1 receptor in the pathogenesis of psychological trauma. In other words, learning from the virus, clarify the molecular underpinnings of stress related disorders, and design better therapies for these conditions. In this context, we emphasize new potential treatments, including furin and bromodomains inhibitors.
Collapse
Affiliation(s)
- Adonis Sfera
- Department of Psychiatry, Loma Linda University, Loma Linda, CA, United States
- Patton State Hospital, San Bernardino, CA, United States
| | - Carolina Osorio
- Department of Psychiatry, Loma Linda University, Loma Linda, CA, United States
| | - Leah Rahman
- Patton State Hospital, San Bernardino, CA, United States
| | | | - Jose Campo Maldonado
- Department of Medicine, The University of Texas Rio Grande Valley, Edinburg, TX, United States
| | - Nyla Jafri
- Patton State Hospital, San Bernardino, CA, United States
| | | | - Steve Maurer
- Patton State Hospital, San Bernardino, CA, United States
| | - Zisis Kozlakidis
- International Agency For Research On Cancer (IARC), Lyon, France
| |
Collapse
|
58
|
Sheikh ZP, Wulf M, Friis S, Althaus M, Lynagh T, Pless SA. The M1 and pre-M1 segments contribute differently to ion selectivity in ASICs and ENaCs. J Gen Physiol 2021; 153:212604. [PMID: 34436511 PMCID: PMC8404453 DOI: 10.1085/jgp.202112899] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 07/26/2021] [Indexed: 11/20/2022] Open
Abstract
The ability to discriminate between different ionic species, termed ion selectivity, is a key feature of ion channels and forms the basis for their physiological function. Members of the degenerin/epithelial sodium channel (DEG/ENaC) superfamily of trimeric ion channels are typically sodium selective, but to a surprisingly variable degree. While acid-sensing ion channels (ASICs) are weakly sodium selective (sodium:potassium ratio ∼10:1), ENaCs show a remarkably high preference for sodium over potassium (>500:1). This discrepancy may be expected to originate from differences in the pore-lining second transmembrane segment (M2). However, these show a relatively high degree of sequence conservation between ASICs and ENaCs, and previous functional and structural studies could not unequivocally establish that differences in M2 alone can account for the disparate degrees of ion selectivity. By contrast, surprisingly little is known about the contributions of the first transmembrane segment (M1) and the preceding pre-M1 region. In this study, we used conventional and noncanonical amino acid-based mutagenesis in combination with a variety of electrophysiological approaches to show that the pre-M1 and M1 regions of mASIC1a channels are major determinants of ion selectivity. Mutational investigations of the corresponding regions in hENaC show that these regions contribute less to ion selectivity, despite affecting ion conductance. In conclusion, our work suggests that the remarkably different degrees of sodium selectivity in ASICs and ENaCs are achieved through different mechanisms. These results further highlight how M1 and pre-M1 are likely to differentially affect pore structure in these related channels.
Collapse
Affiliation(s)
- Zeshan P Sheikh
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Matthias Wulf
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | | | - Mike Althaus
- School of Natural and Environmental Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Timothy Lynagh
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Stephan A Pless
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
59
|
Vinckier F, Betka S, Nion N, Serresse L, Similowski T. Harnessing the power of anticipation to manage respiratory-related brain suffering and ensuing dyspnoea: insights from the neurobiology of the respiratory nocebo effect. Eur Respir J 2021; 58:58/3/2101876. [PMID: 34556533 DOI: 10.1183/13993003.01876-2021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Accepted: 08/09/2021] [Indexed: 12/29/2022]
Affiliation(s)
- Fabien Vinckier
- Université de Paris, Paris, France.,Dept of Psychiatry, Service Hospitalo-Universitaire, GHU Paris Psychiatry and Neurosciences, Paris, France
| | - Sophie Betka
- Laboratory of Cognitive Neuroscience, Brain Mind Institute and Center for Neuroprosthetics, Faculty of Life Sciences, Swiss Federal Institute of Technology (EPFL), Geneva, Switzerland
| | - Nathalie Nion
- Neurophysiologie Respiratoire Expérimentale et Clinique, INSERM, UMRS1158, Sorbonne Université, Paris, France.,Département R3S (Respiration, Réanimation, Réhabilitation respiratoire, Sommeil), AP-HP, Groupe Hospitalier Universitaire APHP-Sorbonne Université, site Pitié-Salpêtrière, Paris, France
| | - Laure Serresse
- Neurophysiologie Respiratoire Expérimentale et Clinique, INSERM, UMRS1158, Sorbonne Université, Paris, France.,Equipe mobile de soins palliatifs, AP-HP, Groupe Hospitalier Universitaire APHP-Sorbonne Université, site Pitié-Salpêtrière, Paris, France
| | - Thomas Similowski
- Neurophysiologie Respiratoire Expérimentale et Clinique, INSERM, UMRS1158, Sorbonne Université, Paris, France .,Département R3S (Respiration, Réanimation, Réhabilitation respiratoire, Sommeil), AP-HP, Groupe Hospitalier Universitaire APHP-Sorbonne Université, site Pitié-Salpêtrière, Paris, France
| |
Collapse
|
60
|
Leisle L, Margreiter M, Ortega-Ramírez A, Cleuvers E, Bachmann M, Rossetti G, Gründer S. Dynorphin Neuropeptides Decrease Apparent Proton Affinity of ASIC1a by Occluding the Acidic Pocket. J Med Chem 2021; 64:13299-13311. [PMID: 34461722 DOI: 10.1021/acs.jmedchem.1c00447] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Prolonged acidosis, as it occurs during ischemic stroke, induces neuronal death via acid-sensing ion channel 1a (ASIC1a). Concomitantly, it desensitizes ASIC1a, highlighting the pathophysiological significance of modulators of ASIC1a acid sensitivity. One such modulator is the opioid neuropeptide big dynorphin (Big Dyn) which binds to ASIC1a and enhances its activity during prolonged acidosis. The molecular determinants and dynamics of this interaction remain unclear, however. Here, we present a molecular interaction model showing a dynorphin peptide inserting deep into the acidic pocket of ASIC1a. We confirmed experimentally that the interaction is predominantly driven by electrostatic forces, and using noncanonical amino acids as photo-cross-linkers, we identified 16 residues in ASIC1a contributing to Big Dyn binding. Covalently tethering Big Dyn to its ASIC1a binding site dramatically decreased the proton sensitivity of channel activation, suggesting that Big Dyn stabilizes a resting conformation of ASIC1a and dissociates from its binding site during channel opening.
Collapse
Affiliation(s)
- Lilia Leisle
- Institute of Physiology, RWTH Aachen University, 52074 Aachen, Germany
| | - Michael Margreiter
- Computational Biomedicine-Institute for Advanced Simulation/Institute of Neuroscience and Medicine, Forschungszentrum Jülich, 52425 Jülich, Germany.,Institute of Organic Chemistry, RWTH Aachen University, Landoltweg 1, 52074 Aachen, Germany
| | | | - Elinor Cleuvers
- Institute of Physiology, RWTH Aachen University, 52074 Aachen, Germany
| | - Michèle Bachmann
- Institute of Physiology, RWTH Aachen University, 52074 Aachen, Germany
| | - Giulia Rossetti
- Computational Biomedicine-Institute for Advanced Simulation/Institute of Neuroscience and Medicine, Forschungszentrum Jülich, 52425 Jülich, Germany.,Jülich Supercomputing Center (JSC), Forschungszentrum Jülich, 52425 Jülich, Germany.,Department of Neurology, RWTH Aachen University, 52074 Aachen, Germany
| | - Stefan Gründer
- Institute of Physiology, RWTH Aachen University, 52074 Aachen, Germany
| |
Collapse
|
61
|
Foster VS, Rash LD, King GF, Rank MM. Acid-Sensing Ion Channels: Expression and Function in Resident and Infiltrating Immune Cells in the Central Nervous System. Front Cell Neurosci 2021; 15:738043. [PMID: 34602982 PMCID: PMC8484650 DOI: 10.3389/fncel.2021.738043] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 08/30/2021] [Indexed: 11/15/2022] Open
Abstract
Peripheral and central immune cells are critical for fighting disease, but they can also play a pivotal role in the onset and/or progression of a variety of neurological conditions that affect the central nervous system (CNS). Tissue acidosis is often present in CNS pathologies such as multiple sclerosis, epileptic seizures, and depression, and local pH is also reduced during periods of ischemia following stroke, traumatic brain injury, and spinal cord injury. These pathological increases in extracellular acidity can activate a class of proton-gated channels known as acid-sensing ion channels (ASICs). ASICs have been primarily studied due to their ubiquitous expression throughout the nervous system, but it is less well recognized that they are also found in various types of immune cells. In this review, we explore what is currently known about the expression of ASICs in both peripheral and CNS-resident immune cells, and how channel activation during pathological tissue acidosis may lead to altered immune cell function that in turn modulates inflammatory pathology in the CNS. We identify gaps in the literature where ASICs and immune cell function has not been characterized, such as neurotrauma. Knowledge of the contribution of ASICs to immune cell function in neuropathology will be critical for determining whether the therapeutic benefits of ASIC inhibition might be due in part to an effect on immune cells.
Collapse
Affiliation(s)
- Victoria S. Foster
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia
| | - Lachlan D. Rash
- School of Biomedical Sciences, The University of Queensland, St Lucia, QLD, Australia
| | - Glenn F. King
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, St Lucia, QLD, Australia
| | - Michelle M. Rank
- Anatomy and Physiology, Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
62
|
Tenorio-Lopes L, Kinkead R. Sex-Specific Effects of Stress on Respiratory Control: Plasticity, Adaptation, and Dysfunction. Compr Physiol 2021; 11:2097-2134. [PMID: 34107062 DOI: 10.1002/cphy.c200022] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
As our understanding of respiratory control evolves, we appreciate how the basic neurobiological principles of plasticity discovered in other systems shape the development and function of the respiratory control system. While breathing is a robust homeostatic function, there is growing evidence that stress disrupts respiratory control in ways that predispose to disease. Neonatal stress (in the form of maternal separation) affects "classical" respiratory control structures such as the peripheral O2 sensors (carotid bodies) and the medulla (e.g., nucleus of the solitary tract). Furthermore, early life stress disrupts the paraventricular nucleus of the hypothalamus (PVH), a structure that has emerged as a primary determinant of the intensity of the ventilatory response to hypoxia. Although underestimated, the PVH's influence on respiratory function is a logical extension of the hypothalamic control of metabolic demand and supply. In this article, we review the functional and anatomical links between the stress neuroendocrine axis and the medullary network regulating breathing. We then present the persistent and sex-specific effects of neonatal stress on respiratory control in adult rats. The similarities between the respiratory phenotype of stressed rats and clinical manifestations of respiratory control disorders such as sleep-disordered breathing and panic attacks are remarkable. These observations are in line with the scientific consensus that the origins of adult disease are often found among developmental and biological disruptions occurring during early life. These observations bring a different perspective on the structural hierarchy of respiratory homeostasis and point to new directions in our understanding of the etiology of respiratory control disorders. © 2021 American Physiological Society. Compr Physiol 11:1-38, 2021.
Collapse
Affiliation(s)
- Luana Tenorio-Lopes
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, The University of Calgary, Calgary, Alberta, Canada
| | - Richard Kinkead
- Département de Pédiatrie, Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Quebec City, Quebec, Canada
| |
Collapse
|
63
|
Citric Acid in Drug Formulations Causes Pain by Potentiating Acid-Sensing Ion Channel 1. J Neurosci 2021; 41:4596-4606. [PMID: 33888605 PMCID: PMC8260239 DOI: 10.1523/jneurosci.2087-20.2021] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 12/08/2020] [Accepted: 04/10/2021] [Indexed: 11/21/2022] Open
Abstract
Pain at the injection site is a common complaint of patients receiving therapeutic formulations containing citric acid. Despite the widely acknowledged role of acid-sensing ion channels (ASICs) in acid-related perception, the specific ASIC subtype mediating pain caused by subcutaneous acid injection and the mechanism by which citrate affects this process are less clear. Here, male mice subjected to intraplantar acid injection responded by executing a withdrawal reflex, and this response was abolished by ASIC1 but not ASIC2 knockout. Although intraplantar injection of neutral citrate solution did not produce this response, intraplantar injection of acidic citrate solution produced a withdrawal reflex greater than that produced by acidity alone. Consistent with the behavioral data, neutral citrate failed to produce an electrophysiological response in HEK293 cells, which express ASIC1, but acidic citrate produced a whole-cell inward current greater than that produced by acidity alone. Saturating the intracellular solution with citrate had no effect on the potentiating effect of extracellular citrate, suggesting that citrate acted extracellularly to potentiate ASIC1. Moreover, exposure to citrate immediately before acid stimulation failed to potentiate ASIC1 currents, which ruled out the involvement of a metabotropic receptor gated by a citrate metabolite. Finally, removal of calcium ions from the extracellular solution mimicked the potentiating effect of citrate and prevented citrate from further potentiating ASIC1. Our data demonstrate that ASIC1 is necessary for the nociceptive response caused by subcutaneous acid infusion and that neutral citrate, despite not inducing ASIC1 currents or nociceptive behavior on its own, potentiates acid nociception by removing the inhibitory effect of extracellular calcium ions on ASIC1. SIGNIFICANCE STATEMENT Citric acid is a common ingredient used in pharmaceutical formulations. Despite the widespread clinical use of these formulations, it remains unclear how citric acid causes pain when injected into patients. We identified ASIC1 as the key receptor used to detect injection-site pain caused by acid, and we showed that neutral citrate does not stimulate ASIC1; instead, citrate substantially potentiates ASIC1 activation when injected simultaneously with acid. In addition, we demonstrated that citrate potentiates ASIC1 by removing the inhibitory action of calcium on the extracellular side of the receptor. Given that injection-site pain is the primary complaint of patients receiving citrate-containing medical products, our data provide mechanistic insight into a common medical complaint and suggest a means of avoiding injection pain.
Collapse
|
64
|
Phelan DE, Mota C, Lai C, Kierans SJ, Cummins EP. Carbon dioxide-dependent signal transduction in mammalian systems. Interface Focus 2021; 11:20200033. [PMID: 33633832 PMCID: PMC7898142 DOI: 10.1098/rsfs.2020.0033] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/09/2020] [Indexed: 12/15/2022] Open
Abstract
Carbon dioxide (CO2) is a fundamental physiological gas known to profoundly influence the behaviour and health of millions of species within the plant and animal kingdoms in particular. A recent Royal Society meeting on the topic of 'Carbon dioxide detection in biological systems' was extremely revealing in terms of the multitude of roles that different levels of CO2 play in influencing plants and animals alike. While outstanding research has been performed by leading researchers in the area of plant biology, neuronal sensing, cell signalling, gas transport, inflammation, lung function and clinical medicine, there is still much to be learned about CO2-dependent sensing and signalling. Notably, while several key signal transduction pathways and nodes of activity have been identified in plants and animals respectively, the precise wiring and sensitivity of these pathways to CO2 remains to be fully elucidated. In this article, we will give an overview of the literature relating to CO2-dependent signal transduction in mammalian systems. We will highlight the main signal transduction hubs through which CO2-dependent signalling is elicited with a view to better understanding the complex physiological response to CO2 in mammalian systems. The main topics of discussion in this article relate to how changes in CO2 influence cellular function through modulation of signal transduction networks influenced by pH, mitochondrial function, adenylate cyclase, calcium, transcriptional regulators, the adenosine monophosphate-activated protein kinase pathway and direct CO2-dependent protein modifications. While each of these topics will be discussed independently, there is evidence of significant cross-talk between these signal transduction pathways as they respond to changes in CO2. In considering these core hubs of CO2-dependent signal transduction, we hope to delineate common elements and identify areas in which future research could be best directed.
Collapse
Affiliation(s)
- D. E. Phelan
- School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - C. Mota
- School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - C. Lai
- School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - S. J. Kierans
- School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - E. P. Cummins
- School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| |
Collapse
|
65
|
Abbott SBG, Souza GMPR. Chemoreceptor mechanisms regulating CO 2 -induced arousal from sleep. J Physiol 2021; 599:2559-2571. [PMID: 33759184 DOI: 10.1113/jp281305] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 03/16/2021] [Indexed: 12/24/2022] Open
Abstract
Arousal from sleep in response to CO2 is a life-preserving reflex that enhances ventilatory drive and facilitates behavioural adaptations to restore eupnoeic breathing. Recurrent activation of the CO2 -arousal reflex is associated with sleep disruption in obstructive sleep apnoea. In this review we examine the role of chemoreceptors in the carotid bodies, the retrotrapezoid nucleus and serotonergic neurons in the dorsal raphe in the CO2 -arousal reflex. We also provide an overview of the supra-medullary structures that mediate CO2 -induced arousal. We propose a framework for the CO2 -arousal reflex in which the activity of the chemoreceptors converges in the parabrachial nucleus to trigger cortical arousal.
Collapse
Affiliation(s)
- Stephen B G Abbott
- Department of Pharmacology, University of Virginia, Charlottesville, VA, 29903, USA
| | - George M P R Souza
- Department of Pharmacology, University of Virginia, Charlottesville, VA, 29903, USA
| |
Collapse
|
66
|
Locus Coeruleus Acid-Sensing Ion Channels Modulate Sleep-Wakefulness and State Transition from NREM to REM Sleep in the Rat. Neurosci Bull 2021; 37:684-700. [PMID: 33638800 DOI: 10.1007/s12264-020-00625-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 06/18/2020] [Indexed: 12/11/2022] Open
Abstract
The locus coeruleus (LC) is one of the essential chemoregulatory and sleep-wake (S-W) modulating centers in the brain. LC neurons remain highly active during wakefulness, and some implicitly become silent during rapid eye movement (REM) sleep. LC neurons are also involved in CO2-dependent modulation of the respiratory drive. Acid-sensing ion channels (ASICs) are highly expressed in some brainstem chemosensory breathing regulatory areas, but their localization and functions in the LC remain unknown. Mild hypercapnia increases the amount of non-REM (NREM) sleep and the number of REM sleep episodes, but whether ASICs in the LC modulate S-W is unclear. Here, we investigated the presence of ASICs in the LC and their role in S-W modulation and the state transition from NREM to REM sleep. Male Wistar rats were surgically prepared for chronic polysomnographic recordings and drug microinjections into the LC. The presence of ASIC-2 and ASIC-3 in the LC was immunohistochemically characterized. Microinjections of amiloride (an ASIC blocker) and APETx2 (a blocker of ASIC-2 and -3) into the LC significantly decreased wakefulness and REM sleep, but significantly increased NREM sleep. Mild hypercapnia increased the amount of NREM and the number of REM episodes. However, APETx2 microinjection inhibited this increase in REM frequency. These results suggest that the ASICs of LC neurons modulate S-W, indicating that ASICs could play an important role in vigilance-state transition. A mild increase in CO2 level during NREM sleep sensed by ASICs could be one of the determinants of state transition from NREM to REM sleep.
Collapse
|
67
|
Song XL, Liu DS, Qiang M, Li Q, Liu MG, Li WG, Qi X, Xu NJ, Yang G, Zhu MX, Xu TL. Postsynaptic Targeting and Mobility of Membrane Surface-Localized hASIC1a. Neurosci Bull 2021; 37:145-165. [PMID: 32996060 PMCID: PMC7870742 DOI: 10.1007/s12264-020-00581-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 05/14/2020] [Indexed: 01/19/2023] Open
Abstract
Acid-sensing ion channels (ASICs), the main H+ receptors in the central nervous system, sense extracellular pH fluctuations and mediate cation influx. ASIC1a, the major subunit responsible for acid-activated current, is widely expressed in brain neurons, where it plays pivotal roles in diverse functions including synaptic transmission and plasticity. However, the underlying molecular mechanisms for these functions remain mysterious. Using extracellular epitope tagging and a novel antibody recognizing the hASIC1a ectodomain, we examined the membrane targeting and dynamic trafficking of hASIC1a in cultured cortical neurons. Surface hASIC1a was distributed throughout somata and dendrites, clustered in spine heads, and co-localized with postsynaptic markers. By extracellular pHluorin tagging and fluorescence recovery after photobleaching, we detected movement of hASIC1a in synaptic spine heads. Single-particle tracking along with use of the anti-hASIC1a ectodomain antibody revealed long-distance migration and local movement of surface hASIC1a puncta on dendrites. Importantly, enhancing synaptic activity with brain-derived neurotrophic factor accelerated the trafficking and lateral mobility of hASIC1a. With this newly-developed toolbox, our data demonstrate the synaptic location and high dynamics of functionally-relevant hASIC1a on the surface of excitatory synapses, supporting its involvement in synaptic functions.
Collapse
Affiliation(s)
- Xing-Lei Song
- Center for Brain Science of Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Di-Shi Liu
- Center for Brain Science of Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Min Qiang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China
| | - Qian Li
- Center for Brain Science of Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai, 201210, China
| | - Ming-Gang Liu
- Center for Brain Science of Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai, 201210, China
| | - Wei-Guang Li
- Center for Brain Science of Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai, 201210, China
| | - Xin Qi
- Center for Brain Science of Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Nan-Jie Xu
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai, 201210, China
| | - Guang Yang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China
| | - Michael Xi Zhu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.
| | - Tian-Le Xu
- Center for Brain Science of Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai, 201210, China.
| |
Collapse
|
68
|
Sandford DG. Preparatory and Anticipatory Grief, Anxiety and Depression in Life-Limiting Lung Disease. Respir Med 2021. [DOI: 10.1007/978-3-030-81788-6_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
69
|
Leibold NK, van den Hove DLA, Weidner MT, Buchanan GF, Steinbusch HWM, Lesch KP, Schruers KRJ. Effect of serotonin transporter genotype on carbon dioxide-induced fear-related behavior in mice. J Psychopharmacol 2020; 34:1408-1417. [PMID: 33103571 PMCID: PMC7708670 DOI: 10.1177/0269881120959611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Inhaling 35% carbon dioxide induces an emotional and symptomatic state in humans closely resembling naturally occurring panic attacks, the core symptom of panic disorder. Previous research has suggested a role of the serotonin system in the individual sensitivity to carbon dioxide. In line with this, we previously showed that a variant in the SLC6A4 gene, encoding the serotonin transporter, moderates the fear response to carbon dioxide in humans. To study the etiological basis of carbon dioxide-reactivity and panic attacks in more detail, we recently established a translational mouse model. AIM The purpose of this study was to investigate whether decreased expression of the serotonin transporter affects the sensitivity to carbon dioxide. METHODS Based on our previous work, wildtype and serotonin transporter deficient (+/-, -/-) mice were monitored while being exposed to carbon dioxide-enriched air. In wildtype and serotonin transporter +/- mice, also cardio-respiration was assessed. RESULTS For most behavioral measures under air exposure, wildtype and serotonin transporter +/- mice did not differ, while serotonin transporter -/- mice showed more fear-related behavior. Carbon dioxide exposure evoked a marked increase in fear-related behaviors, independent of genotype, with the exception of time serotonin transporter -/- mice spent in the center zone of the modified open field test and freezing in the two-chamber test. On the physiological level, when inhaling carbon dioxide, the respiratory system was strongly activated and heart rate decreased independent of genotype. CONCLUSION Carbon dioxide is a robust fear-inducing stimulus. It evokes inhibitory behavioral responses such as decreased exploration and is associated with a clear respiratory profile independent of serotonin transporter genotype.
Collapse
Affiliation(s)
- Nicole K Leibold
- Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, The Netherlands,Department of Neurology, Yale School of Medicine, New Haven, USA,Nicole K Leibold, Department of Psychiatry and Neuropsychology, Maastricht University, P.O. Box 616 (Vijverdal), 6200 MD Maastricht, The Netherlands.
| | - Daniel LA van den Hove
- Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, The Netherlands,School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands,Division of Molecular Psychiatry, Center of Mental Health, University of Wuerzburg, Wuerzburg, Germany
| | - Magdalena T Weidner
- Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, The Netherlands,School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands,Division of Molecular Psychiatry, Center of Mental Health, University of Wuerzburg, Wuerzburg, Germany,Department of Psychiatry and Psychotherapy, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Gordon F Buchanan
- Department of Neurology, Yale School of Medicine, New Haven, USA,Department of Neurology, University of Iowa, Iowa City, USA,University of Iowa Graduate College, Iowa City, USA
| | - Harry WM Steinbusch
- Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, The Netherlands,School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands,Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technoglogy (DGIST), Daegu, South Korea
| | - Klaus-Peter Lesch
- Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, The Netherlands,School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands,Division of Molecular Psychiatry, Center of Mental Health, University of Wuerzburg, Wuerzburg, Germany,Laboratory of Psychiatric Neurobiology, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Koen RJ Schruers
- Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, The Netherlands,School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands,Department of Psychology, University of Leuven, Leuven, Belgium
| |
Collapse
|
70
|
Traverso S. Anxiety and depression: A matter of stiffness? Med Hypotheses 2020; 145:110344. [PMID: 33075584 DOI: 10.1016/j.mehy.2020.110344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 09/06/2020] [Accepted: 10/07/2020] [Indexed: 10/23/2022]
Abstract
Cells react to stress by the universal responses of "fluidization" or "reinforcement" (stiffening) of the cytoplasm, through dramatic re-arrangements of the cytoskeleton. Here it is suggested that, at a supracellular level, the brain exhibits such a fundamental behavior as part of its complex response to stress: it is hypothesized that the soft gel formed by brain cell cytoskeletons and the surrounding extracellular matrix (the "cytoskeletons-matrix system") undergoes transitions either to sol (fluidization) or stiff gel (reinforcement) as a very fundamental and evolutionarily conserved brain response to stress, alongside more sophisticated neural pathways. Sol state corresponds to increased cell activity (a sort of "fight or flight" response), whereas stiff gel state corresponds to inactivity (an "immobility" strategy). Psychological stress, through simple stress signals such as pH changes, would lead to an initial tissue fluidization in key regions of the brain, followed, if the stress stimuli persist, by reinforcement (slow formation of actomyosin stress fibers and matrix stiffening). It is also hypothesized that the cytoskeletons-matrix system is one of the biological correlates of so-called "background feelings", i.e conscious feelings built on inner chemical-physical states of the body. Optimal dynamics of the cytoskeletons-matrix system would contribute to a core feeling of well-being, while shifts towards fluidization (activation) or stiffening (inactivation) would contribute to background feelings at the basis of anxiety and stress-induced depression, respectively. It is suggested that the cytoskeletons-matrix system behaves as a "self-organized critical system", anxiety and depression arising whenever the system is driven too far from the optimal critical point. Finally, some application hints from the proposed ideas are given.
Collapse
|
71
|
Park J, Tabet A, Moon J, Chiang PH, Koehler F, Sahasrabudhe A, Anikeeva P. Remotely Controlled Proton Generation for Neuromodulation. NANO LETTERS 2020; 20:6535-6541. [PMID: 32786937 PMCID: PMC8558523 DOI: 10.1021/acs.nanolett.0c02281] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Understanding and modulating proton-mediated biochemical processes in living organisms have been impeded by the lack of tools to control local pH. Here, we design nanotransducers capable of converting noninvasive alternating magnetic fields (AMFs) into protons in physiological environments by combining magnetic nanoparticles (MNPs) with polymeric scaffolds. When exposed to AMFs, the heat dissipated by MNPs triggered a hydrolytic degradation of surrounding polyanhydride or polyester, releasing protons into the extracellular space. pH changes induced by these nanotransducers can be tuned by changing the polymer chemistry or AMF stimulation parameters. Remote magnetic control of local protons was shown to trigger acid-sensing ion channels and to evoke intracellular calcium influx in neurons. By offering a wireless modulation of local pH, our approach can accelerate the mechanistic investigation of the role of protons in biochemical signaling in the nervous system.
Collapse
Affiliation(s)
- Jimin Park
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge Massachusetts 02139, United States
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge Massachusetts 02139, United States
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Anthony Tabet
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge Massachusetts 02139, United States
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Junsang Moon
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge Massachusetts 02139, United States
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge Massachusetts 02139, United States
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Po-Han Chiang
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge Massachusetts 02139, United States
- Institute of Biomedical Engineering, National Chiao Tung University, Hsinchu City, Taiwan 30010, ROC
| | - Florian Koehler
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge Massachusetts 02139, United States
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Atharva Sahasrabudhe
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge Massachusetts 02139, United States
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Polina Anikeeva
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge Massachusetts 02139, United States
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge Massachusetts 02139, United States
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
72
|
Histidine Residues Are Responsible for Bidirectional Effects of Zinc on Acid-Sensing Ion Channel 1a/3 Heteromeric Channels. Biomolecules 2020; 10:biom10091264. [PMID: 32887365 PMCID: PMC7565092 DOI: 10.3390/biom10091264] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 08/26/2020] [Accepted: 08/31/2020] [Indexed: 12/16/2022] Open
Abstract
Acid-sensing ion channel (ASIC) subunits 1a and 3 are highly expressed in central and peripheral sensory neurons, respectively. Endogenous biomolecule zinc plays a critical role in physiological and pathophysiological conditions. Here, we found that currents recorded from heterologously expressed ASIC1a/3 channels using the whole-cell patch-clamp technique were regulated by zinc with dual effects. Co-application of zinc dose-dependently potentiated both peak amplitude and the sustained component of heteromeric ASIC1a/3 currents; pretreatment with zinc between 3 to 100 µM exerted the same potentiation as co-application. However, pretreatment with zinc induced a significant inhibition of heteromeric ASIC1a/3 channels when zinc concentrations were over 250 µM. The potentiation of heteromeric ASIC1a/3 channels by zinc was pH dependent, as zinc shifted the pH dependence of ASIC1a/3 currents from a pH50 of 6.54 to 6.77; whereas the inhibition of ASIC1a/3 currents by zinc was also pH dependent. Furthermore, we systematically mutated histidine residues in the extracellular domain of ASIC1a or ASIC3 and found that histidine residues 72 and 73 in both ASIC1a and ASIC3, and histidine residue 83 in the ASIC3 were responsible for bidirectional effects on heteromeric ASIC1a/3 channels by zinc. These findings suggest that histidine residues in the extracellular domain of heteromeric ASIC1a/3 channels are critical for zinc-mediated effects.
Collapse
|
73
|
Hossain MZ, Ando H, Unno S, Kitagawa J. Targeting Chemosensory Ion Channels in Peripheral Swallowing-Related Regions for the Management of Oropharyngeal Dysphagia. Int J Mol Sci 2020; 21:E6214. [PMID: 32867366 PMCID: PMC7503421 DOI: 10.3390/ijms21176214] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 08/24/2020] [Accepted: 08/26/2020] [Indexed: 12/22/2022] Open
Abstract
Oropharyngeal dysphagia, or difficulty in swallowing, is a major health problem that can lead to serious complications, such as pulmonary aspiration, malnutrition, dehydration, and pneumonia. The current clinical management of oropharyngeal dysphagia mainly focuses on compensatory strategies and swallowing exercises/maneuvers; however, studies have suggested their limited effectiveness for recovering swallowing physiology and for promoting neuroplasticity in swallowing-related neuronal networks. Several new and innovative strategies based on neurostimulation in peripheral and cortical swallowing-related regions have been investigated, and appear promising for the management of oropharyngeal dysphagia. The peripheral chemical neurostimulation strategy is one of the innovative strategies, and targets chemosensory ion channels expressed in peripheral swallowing-related regions. A considerable number of animal and human studies, including randomized clinical trials in patients with oropharyngeal dysphagia, have reported improvements in the efficacy, safety, and physiology of swallowing using this strategy. There is also evidence that neuroplasticity is promoted in swallowing-related neuronal networks with this strategy. The targeting of chemosensory ion channels in peripheral swallowing-related regions may therefore be a promising pharmacological treatment strategy for the management of oropharyngeal dysphagia. In this review, we focus on this strategy, including its possible neurophysiological and molecular mechanisms.
Collapse
Affiliation(s)
- Mohammad Zakir Hossain
- Department of Oral Physiology, School of Dentistry, Matsumoto Dental University, 1780 Gobara Hirooka, Shiojiri, Nagano 399-0781, Japan;
| | - Hiroshi Ando
- Department of Biology, School of Dentistry, Matsumoto Dental University, 1780 Gobara, Hirooka, Shiojiri, Nagano 399-0781, Japan;
| | - Shumpei Unno
- Department of Oral Physiology, School of Dentistry, Matsumoto Dental University, 1780 Gobara Hirooka, Shiojiri, Nagano 399-0781, Japan;
| | - Junichi Kitagawa
- Department of Oral Physiology, School of Dentistry, Matsumoto Dental University, 1780 Gobara Hirooka, Shiojiri, Nagano 399-0781, Japan;
| |
Collapse
|
74
|
Protein Kinase C Regulates ASIC1a Protein Expression and Channel Function via NF-kB Signaling Pathway. Mol Neurobiol 2020; 57:4754-4766. [PMID: 32783140 DOI: 10.1007/s12035-020-02056-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 08/03/2020] [Indexed: 10/23/2022]
Abstract
Tissue acidosis is a common feature in many pathological conditions. Activation of acid-sensing ion channel 1a (ASIC1a) plays a key role in acidosis-mediated neurotoxicity. Protein kinase C (PKC) activity has been proved to be associated with many physiological processes and pathological conditions; however, whether PKC activation regulates ASIC1a protein expression and channel function remains ill defined. In this study, we demonstrated that treatment with phorbol 12-myristate 13-acetate (PMA, a PKC activator) for 6 h significantly increased ASIC1a protein expression and ASIC currents in NS20Y cells, a neuronal cell line, and in primary cultured mouse cortical neurons. In contrast, treatment with Calphostin C (a nonselective PKC inhibitor) for 6 h or longer decreased ASIC1a protein expression and ASIC currents. Similar to Calphostin C, PKC α and βI inhibitor Go6976 exposure also reduced ASIC1a protein expression. The reduction in ASIC1a protein expression by PKC inhibition involves a change in ASIC1a protein degradation, which is mediated by ubiquitin-proteasome system (UPS)-dependent degradation pathway. In addition, we showed that PKC regulation of ASIC1a protein expression involves NF-κB signaling pathway. Consistent with their effects on ASIC1a protein expression and channel function, PKC inhibition protected NS20Y cells against acidosis-induced cytotoxicity, while PKC activation potentiated acidosis-induced cells injury. Together, these results indicate that ASIC1a protein expression and channel function are closely regulated by the activity of protein kinase C and its downstream signaling pathway(s).
Collapse
|
75
|
Stein DJ. The respiratory subtype of panic disorder: reflections on the past and future of biological psychiatry. REVISTA BRASILEIRA DE PSIQUIATRIA (SAO PAULO, BRAZIL : 1999) 2020; 42:340-341. [PMID: 32638918 PMCID: PMC7430390 DOI: 10.1590/1516-4446-2020-1076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 04/29/2020] [Indexed: 11/22/2022]
Affiliation(s)
- Dan J Stein
- UCT Department of Psychiatry, Groote Schuur Hospital J2, Cape Town, South Africa
| |
Collapse
|
76
|
Modulation of fear behavior and neuroimmune alterations in house dust mite exposed A/J mice, a model of severe asthma. Brain Behav Immun 2020; 88:688-698. [PMID: 32380274 PMCID: PMC8988097 DOI: 10.1016/j.bbi.2020.04.084] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 04/23/2020] [Accepted: 04/30/2020] [Indexed: 12/24/2022] Open
Abstract
Fear-associated conditions such as posttraumatic stress disorder (PTSD) and panic disorder (PD) are highly prevalent. There is considerable interest in understanding contributory risk and vulnerability factors. Accumulating evidence suggests that chronically elevated inflammatory load may be a potential risk factor for these disorders. In this regard, an association of asthma, a chronic inflammatory condition, with PTSD and PD has been reported. Symptoms of PD and PTSD are more prevalent in severe asthmatics, compared to those with mild or moderate asthma suggesting that factors that influence the severity of asthma, may also influence susceptibility to the development of fear-related disorders. There has been relatively little progress in identifying contributory factors and underlying mechanisms, particularly, the translation of severe asthma-associated lung inflammation to central neuroimmune alterations and behavioral manifestations remains unclear. The current study investigated the expression of behaviors relevant to PD and PTSD (CO2 inhalation and fear conditioning/extinction) in A/J mice using a model of severe allergic asthma associated with a mixed T helper 2 (Th2) and Th17 immune response. We also investigated the accumulation of Th2- and Th17-cytokine expressing cells in lung and brain tissue, microglial alterations, as well as neuronal activation marker, delta FosB (ΔFosB)) in fear and panic regulatory brain areas. HDM-exposed mice elicited higher freezing during fear extinction. CO2-associated spontaneous and conditioned freezing, as well as anxiety or depression-relevant exploratory and coping behaviors were not altered by HDM treatment. A significant increase in brain Th17-associated inflammatory mediators was observed prior to behavioral testing, accompanied by microglial alterations in specialized blood brain barrier-compromised circumventricular area, subfornical organ. Post extinction measurements revealed increased ΔFosB staining within the medial prefrontal cortex and basolateral amygdala in HDM-treated mice. Collectively, our data show modulation of brain immune mechanisms and fear circuits by peripheral airway inflammation, and is relevant to understanding the risk and comorbidity of asthma with fear-associated disorders such as PTSD.
Collapse
|
77
|
Abstract
The aim of this review is to summarize evidence regarding rat emotional experiences during carbon dioxide (CO2) exposure. The studies reviewed show that CO2 exposure is aversive to rats, and that rats respond to CO2 exposure with active and passive defense behaviors. Plasma corticosterone and bradycardia increased in rats exposed to CO2. As with anxiogenic drugs, responses to CO2 are counteracted by the administration of anxiolytics, SRIs, and SSRI's. Human studies reviewed indicate that, when inhaling CO2, humans experience feelings of anxiety fear and panic, and that administration of benzodiazepines, serotonin precursors, and SSRIs ameliorate these feelings. In vivo and in vitro rat studies reviewed show that brain regions, ion channels, and neurotransmitters involved in negative emotional responses are activated by hypercapnia and acidosis associated with CO2 exposure. On the basis of the behavioral, physiological, and neurobiological evidence reviewed, we conclude that CO2 elicits negative emotions in rats.
Collapse
|
78
|
Turner PV, Hickman DL, van Luijk J, Ritskes-Hoitinga M, Sargeant JM, Kurosawa TM, Agui T, Baumans V, Choi WS, Choi YK, Flecknell PA, Lee BH, Otaegui PJ, Pritchett-Corning KR, Shimada K. Welfare Impact of Carbon Dioxide Euthanasia on Laboratory Mice and Rats: A Systematic Review. Front Vet Sci 2020; 7:411. [PMID: 32793645 PMCID: PMC7387666 DOI: 10.3389/fvets.2020.00411] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 06/09/2020] [Indexed: 12/27/2022] Open
Abstract
Background: There has been increased concern about the suitability of CO2 as a method for euthanasia of laboratory mice and rats, including the potential discomfort, pain or distress that animals may experience prior to loss of consciousness; time to loss of consciousness; best methods for use of CO2; and the availability of better alternatives. These discussions have been useful in providing new information, but have resulted in significant confusion regarding the acceptability of CO2 for rodent euthanasia. In some cases, researchers and veterinarians have become uncertain as to which techniques to recommend or use for euthanasia of laboratory mice and rats. Methods: The International Association of Colleges of Laboratory Animal Medicine (IACLAM) convened a taskforce to examine the evidence for adverse welfare indicators in laboratory rats and mice undergoing CO2 euthanasia using a SYRCLE-registered systematic review protocol. Of 3,772 papers identified through a database search (PubMed, Web of Science, CAB Direct, Agricola, and grey literature) from 1900 to 2017, 37 studies were identified for detailed review (some including more than one species or age group), including 15 in adult mice, 21 in adult rats, and 5 in neonates of both species. Experiments or reports were excluded if they only assessed parameters other than those directly affecting animal welfare during CO2 induction and/or euthanasia. Results: Study design and outcome measures were highly variable and there was an unclear to high risk of bias in many of the published studies. Changes in the outcome measures evaluated were inconsistent or poorly differentiated. It is likely that repeated exposures to carbon dioxide inhalation are aversive to adult rats and mice, based on avoidance behavior studies; however, this effect is largely indistinguishable from aversion induced by repeated exposures to other inhalant anesthetic gasses. Conclusion: There is insufficient evidence to permit an unbiased assessment of the effect of CO2 inhalation during euthanasia on welfare indicators in laboratory mice and rats. Additional well-designed, unbiased, and adequately powered studies are needed to accurately assess the welfare of laboratory mice and rats undergoing euthanasia via CO2 gas.
Collapse
Affiliation(s)
- Patricia V. Turner
- Department of Pathobiology, University of Guelph, Guelph, ON, Canada
- Global Animal Welfare and Training, Charles River, Wilmington, MA, United States
| | - Debra L. Hickman
- Laboratory Animal Resource Center, School of Medicine, Indiana University, Indianapolis, IN, United States
| | - Judith van Luijk
- Department of Health Evidence, SYstematic Review Center for Laboratory Experimentation (SYRCLE), Radboud University, Nijmegen, Netherlands
| | - Merel Ritskes-Hoitinga
- Department of Health Evidence, SYstematic Review Center for Laboratory Experimentation (SYRCLE), Radboud University, Nijmegen, Netherlands
| | - Jan M. Sargeant
- Department of Population Medicine, University of Guelph, Guelph, ON, Canada
- Centre for Public Health and Zoonoses, University of Guelph, Guelph, ON, Canada
| | - T. Miki Kurosawa
- Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan
| | - Takashi Agui
- Department of Applied Veterinary Science, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Vera Baumans
- Department of Animals, Science and Society, Utrecht University, Utrecht, Netherlands
| | - Woo Sung Choi
- National New Drug Development Cluster, Woojung Bio, Suwon-si, South Korea
| | - Yang-Kyu Choi
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, Seoul, South Korea
| | - Paul A. Flecknell
- Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Byeong H. Lee
- Osong Medical Innovation Foundation, Cheongju, South Korea
| | - Pedro J. Otaegui
- Laboratory Animal Facilities, Autonomous University of Barcelona, Barcelona, Spain
| | - Kathleen R. Pritchett-Corning
- Office of Animal Resources, Harvard University Faculty of Arts and Sciences, Cambridge, MA, United States
- Department of Comparative Medicine, University of Washington, Seattle, WA, United States
| | - Keisuke Shimada
- Animal Resource Center for Infectious Diseases, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| |
Collapse
|
79
|
Osmakov DI, Khasanov TA, Andreev YA, Lyukmanova EN, Kozlov SA. Animal, Herb, and Microbial Toxins for Structural and Pharmacological Study of Acid-Sensing Ion Channels. Front Pharmacol 2020; 11:991. [PMID: 32733241 PMCID: PMC7360831 DOI: 10.3389/fphar.2020.00991] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 06/19/2020] [Indexed: 12/22/2022] Open
Abstract
Acid-sensing ion channels (ASICs) are of the most sensitive molecular sensors of extracellular pH change in mammals. Six isoforms of these channels are widely represented in membranes of neuronal and non-neuronal cells, where these molecules are involved in different important regulatory functions, such as synaptic plasticity, learning, memory, and nociception, as well as in various pathological states. Structural and functional studies of both wild-type and mutant ASICs are essential for human care and medicine for the efficient treatment of socially significant diseases and ensure a comfortable standard of life. Ligands of ASICs serve as indispensable tools for these studies. Such bioactive compounds can be synthesized artificially. However, to date, the search for such molecules has been most effective amongst natural sources, such as animal venoms or plants and microbial extracts. In this review, we provide a detailed and comprehensive structural and functional description of natural compounds acting on ASICs, as well as the latest information on structural aspects of their interaction with the channels. Many of the examples provided in the review demonstrate the undoubted fundamental and practical successes of using natural toxins. Without toxins, it would not be possible to obtain data on the mechanisms of ASICs' functioning, provide detailed study of their pharmacological properties, or assess the contribution of the channels to development of different pathologies. The selectivity to different isoforms and variety in the channel modulation mode allow for the appraisal of prospective candidates for the development of new drugs.
Collapse
Affiliation(s)
- Dmitry I Osmakov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Science, Moscow, Russia
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Timur A Khasanov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Science, Moscow, Russia
| | - Yaroslav A Andreev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Science, Moscow, Russia
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Ekaterina N Lyukmanova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Science, Moscow, Russia
| | - Sergey A Kozlov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Science, Moscow, Russia
| |
Collapse
|
80
|
Liu X, Liu C, Ye J, Zhang S, Wang K, Su R. Distribution of Acid Sensing Ion Channels in Axonal Growth Cones and Presynaptic Membrane of Cultured Hippocampal Neurons. Front Cell Neurosci 2020; 14:205. [PMID: 32733209 PMCID: PMC7358772 DOI: 10.3389/fncel.2020.00205] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 06/10/2020] [Indexed: 12/24/2022] Open
Abstract
Although acid-sensing ion channels (ASICs) are widely expressed in the central nervous system, their distribution and roles in axonal growth cones remain unclear. In this study, we examined ASIC localization and function in the axonal growth cones of cultured immature hippocampal neurons. Our immunocytochemical data showed that native and overexpressed ASIC1a and ASIC2a are both localized in growth cones of cultured young hippocampal neurons. Calcium imaging and electrophysiological assay results were utilized to validate their function. The calcium imaging test results indicated that the ASICs (primarily ASIC1a) present in growth cones mediate calcium influx despite the addition of voltage-gated Ca2+ channels antagonists and the depletion of intracellular calcium stores. The electrophysiological tests results suggested that a rapid decrease in extracellular pH at the growth cones of voltage-clamped neurons elicits inward currents that were blocked by bath application of the ASIC antagonist amiloride, showing that the ASICs expressed at growth cones are functional. The subsequent immuno-colocalization test results demonstrated that ASIC1a and ASIC2a are both colocalized with Neurofilament-H and Bassoon in mature hippocampal neurons. This finding demonstrated that after reaching maturity, ASIC1a and ASIC2a are both distributed in axons and the presynaptic membrane. Our data reveal the distribution of functional ASICs in growth cones of immature hippocampal neurons and the presence of ASICs in the axons and presynaptic membrane of mature hippocampal neurons, indicating a possible role for ASICs in axonal guidance, synapse formation and neurotransmitter release.
Collapse
Affiliation(s)
- Xiaoyan Liu
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Can Liu
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Jiamin Ye
- School of Pharmacy, North China University of Science and Technology, Tangshan, China
| | - Shuzhuo Zhang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Kai Wang
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Ruibin Su
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| |
Collapse
|
81
|
Bignucolo O, Vullo S, Ambrosio N, Gautschi I, Kellenberger S. Structural and Functional Analysis of Gly212 Mutants Reveals the Importance of Intersubunit Interactions in ASIC1a Channel Function. Front Mol Biosci 2020; 7:58. [PMID: 32411719 PMCID: PMC7198790 DOI: 10.3389/fmolb.2020.00058] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 03/23/2020] [Indexed: 12/27/2022] Open
Abstract
Acid-sensing ion channels (ASICs) act as pH sensors in neurons. ASICs contribute to pain sensation, learning, fear behavior and to neuronal death after ischemic stroke. Extracellular acidification induces a transient activation and subsequent desensitization of these Na+-selective channels. ASICs are trimeric channels made of identical or homologous subunits. We have previously shown that mutation of the highly conserved Gly212 residue of human ASIC1a to Asp affects the channel function. Gly212 is located in the proximity of a predicted Cl– binding site at a subunit interface. Here, we have measured the function of a series of Gly212 mutants. We show that substitution of Gly212 affects the ASIC1a pH dependence and current decay kinetics. Intriguingly, the mutations to the acidic residues Asp and Glu have opposing effects on the pH dependence and the current decay kinetics. Analysis of molecular dynamics simulation trajectories started with the coordinates of the closed conformation indicates that the immediate environment of residue 212 in G212E, which shifts the pH dependence to more alkaline values, adopts a conformation closer to the open state. The G212D and G212E mutants have a different pattern of intersubunit salt bridges, that, in the case of G212E, leads to an approaching of neighboring subunits. Based on the comparison of crystal structures, the conformational changes in this zone appear to be smaller during the open-desensitized transition. Nevertheless, MD simulations highlight differences between mutants, suggesting that the changed function upon substitution of residue 212 is due to differences in intra- and intersubunit interactions in its proximity.
Collapse
Affiliation(s)
- Olivier Bignucolo
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland.,SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Sabrina Vullo
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Nicolas Ambrosio
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Ivan Gautschi
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Stephan Kellenberger
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
82
|
Potential Role of the Amygdala and Posterior Claustrum in Exercise Intensity-dependent Cardiovascular Regulation in Rats. Neuroscience 2020; 432:150-159. [PMID: 32109531 DOI: 10.1016/j.neuroscience.2020.02.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Revised: 02/14/2020] [Accepted: 02/17/2020] [Indexed: 11/21/2022]
Abstract
Tuning of the cardiovascular response is crucial to maintain performance during high-intensity exercise. It is well known that the nucleus of the solitary tract (NTS) in the brainstem medulla plays a central role in cardiovascular regulation; however, where and how upper brain regions form circuits with NTS and coordinately control cardiovascular responses during high-intensity exercise remain unclear. Here focusing on the amygdala and claustrum, we investigated part of the mechanism for regulation of the cardiovascular system during exercise. In rats, c-Fos immunostaining was used to examine whether the amygdala and claustrum were activated during treadmill exercise. Further, we examined arterial pressure responses to electrical and chemical stimulation of the claustrum region. We also confirmed the anatomical connections between the amygdala, claustrum, and NTS by retrograde tracer injections. Finally, we performed simultaneous electrical stimulation of the claustrum and amygdala to examine their functional connectivity. c-Fos expression was observed in the amygdala and the posterior part of the claustrum (pCL), but not in the anterior part, in an exercise intensity-dependent manner. pCL stimulation induced a depressor response. Using a retrograde tracer, we confirmed direct projections from the amygdala to the pCL and NTS. Simultaneous stimulation of the central nucleus of the amygdala and pCL showed a greater pressor response compared with the stimulation of the amygdala alone. These results suggest the amygdala and pCL are involved in different phases of exercise. More speculatively, these areas might coordinately tune cardiovascular responses that help maintain performance during high-intensity exercise.
Collapse
|
83
|
A molecular view of the function and pharmacology of acid-sensing ion channels. Pharmacol Res 2020; 154:104166. [DOI: 10.1016/j.phrs.2019.02.005] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 02/01/2019] [Accepted: 02/02/2019] [Indexed: 02/06/2023]
|
84
|
High Behavioral Sensitivity to Carbon Dioxide Associates with Enhanced Fear Memory and Altered Forebrain Neuronal Activation. Neuroscience 2020; 429:92-105. [PMID: 31930959 DOI: 10.1016/j.neuroscience.2019.12.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 11/13/2019] [Accepted: 12/08/2019] [Indexed: 01/06/2023]
Abstract
There is considerable interest in pre-trauma individual differences that may contribute to increased risk for developing post-traumatic stress disorder (PTSD). Identification of underlying vulnerability factors that predict differential responses to traumatic experiences is important. Recently, the relevance of homeostatic perturbations in shaping long-term behavior has been recognized. Sensitivity to CO2 inhalation, a homeostatic threat to survival, was shown to associate with the later development of PTSD symptoms in veterans. Here, we investigated whether behavioral sensitivity to CO2 associates with PTSD-relevant behaviors and alters forebrain fear circuitry in mice. Mice were exposed to 5% CO2 or air inhalation and tested one week later on acoustic startle and footshock contextual fear conditioning, extinction and reinstatement. CO2 inhalation evoked heterogenous freezing behaviors (high freezing CO2-H and low freezing CO2-L) that significantly associated with fear conditioning and extinction behaviors. CO2-H mice elicited potentiated conditioned fear and delayed extinction while behavioral responses in CO2-L mice were similar to the air group. Persistent neuronal activation marker ΔFosB immunostaining revealed altered regional neuronal activation within the hippocampus, amygdala and medial pre-frontal cortex that correlated with conditioned fear and extinction. Inter-regional co-activation mapping revealed disruptions in the coordinated activity of hippocampal dentate-amygdala-infralimbic regions and infralimbic-prelimbic associations in CO2-H mice that may explain their enhanced fear phenotype. In conclusion, our data support an association of behavioral sensitivity to interoceptive threats such as CO2 with altered fear responding to exteroceptive threats and suggest that "CO2-sensitive" individuals may be susceptible to developing PTSD.
Collapse
|
85
|
Okuro RT, Freire RC, Zin WA, Quagliato LA, Nardi AE. Panic disorder respiratory subtype: psychopathology and challenge tests - an update. ACTA ACUST UNITED AC 2020; 42:420-430. [PMID: 32074230 PMCID: PMC7430397 DOI: 10.1590/1516-4446-2019-0717] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 12/01/2019] [Indexed: 02/06/2023]
Abstract
Panic disorder (PD) pathophysiology is very heterogeneous, and the discrimination of distinct subtypes could be very useful. A subtype based on respiratory symptoms is known to constitute a specific subgroup. However, evidence to support the respiratory subtype (RS) as a distinct subgroup of PD with a well-defined phenotype remains controversial. Studies have focused on characterization of the RS based on symptoms and response to CO2. In this line, we described clinical and biological aspects focused on symptomatology and CO2 challenge tests in PD RS. The main symptoms that characterize RS are dyspnea (shortness of breath) and a choking sensation. Moreover, patients with the RS tended to be more responsive to CO2 challenge tests, which triggered more panic attacks in this subgroup. Future studies should focus on discriminating respiratory-related clusters and exploring psychophysiological and neuroimaging outcomes in order to provide robust evidence to confirm RS as a distinct subtype of PD.
Collapse
Affiliation(s)
- Renata T Okuro
- Laboratório Pânico e Respiração, Instituto de Psiquiatria (IPUB), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| | - Rafael C Freire
- Laboratório Pânico e Respiração, Instituto de Psiquiatria (IPUB), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| | - Walter A Zin
- Instituto de Biofísica Carlos Chagas Filho, UFRJ, Rio de Janeiro, RJ, Brazil
| | - Laiana A Quagliato
- Laboratório Pânico e Respiração, Instituto de Psiquiatria (IPUB), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| | - Antonio E Nardi
- Laboratório Pânico e Respiração, Instituto de Psiquiatria (IPUB), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| |
Collapse
|
86
|
Takagaki N, Ohta A, Ohnishi K, Kawanabe A, Minakuchi Y, Toyoda A, Fujiwara Y, Kuhara A. The mechanoreceptor DEG-1 regulates cold tolerance in Caenorhabditis elegans. EMBO Rep 2020; 21:e48671. [PMID: 32009302 PMCID: PMC7054665 DOI: 10.15252/embr.201948671] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 12/19/2019] [Accepted: 12/20/2019] [Indexed: 12/13/2022] Open
Abstract
Caenorhabditis elegans mechanoreceptors located in ASG sensory neurons have been found to sense ambient temperature, which is a key trait for animal survival. Here, we show that experimental loss of xanthine dehydrogenase (XDH-1) function in AIN and AVJ interneurons results in reduced cold tolerance and atypical neuronal response to changes in temperature. These interneurons connect with upstream neurons such as the mechanoreceptor-expressing ASG. Ca2+ imaging revealed that ASG neurons respond to warm temperature via the mechanoreceptor DEG-1, a degenerin/epithelial Na+ channel (DEG/ENaC), which in turn affects downstream AIN and AVJ circuits. Ectopic expression of DEG-1 in the ASE gustatory neuron results in the acquisition of warm sensitivity, while electrophysiological analysis revealed that DEG-1 and human MDEG1 were involved in warm sensation. Taken together, these results suggest that cold tolerance is regulated by mechanoreceptor-mediated circuit calculation.
Collapse
Affiliation(s)
- Natsune Takagaki
- Graduate School of Natural Science, Konan University, Kobe, Japan.,Institute for Integrative Neurobiology, Konan University, Kobe, Japan
| | - Akane Ohta
- Graduate School of Natural Science, Konan University, Kobe, Japan.,Institute for Integrative Neurobiology, Konan University, Kobe, Japan.,Faculty of Science and Engineering, Konan University, Kobe, Japan
| | - Kohei Ohnishi
- Graduate School of Natural Science, Konan University, Kobe, Japan.,Institute for Integrative Neurobiology, Konan University, Kobe, Japan
| | - Akira Kawanabe
- Laboratory of Molecular Physiology & Biophysics, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Yohei Minakuchi
- Advanced Genomics Center, National Institute of Genetics, Mishima, Shizuoka, Japan.,Comparative Genomics Laboratory, National Institute of Genetics, Mishima, Shizuoka, Japan
| | - Atsushi Toyoda
- Advanced Genomics Center, National Institute of Genetics, Mishima, Shizuoka, Japan.,Comparative Genomics Laboratory, National Institute of Genetics, Mishima, Shizuoka, Japan
| | - Yuichiro Fujiwara
- Laboratory of Molecular Physiology & Biophysics, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Atsushi Kuhara
- Graduate School of Natural Science, Konan University, Kobe, Japan.,Institute for Integrative Neurobiology, Konan University, Kobe, Japan.,Faculty of Science and Engineering, Konan University, Kobe, Japan.,AMED-PRIME, Japan Agency for Medical Research and Development, Tokyo, Japan
| |
Collapse
|
87
|
Impaired endothelium-mediated cerebrovascular reactivity promotes anxiety and respiration disorders in mice. Proc Natl Acad Sci U S A 2020; 117:1753-1761. [PMID: 31896584 DOI: 10.1073/pnas.1907467117] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Carbon dioxide (CO2), the major product of metabolism, has a strong impact on cerebral blood vessels, a phenomenon known as cerebrovascular reactivity. Several vascular risk factors such as hypertension or diabetes dampen this response, making cerebrovascular reactivity a useful diagnostic marker for incipient vascular pathology, but its functional relevance, if any, is still unclear. Here, we found that GPR4, an endothelial H+ receptor, and endothelial Gαq/11 proteins mediate the CO2/H+ effect on cerebrovascular reactivity in mice. CO2/H+ leads to constriction of vessels in the brainstem area that controls respiration. The consequential washout of CO2, if cerebrovascular reactivity is impaired, reduces respiration. In contrast, CO2 dilates vessels in other brain areas such as the amygdala. Hence, an impaired cerebrovascular reactivity amplifies the CO2 effect on anxiety. Even at atmospheric CO2 concentrations, impaired cerebrovascular reactivity caused longer apneic episodes and more anxiety, indicating that cerebrovascular reactivity is essential for normal brain function. The site-specific reactivity of vessels to CO2 is reflected by regional differences in their gene expression and the release of vasoactive factors from endothelial cells. Our data suggest the central nervous system (CNS) endothelium as a target to treat respiratory and affective disorders associated with vascular diseases.
Collapse
|
88
|
Steiner AR, Flammer SA, Beausoleil NJ, Berg C, Bettschart-Wolfensberger R, Pinillos RG, Golledge HDW, Marahrens M, Meyer R, Schnitzer T, Toscano MJ, Turner PV, Weary DM, Gent TC. Humanely Ending the Life of Animals: Research Priorities to Identify Alternatives to Carbon Dioxide. Animals (Basel) 2019; 9:E911. [PMID: 31684044 PMCID: PMC6912382 DOI: 10.3390/ani9110911] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 10/14/2019] [Accepted: 10/25/2019] [Indexed: 02/06/2023] Open
Abstract
: The use of carbon dioxide (CO2) for stunning and killing animals is considered to compromise welfare due to air hunger, anxiety, fear, and pain. Despite decades of research, no alternatives have so far been found that provide a safe and reliable way to induce unconsciousness in groups of animals, and also cause less distress than CO2. Here, we revisit the current and historical literature to identify key research questions that may lead to the identification and implementation of more humane alternatives to induce unconsciousness in mice, rats, poultry, and pigs. In addition to the evaluation of novel methods and agents, we identify the need to standardise the terminology and behavioural assays within the field. We further reason that more accurate measurements of consciousness state are needed and serve as a central component in the assessment of suffering. Therefore, we propose a roadmap toward improving animal welfare during end-of-life procedures.
Collapse
Affiliation(s)
- Aline R Steiner
- Department of Clinical and Diagnostic Services, Section of Anaesthesiology, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 258c, 8057 Zurich, Switzerland.
| | - Shannon Axiak Flammer
- Department of Clinical Veterinary Medicine, Section of Anesthesia and Analgesia, Vetsuisse Faculty, University of Berne, Laenggassstrasse 124, 3012 Bern, Switzerland.
| | - Ngaio J Beausoleil
- Animal Welfare Science and Bioethics Centre, School of Veterinary Science, Massey University, Palmerston North 4410, New Zealand.
| | - Charlotte Berg
- Department of Animal Environment and Health, Swedish University of Agricultural Sciences, Box 234, SE-53223 Skara, Sweden.
| | - Regula Bettschart-Wolfensberger
- Department of Clinical and Diagnostic Services, Section of Anaesthesiology, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 258c, 8057 Zurich, Switzerland.
| | - Rebeca García Pinillos
- Animal and Plant Health Agency and Department for Environment, Food and Rural Affairs, Nobel House, 17 Smith Square, London SW1P 3JR, UK.
| | - Huw D W Golledge
- Universities Federation for Animal Welfare (UFAW), The Old School, Brewhouse Hill, Wheathampstead, Hertfordshire AL4 8AN, UK.
| | - Michael Marahrens
- Institute of Animal Welfare and Animal Husbandry, Friedrich-Loeffler-Institut, Dörnbergstraße 25/27, 29223 Celle, Germany.
| | - Robert Meyer
- Department of Clinical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, MS 39762, USA.
| | - Tobias Schnitzer
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, 4070 Basel, Switzerland.
| | - Michael J Toscano
- Center for Proper Housing: Poultry and Rabbits (ZTHZ), Animal Welfare Division, VPH Institute, University of Bern, 3052 Zollikofen, Switzerland.
| | - Patricia V Turner
- Department of Pathobiology, University of Guelph, Guelph, ON, N1G 2W1, Canada and Charles River, Wilmington, MA 01887, USA.
| | - Daniel M Weary
- Animal Welfare Program, University of British Colombia, 2357 Main Mall, Vancouver, BC V6T 1Z4, Canada.
| | - Thomas C Gent
- Department of Clinical and Diagnostic Services, Section of Anaesthesiology, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 258c, 8057 Zurich, Switzerland.
| |
Collapse
|
89
|
Faraci FM, Taugher RJ, Lynch C, Fan R, Gupta S, Wemmie JA. Acid-Sensing Ion Channels: Novel Mediators of Cerebral Vascular Responses. Circ Res 2019; 125:907-920. [PMID: 31451088 PMCID: PMC6813889 DOI: 10.1161/circresaha.119.315024] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
RATIONALE Precise regulation of cerebral blood flow is critical for normal brain function. Insufficient cerebral blood flow contributes to brain dysfunction and neurodegeneration. Carbon dioxide (CO2), via effects on local acidosis, is one of the most potent regulators of cerebral blood flow. Although a role for nitric oxide in intermediate signaling has been implicated, mechanisms that initiate CO2-induced vasodilation remain unclear. OBJECTIVE Acid-sensing ion channel-1A (ASIC1A) is a proton-gated cation channel that is activated by extracellular acidosis. Based on work that implicated ASIC1A in the amygdala and bed nucleus of the stria terminalis in CO2-evoked and acid-evoked behaviors, we hypothesized that ASIC1A might also mediate microvascular responses to CO2. METHODS AND RESULTS To test this hypothesis, we genetically and pharmacologically manipulated ASIC1A and assessed effects on CO2-induced dilation of cerebral arterioles in vivo. Effects of inhalation of 5% or 10% CO2 on arteriolar diameter were greatly attenuated in mice with global deficiency in ASIC1A (Asic1a-/-) or by local treatment with the ASIC inhibitor, psalmotoxin. Vasodilator effects of acetylcholine, which acts via endothelial nitric oxide synthase were unaffected, suggesting a nonvascular source of nitric oxide may be key for CO2 responses. Thus, we tested whether neurons may be the cell type through which ASIC1A influences microvessels. Using mice in which Asic1a was specifically disrupted in neurons, we found effects of CO2 on arteriolar diameter were also attenuated. CONCLUSIONS Together, these data are consistent with a model wherein activation of ASIC1A, particularly in neurons, is critical for CO2-induced nitric oxide production and vasodilation. With these findings, ASIC1A emerges as major regulator of microvascular tone.
Collapse
Affiliation(s)
- Frank M. Faraci
- Department of Internal Medicine, Francois M. Abboud Cardiovascular Center, Papajohn Biomedical Institute, Carver College of Medicine, University of Iowa, Department of Veterans Affairs Medical Center, Iowa City, IA 52242
- Department of Pharmacology, Francois M. Abboud Cardiovascular Center, Papajohn Biomedical Institute, Carver College of Medicine, University of Iowa, Department of Veterans Affairs Medical Center, Iowa City, IA 52242
| | - Rebecca J. Taugher
- Department of Psychiatry, Francois M. Abboud Cardiovascular Center, Papajohn Biomedical Institute, Carver College of Medicine, University of Iowa, Department of Veterans Affairs Medical Center, Iowa City, IA 52242
| | - Cynthia Lynch
- Department of Internal Medicine, Francois M. Abboud Cardiovascular Center, Papajohn Biomedical Institute, Carver College of Medicine, University of Iowa, Department of Veterans Affairs Medical Center, Iowa City, IA 52242
| | - Rong Fan
- Department of Psychiatry, Francois M. Abboud Cardiovascular Center, Papajohn Biomedical Institute, Carver College of Medicine, University of Iowa, Department of Veterans Affairs Medical Center, Iowa City, IA 52242
| | - Subhash Gupta
- Department of Psychiatry, Francois M. Abboud Cardiovascular Center, Papajohn Biomedical Institute, Carver College of Medicine, University of Iowa, Department of Veterans Affairs Medical Center, Iowa City, IA 52242
| | - John A. Wemmie
- Department of Psychiatry, Francois M. Abboud Cardiovascular Center, Papajohn Biomedical Institute, Carver College of Medicine, University of Iowa, Department of Veterans Affairs Medical Center, Iowa City, IA 52242
| |
Collapse
|
90
|
Cummins EP, Strowitzki MJ, Taylor CT. Mechanisms and Consequences of Oxygen and Carbon Dioxide Sensing in Mammals. Physiol Rev 2019; 100:463-488. [PMID: 31539306 DOI: 10.1152/physrev.00003.2019] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Molecular oxygen (O2) and carbon dioxide (CO2) are the primary gaseous substrate and product of oxidative phosphorylation in respiring organisms, respectively. Variance in the levels of either of these gasses outside of the physiological range presents a serious threat to cell, tissue, and organism survival. Therefore, it is essential that endogenous levels are monitored and kept at appropriate concentrations to maintain a state of homeostasis. Higher organisms such as mammals have evolved mechanisms to sense O2 and CO2 both in the circulation and in individual cells and elicit appropriate corrective responses to promote adaptation to commonly encountered conditions such as hypoxia and hypercapnia. These can be acute and transient nontranscriptional responses, which typically occur at the level of whole animal physiology or more sustained transcriptional responses, which promote chronic adaptation. In this review, we discuss the mechanisms by which mammals sense changes in O2 and CO2 and elicit adaptive responses to maintain homeostasis. We also discuss crosstalk between these pathways and how they may represent targets for therapeutic intervention in a range of pathological states.
Collapse
Affiliation(s)
- Eoin P Cummins
- UCD Conway Institute, Systems Biology Ireland and the School of Medicine, University College Dublin, Belfield, Dublin, Ireland
| | - Moritz J Strowitzki
- UCD Conway Institute, Systems Biology Ireland and the School of Medicine, University College Dublin, Belfield, Dublin, Ireland
| | - Cormac T Taylor
- UCD Conway Institute, Systems Biology Ireland and the School of Medicine, University College Dublin, Belfield, Dublin, Ireland
| |
Collapse
|
91
|
Taugher RJ, Dlouhy BJ, Kreple CJ, Ghobbeh A, Conlon MM, Wang Y, Wemmie JA. The amygdala differentially regulates defensive behaviors evoked by CO 2. Behav Brain Res 2019; 377:112236. [PMID: 31536735 DOI: 10.1016/j.bbr.2019.112236] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 09/11/2019] [Accepted: 09/11/2019] [Indexed: 10/26/2022]
Abstract
CO2 inhalation can provoke panic attacks in humans, and the likelihood is increased in patients with panic disorder. Identifying brain sites involved could provide important mechanistic insight into the illness. In mice, the amygdala has been suggested to promote CO2 responses; however, recent studies in humans with amygdala damage indicate the amygdala is not required for CO2-induced fear and panic and might actually oppose these responses. To clarify the role of the amygdala, we produced lesions in mice paralleling the human lesions, and characterized behavioral responses to CO2. Compared to sham controls, we found that amygdala-lesioned mice froze less to 10% CO2, and unlike shams they also began to jump frenetically. At 20% CO2, controls also exhibited jumping, suggesting it is a normal response to more extreme CO2 concentrations. The effect of amygdala lesions was specific to CO2 as amygdala-lesioned mice did not jump in response to a predator odor or to an auditory conditioned stimulus. In amygdala-lesioned mice, jumping evoked by 10% CO2 was eliminated by co-lesioning the dorsal periaqueductal gray, a structure implicated in panic and escape-related behaviors. Together, these observations suggest a dual role for the amygdala in the CO2 response: promoting CO2-induced freezing, and opposing CO2-induced jumping, which may help explain the exaggerated CO2 responses in humans with amygdala lesions.
Collapse
Affiliation(s)
- R J Taugher
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA; Department of Veterans Affairs Medical Center, Iowa City, IA, USA
| | - B J Dlouhy
- Department of Neurosurgery, University of Iowa, Iowa City, IA, USA; Pappajohn Biomedical Institute, University of Iowa, Iowa City, IA, USA; Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
| | - C J Kreple
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, USA; Medical Scientist Training Program, University of Iowa, Iowa City, IA, USA
| | - A Ghobbeh
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA; Department of Veterans Affairs Medical Center, Iowa City, IA, USA
| | - M M Conlon
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA; Department of Veterans Affairs Medical Center, Iowa City, IA, USA
| | - Y Wang
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
| | - J A Wemmie
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA; Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
92
|
Ghobbeh A, Taugher RJ, Alam SM, Fan R, LaLumiere RT, Wemmie JA. A novel role for acid-sensing ion channels in Pavlovian reward conditioning. GENES, BRAIN, AND BEHAVIOR 2019; 18:e12531. [PMID: 30375184 PMCID: PMC6818262 DOI: 10.1111/gbb.12531] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 10/19/2018] [Accepted: 10/19/2018] [Indexed: 12/01/2022]
Abstract
Pavlovian fear conditioning has been shown to depend on acid-sensing ion channel-1A (ASIC1A); however, it is unknown whether conditioning to rewarding stimuli also depends on ASIC1A. Here, we tested the hypothesis that ASIC1A contributes to Pavlovian conditioning to a non-drug reward. We found effects of ASIC1A disruption depended on the relationship between the conditional stimulus (CS) and the unconditional stimulus (US), which was varied between five experiments. In experiment 1, when the CS preceded the US signaling an upcoming reward, Asic1a-/- mice exhibited a deficit in conditioning compared to Asic1a+/+ mice. Alternatively, in experiment 2, when the CS coinitiated with the US and signaled immediate reward availability, the Asic1a-/- mice exhibited an increase in conditioned responses compared to Asic1a+/+ mice, which contrasted with the deficits in the first experiment. Furthermore, in experiments 3 and 4, when the CS partially overlapped in time with the US, or the CS was shortened and coinitiated with the US, the Asic1a-/- mice did not differ from control mice. The contrasting outcomes were likely because of differences in conditioning because in experiment 5 neither the Asic1a-/- nor Asic1a+/+ mice acquired conditioned responses when the CS and US were explicitly unpaired. Taken together, these results suggest that the effects of ASIC1A disruption on reward conditioning depend on the temporal relationship between the CS and US. Furthermore, these results suggest that ASIC1A plays a critical, yet nuanced role in Pavlovian conditioning. More research will be needed to deconstruct the roles of ASIC1A in these fundamental forms of learning and memory.
Collapse
Affiliation(s)
- Ali Ghobbeh
- Department of Psychiatry, University of Iowa, Iowa City, Iowa, USA
- Department of Veterans Affairs Medical Center, Iowa City, Iowa, USA
| | - Rebecca J. Taugher
- Department of Psychiatry, University of Iowa, Iowa City, Iowa, USA
- Department of Veterans Affairs Medical Center, Iowa City, Iowa, USA
| | - Syed M. Alam
- Department of Psychiatry, University of Iowa, Iowa City, Iowa, USA
- Department of Veterans Affairs Medical Center, Iowa City, Iowa, USA
| | - Rong Fan
- Department of Psychiatry, University of Iowa, Iowa City, Iowa, USA
- Department of Veterans Affairs Medical Center, Iowa City, Iowa, USA
| | - Ryan T. LaLumiere
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, Iowa, USA
| | - John A. Wemmie
- Department of Psychiatry, University of Iowa, Iowa City, Iowa, USA
- Department of Veterans Affairs Medical Center, Iowa City, Iowa, USA
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa, USA
- Medical Scientist Training Program, University of Iowa, Iowa City, Iowa, USA
- Department of Neurosurgery, University of Iowa, Iowa City, Iowa, USA
- Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
93
|
Alternatives to Carbon Dioxide-Taking Responsibility for Humanely Ending the Life of Animals. Animals (Basel) 2019; 9:ani9080482. [PMID: 31344949 PMCID: PMC6720529 DOI: 10.3390/ani9080482] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 07/16/2019] [Accepted: 07/19/2019] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Carbon dioxide has long been considered one of the better methods for euthanizing laboratory rodents because it allows termination of several animals at one time, does not require handling of the animal, is easy to use, is inexpensive, and is environmentally friendly. Research, though, has shown that this gas is aversive to rodents and that it may be inhumane to expose them to this gas while they are conscious. Therefore, the Swiss Federal Food Safety and Veterinary Office has set out to find a suitable replacement and organized a meeting that included representatives and experts of the different stakeholders involved in this process in order to find a solution. The conclusion of this meeting was that a replacement is required, and the next step would be to draft a research strategy to find a suitable replacement. Abstract Carbon dioxide (CO2) is commonly used to kill rodents. However, a large body of research has now established that CO2 is aversive to them. A multidisciplinary symposium organized by the Swiss Federal Food Safety and Veterinary Office discussed the drawbacks and alternatives to CO2 in euthanasia protocols for laboratory animals. Dialogue was facilitated by brainstorming sessions in small groups and a “World Café”. A conclusion from this process was that alternatives to CO2 were urgently required, including a program of research and extension to meet the needs for humane killing of these animals. The next step will involve gathering a group of international experts to formulate, draft, and publish a research strategy on alternatives to CO2.
Collapse
|
94
|
Uchitel OD, González Inchauspe C, Weissmann C. Synaptic signals mediated by protons and acid-sensing ion channels. Synapse 2019; 73:e22120. [PMID: 31180161 DOI: 10.1002/syn.22120] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 06/05/2019] [Accepted: 06/05/2019] [Indexed: 01/04/2023]
Abstract
Extracellular pH changes may constitute significant signals for neuronal communication. During synaptic transmission, changes in pH in the synaptic cleft take place. Its role in the regulation of presynaptic Ca2+ currents through multivesicular release in ribbon-type synapses is a proven phenomenon. In recent years, protons have been recognized as neurotransmitters that participate in neuronal communication in synapses of several regions of the CNS such as amygdala, nucleus accumbens, and brainstem. Protons are released by nerve stimulation and activate postsynaptic acid-sensing ion channels (ASICs). Several types of ASIC channels are expressed in the peripheral and central nervous system. The influx of Ca2+ through some subtypes of ASICs, as a result of synaptic transmission, agrees with the participation of ASICs in synaptic plasticity. Pharmacological and genetical inhibition of ASIC1a results in alterations in learning, memory, and phenomena like fear and cocaine-seeking behavior. The recognition of endogenous molecules, such as arachidonic acid, cytokines, histamine, spermine, lactate, and neuropeptides, capable of inhibiting or potentiating ASICs suggests the existence of mechanisms of synaptic modulation that have not yet been fully identified and that could be tuned by new emerging pharmacological compounds with potential therapeutic benefits.
Collapse
Affiliation(s)
- Osvaldo D Uchitel
- Departamento de Fisiología, Biología Molecular y Celular "Dr. Héctor Maldonado", Facultad de Ciencias Exactas y Naturales, Instituto de Fisiología, Biología molecular y Neurociencias (IFIBYNE) CONICET, Universidad de Buenos Aires, Ciudad Universitaria, (C1428EGA), Ciudad Autónoma de Buenos Aires, Argentina
| | - Carlota González Inchauspe
- Departamento de Fisiología, Biología Molecular y Celular "Dr. Héctor Maldonado", Facultad de Ciencias Exactas y Naturales, Instituto de Fisiología, Biología molecular y Neurociencias (IFIBYNE) CONICET, Universidad de Buenos Aires, Ciudad Universitaria, (C1428EGA), Ciudad Autónoma de Buenos Aires, Argentina
| | - Carina Weissmann
- Departamento de Fisiología, Biología Molecular y Celular "Dr. Héctor Maldonado", Facultad de Ciencias Exactas y Naturales, Instituto de Fisiología, Biología molecular y Neurociencias (IFIBYNE) CONICET, Universidad de Buenos Aires, Ciudad Universitaria, (C1428EGA), Ciudad Autónoma de Buenos Aires, Argentina
| |
Collapse
|
95
|
Moffitt AD, Brignolo LL, Ardeshir A, Creamer-Hente MA. The Role of Emotional Contagion in the Distress Exhibited by Grouped Mice Exposed to CO₂. JOURNAL OF THE AMERICAN ASSOCIATION FOR LABORATORY ANIMAL SCIENCE 2019; 58:430-437. [PMID: 31266585 DOI: 10.30802/aalas-jaalas-18-000098] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The 2013 AVMA Guidelines for the Euthanasia of Animals recommends a chamber volume displacement rate of 10% to 30% per minute (v/min) when euthanizing small laboratory rodents with CO₂. Group euthanasia of mice is a common practice, and grouping strangers is often avoided to minimize distress; however, emotional contagion, which occurs between familiar animals but not strangers, has not been studied in the context of group CO₂ euthanasia. This study examined cagemate- and stranger-grouped mice exposed to 10%, 30%, or 50% v/min CO₂ to determine whether emotional contagion plays a role in this context and whether that role is influenced by CO₂ flow rate. Videos of adult male C57BL/6J mice exposed to different CO₂ flow rates were scored for durations of dyspnea, ataxia, and consciousness as well as the numbers of face pawing and jump behaviors. Blood was collected at time of unconsciousness and assayed for ACTH. Cagemates experienced significantly longer durations of conscious dyspnea and ataxia with 10% v/min CO₂ compared with 30% and 50% v/min. Similarly, strangers experienced significantly longer duration of conscious dyspnea with 10% v/min CO₂ compared with 30% and 50% v/min and significantly longer duration of ataxia with 10% compared with 50% v/min. Cagemates showed significantly more jumps with 10% v/min CO₂ compared with 30% and 50% v/min, whereas jumping was unaffected by CO₂ flow rate in strangers. We conclude that more potential for distress exists when cagemate and stranger mice are exposed to a 10% v/min CO₂ flow rate and that emotional contagion may contribute to distress in cagemates at this flow rate. Therefore, we propose that 30% v/min CO₂ should be used for euthanasia of mice, and that 50% v/min should also be considered humane.
Collapse
Affiliation(s)
- Andrea D Moffitt
- Campus Veterinary Services, University of California, Davis, California;,
| | - Laurie L Brignolo
- Campus Veterinary Services, University of California, Davis, California
| | - Amir Ardeshir
- California National Primate Research Center, University of California, Davis, California
| | | |
Collapse
|
96
|
Van Diest I. Interoception, conditioning, and fear: The panic threesome. Psychophysiology 2019; 56:e13421. [DOI: 10.1111/psyp.13421] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 04/11/2019] [Accepted: 05/16/2019] [Indexed: 12/25/2022]
Affiliation(s)
- Ilse Van Diest
- Health, Behavior & Psychopathology, Faculty of Psychology & Educational Sciences; University of Leuven; Leuven Belgium
| |
Collapse
|
97
|
Winter A, Ahlbrand R, Sah R. Recruitment of central angiotensin II type 1 receptor associated neurocircuits in carbon dioxide associated fear. Prog Neuropsychopharmacol Biol Psychiatry 2019; 92:378-386. [PMID: 30776402 DOI: 10.1016/j.pnpbp.2019.02.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 01/15/2019] [Accepted: 02/14/2019] [Indexed: 11/29/2022]
Abstract
Individuals with fear-associated conditions such as panic disorder (PD) and posttraumatic stress disorder (PTSD) display increased emotional responses to interoceptive triggers, such as CO2 inhalation, that signal a threat to physiological homeostasis. Currently, effector systems and mechanisms underlying homeostatic modulation of fear memory are not well understood. In this regard, the renin angiotensin system (RAS), particularly the angiotensin receptor type 1 (AT1R), a primary homeostatic regulatory target, has gained attention. RAS polymorphisms have been reported in PD and PTSD, and recent studies report AT1R-mediated modulation of fear extinction. However, contribution of AT1Rs in fear evoked by the interoceptive threat of CO2 has not been investigated. Using pharmacological, behavioral, and AT1R/ACE gene transcription analyses, we assessed central AT1R recruitment in CO2-associated fear. CO2 inhalation led to significant AT1R and ACE mRNA upregulation in homeostatic regulatory regions, subfornical organ (SFO) and paraventricular nucleus (PVN), in a temporal manner. Intracerebroventricular infusion of selective AT1R antagonist, losartan, significantly attenuated freezing during CO2 inhalation, and during re-exposure to CO2 context, suggestive of AT1R modulation of contextual fear. Regional Fos mapping in losartan-treated mice post-behavior revealed significantly attenuated labeling in areas regulating defensive behavior, contextual fear, and threat responding; such as, the bed nucleus of stria terminalis, dorsal periaqueductal gray, hypothalamic nuclei, hippocampus, and prefrontal areas such as the prelimbic, infralimbic, and anterior cingulate cortices. Sub-regions of the amygdala did not show CO2-associated AT1R regulation or altered Fos labeling. Collectively, our data suggests central AT1R recruitment in modulation of fear behaviors associated with CO2 inhalation via engagement of neurocircuits regulating homeostasis and defensive behaviors. Our data provides mechanistic insights into the interoceptive regulation of fear, relevant to fear related disorders such as PD and PTSD.
Collapse
Affiliation(s)
- Andrew Winter
- Dept. of Pharmacology and Systems Physiology, University of Cincinnati, United States; Neuroscience Graduate Program, University of Cincinnati, United States
| | - Rebecca Ahlbrand
- Dept. of Pharmacology and Systems Physiology, University of Cincinnati, United States; VA Medical Center, Cincinnati, OH, 45221, United States
| | - Renu Sah
- Dept. of Pharmacology and Systems Physiology, University of Cincinnati, United States; Neuroscience Graduate Program, University of Cincinnati, United States; VA Medical Center, Cincinnati, OH, 45221, United States.
| |
Collapse
|
98
|
Weston CSE. Four Social Brain Regions, Their Dysfunctions, and Sequelae, Extensively Explain Autism Spectrum Disorder Symptomatology. Brain Sci 2019; 9:E130. [PMID: 31167459 PMCID: PMC6627615 DOI: 10.3390/brainsci9060130] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 05/31/2019] [Accepted: 06/03/2019] [Indexed: 12/13/2022] Open
Abstract
Autism spectrum disorder (ASD) is a challenging neurodevelopmental disorder with symptoms in social, language, sensory, motor, cognitive, emotional, repetitive behavior, and self-sufficient living domains. The important research question examined is the elucidation of the pathogenic neurocircuitry that underlies ASD symptomatology in all its richness and heterogeneity. The presented model builds on earlier social brain research, and hypothesizes that four social brain regions largely drive ASD symptomatology: amygdala, orbitofrontal cortex (OFC), temporoparietal cortex (TPC), and insula. The amygdala's contributions to ASD largely derive from its major involvement in fine-grained intangible knowledge representations and high-level guidance of gaze. In addition, disrupted brain regions can drive disturbance of strongly interconnected brain regions to produce further symptoms. These and related effects are proposed to underlie abnormalities of the visual cortex, inferior frontal gyrus (IFG), caudate nucleus, and hippocampus as well as associated symptoms. The model is supported by neuroimaging, neuropsychological, neuroanatomical, cellular, physiological, and behavioral evidence. Collectively, the model proposes a novel, parsimonious, and empirically testable account of the pathogenic neurocircuitry of ASD, an extensive account of its symptomatology, a novel physiological biomarker with potential for earlier diagnosis, and novel experiments to further elucidate the mechanisms of brain abnormalities and symptomatology in ASD.
Collapse
|
99
|
Martinez VK, Saldana-Morales F, Sun JJ, Zhu PJ, Costa-Mattioli M, Ray RS. Off-Target Effects of Clozapine-N-Oxide on the Chemosensory Reflex Are Masked by High Stress Levels. Front Physiol 2019; 10:521. [PMID: 31178741 PMCID: PMC6538678 DOI: 10.3389/fphys.2019.00521] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 04/11/2019] [Indexed: 12/28/2022] Open
Abstract
Respiratory chemosensory circuits are implicated in several physiological and behavioral disorders ranging from sudden infant death syndrome to panic disorder. Thus, a comprehensive map of the chemosensory network would be of significant value. To delineate chemosensory neuronal populations, we have utilized pharmacogenetic Designer Receptors Exclusively Activated by Designer Drugs (DREADD) perturbations for acute neuronal perturbations in respiratory circuit mapping. Recent studies show that the biologically inert DREADD ligand clozapine-N-oxide (CNO) is back-metabolized into the bioactive compound clozapine in rodents, emphasizing the need for CNO-only DREADD-free controls, which have been carried out in several studies. However, we show that high CNO doses used in several chemosensory circuit mapping studies nonetheless affect the chemosensory ventilatory reflexes in control mice, which is unmasked by extensive habituation. Here, unhabituated control animals showed no differences in respiratory parameters after CNO administration, whereas habituated animals receiving the commonly used dose of 10 mg/kg of CNO show a deficit in the hypercapnic (high CO2) chemosensory reflex, which is not present in 1 mg/kg CNO treated or saline control groups. Our findings indicate that even in appropriately controlled studies, additional masked CNO off-target effects may exist and underscore the importance of using minimal doses of activating ligand in combination with high levels of habituation.
Collapse
Affiliation(s)
- Vena K Martinez
- Department of Pharmacology, Baylor College of Medicine, Houston, TX, United States.,Memory Brain Research Center, Baylor College of Medicine, Houston, TX, United States
| | - Fatima Saldana-Morales
- Memory Brain Research Center, Baylor College of Medicine, Houston, TX, United States.,Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Jenny J Sun
- Memory Brain Research Center, Baylor College of Medicine, Houston, TX, United States.,Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Ping Jun Zhu
- Memory Brain Research Center, Baylor College of Medicine, Houston, TX, United States.,Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Mauro Costa-Mattioli
- Memory Brain Research Center, Baylor College of Medicine, Houston, TX, United States.,Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Russell S Ray
- Memory Brain Research Center, Baylor College of Medicine, Houston, TX, United States.,Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States.,McNair Medical Institute, Houston, TX, United States
| |
Collapse
|
100
|
Gent TC, Vyssotski AL, Detotto C, Isler S, Wehrle M, Bettschart-Wolfensberger R. Is xenon a suitable euthanasia agent for mice? Vet Anaesth Analg 2019; 46:652-657. [PMID: 31151872 DOI: 10.1016/j.vaa.2019.04.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 03/28/2019] [Accepted: 04/03/2019] [Indexed: 10/27/2022]
Abstract
OBJECTIVE To compare behavioural and electrophysiological variables of mice undergoing gas euthanasia with either xenon (Xe) or carbon dioxide (CO2). STUDY DESIGN Single animals chronically instrumented for electroencephalography (EEG) recording were randomized to undergo euthanasia with either CO2 or Xe (n = 6 animals per group). ANIMALS Twelve adult (>6 weeks old) male C57Bl6/n mice. METHODS Mice were surgically instrumented with EEG and electromyogram electrodes. Following a 7-day recovery period, animals were placed individually in a sealed chamber and a 5-minute baseline recorded in 21% O2. Gas [100% Xe (n = 6) or 100% CO2 (n = 6)] was then added to the chamber at 30% chamber volume minute-1 (2.8 L minute-1) until cessation of breathing. EEG, behaviour (jumping and freezing) and locomotion speed were recorded throughout. RESULTS Mice undergoing single gas euthanasia with Xe did not show jumping or freezing behaviours and had reduced locomotion speed compared to baseline, in contrast to CO2, which resulted in increases in these variables. EEG recordings revealed sedative effects from Xe but heightened arousal from CO2. CONCLUSIONS Our data suggest that Xe may be less aversive than CO2 when using a 30% chamber volume minute-1 fill rate and could improve the welfare of mice undergoing gas euthanasia.
Collapse
Affiliation(s)
- Thomas C Gent
- Section of Anaesthesiology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland.
| | - Alexei L Vyssotski
- Institute for Neuroinformatics, University of Zürich and ETH Zurich, Zurich, Switzerland
| | - Carlotta Detotto
- Section of Anaesthesiology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Sarah Isler
- Natur- und Tierpark Goldau, Goldau, Switzerland
| | | | | |
Collapse
|