1
|
Bastianini S, Alvente S, Berteotti C, Lo Martire V, Matteoli G, Miglioranza E, Silvani A, Zoccoli G. Ageing-related modification of sleep and breathing in orexin-knockout narcoleptic mice. J Sleep Res 2025; 34:e14287. [PMID: 39032099 PMCID: PMC11911059 DOI: 10.1111/jsr.14287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 05/27/2024] [Accepted: 07/08/2024] [Indexed: 07/22/2024]
Abstract
Narcolepsy type-1 (NT1) is a lifelong sleep disease, characterised by impairment of the orexinergic system, with a typical onset during adolescence and young adulthood. Since the wake-sleep cycle physiologically changes with ageing, this study aims to compare sleep patterns between orexin-knockout (KO) and wild type (WT) control mice at different ages. Four groups of age-matched female KO and WT mice (16 weeks of age: 8 KO-YO and 9 WT-YO mice; 87 weeks of age: 13 KO-OLD and 12 WT-OLD mice) were implanted with electrodes for discriminating wakefulness, rapid-eye-movement sleep (REMS), and non-REMS (NREMS). Mice were recorded for 48 h in their home cages and for 7 more hours into a plethysmographic chamber to characterise their sleep-breathing pattern. Regardless of orexin deficiency, OLD mice spent less time awake and had fragmentation of this behavioural state showing more bouts of shorter length than YO mice. OLD mice also had more NREMS bouts and less frequent NREMS apneas than YO mice. Regardless of age, KO mice showed cataplexy-like episodes and shorter REMS latency than WT controls and had a faster breathing rate and an increased minute ventilation during REMS. KO mice also had more wakefulness, NREMS and REMS bouts, and a shorter mean length of wakefulness bouts than WT controls. Our experiment indicated that the lack of orexins as well as ageing importantly modulate the sleep and breathing phenotype in mice. The narcoleptic phenotype caused by orexin deficiency in female mice was substantially preserved with ageing.
Collapse
Affiliation(s)
- Stefano Bastianini
- Laboratory of Physiological Regulation in Sleeping Mice (PRISM), Department of Biomedical and Neuromotor SciencesAlma Mater Studiorum – University of BolognaBolognaItaly
| | - Sara Alvente
- Laboratory of Physiological Regulation in Sleeping Mice (PRISM), Department of Biomedical and Neuromotor SciencesAlma Mater Studiorum – University of BolognaBolognaItaly
| | - Chiara Berteotti
- Laboratory of Physiological Regulation in Sleeping Mice (PRISM), Department of Biomedical and Neuromotor SciencesAlma Mater Studiorum – University of BolognaBolognaItaly
| | - Viviana Lo Martire
- Laboratory of Physiological Regulation in Sleeping Mice (PRISM), Department of Biomedical and Neuromotor SciencesAlma Mater Studiorum – University of BolognaBolognaItaly
| | - Gabriele Matteoli
- Laboratory of Physiological Regulation in Sleeping Mice (PRISM), Department of Biomedical and Neuromotor SciencesAlma Mater Studiorum – University of BolognaBolognaItaly
| | - Elena Miglioranza
- Laboratory of Physiological Regulation in Sleeping Mice (PRISM), Department of Biomedical and Neuromotor SciencesAlma Mater Studiorum – University of BolognaBolognaItaly
| | - Alessandro Silvani
- Laboratory of Physiological Regulation in Sleeping Mice (PRISM), Department of Biomedical and Neuromotor SciencesAlma Mater Studiorum – University of BolognaBolognaItaly
| | - Giovanna Zoccoli
- Laboratory of Physiological Regulation in Sleeping Mice (PRISM), Department of Biomedical and Neuromotor SciencesAlma Mater Studiorum – University of BolognaBolognaItaly
| |
Collapse
|
2
|
Dereli AS, Apaire A, El Tahry R. Sudden Unexpected Death in Epilepsy: Central Respiratory Chemoreception. Int J Mol Sci 2025; 26:1598. [PMID: 40004062 PMCID: PMC11855741 DOI: 10.3390/ijms26041598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 02/03/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
Sudden unexpected death in epilepsy (SUDEP) is a critical concern for individuals suffering from epilepsy, with respiratory dysfunction playing a significant role in its pathology. Fatal seizures are often characterized by central apnea and hypercapnia (elevated CO2 levels), indicating a failure in ventilatory control. Research has shown that both human epilepsy patients and animal models exhibit a reduced hypercapnic ventilatory response in the interictal (non-seizure) period, suggesting an impaired ability to regulate breathing in response to high CO2 levels. This review examines the role of central chemoreceptors-specifically the retrotrapezoid nucleus, raphe nuclei, nucleus tractus solitarius, locus coeruleus, and hypothalamus in this pathology. These structures are critical for sensing CO2 and maintaining respiratory homeostasis. Emerging evidence also implicates neuropeptidergic pathways within these chemoreceptive regions in SUDEP. Neuropeptides like galanin, pituitary adenylate cyclase-activating peptide (PACAP), orexin, somatostatin, and bombesin-like peptides may modulate chemosensitivity and respiratory function, potentially exacerbating respiratory failure during seizures. Understanding the mechanisms linking central chemoreception, respiratory control, and neuropeptidergic signaling is essential to developing targeted interventions to reduce the risk of SUDEP in epilepsy patients.
Collapse
Affiliation(s)
- Ayse S. Dereli
- Clinical Neuroscience, Institute of Neuroscience (IoNS), Université Catholique de Louvain, 1200 Brussels, Belgium; (A.A.); (R.E.T.)
| | - Auriane Apaire
- Clinical Neuroscience, Institute of Neuroscience (IoNS), Université Catholique de Louvain, 1200 Brussels, Belgium; (A.A.); (R.E.T.)
- Walloon Excellence in Life Sciences and Biotechnology (WELBIO), WEL Research Institute, 1300 Wavre, Belgium
| | - Riem El Tahry
- Clinical Neuroscience, Institute of Neuroscience (IoNS), Université Catholique de Louvain, 1200 Brussels, Belgium; (A.A.); (R.E.T.)
- Walloon Excellence in Life Sciences and Biotechnology (WELBIO), WEL Research Institute, 1300 Wavre, Belgium
- Center for Refractory Epilepsy, Department of Neurology, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| |
Collapse
|
3
|
Iyer SH, Hinman JE, Warren T, Matthews SA, Simeone TA, Simeone KA. Altered ventilatory responses to hypercapnia-hypoxia challenges in a preclinical SUDEP model involve orexin neurons. Neurobiol Dis 2024; 199:106592. [PMID: 38971479 PMCID: PMC11648317 DOI: 10.1016/j.nbd.2024.106592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 06/25/2024] [Accepted: 07/02/2024] [Indexed: 07/08/2024] Open
Abstract
Failure to recover from repeated hypercapnia and hypoxemia (HH) challenges caused by severe GCS and postictal apneas may contribute to sudden unexpected death in epilepsy (SUDEP). Our previous studies found orexinergic dysfunction contributes to respiratory abnormalities in a preclinical model of SUDEP, Kcna1-/- mice. Here, we developed two gas challenges consisting of repeated HH exposures and used whole body plethysmography to determine whether Kcna1-/- mice have detrimental ventilatory responses. Kcna1-/- mice exhibited an elevated ventilatory response to a mild repeated hypercapnia-hypoxia (HH) challenge compared to WT. Moreover, 71% of Kcna1-/- mice failed to survive a severe repeated HH challenge, whereas all WT mice recovered. We next determined whether orexin was involved in these differences. Pretreating Kcna1-/- mice with a dual orexin receptor antagonist rescued the ventilatory response during the mild challenge and all subjects survived the severe challenge. In ex vivo extracellular recordings in the lateral hypothalamus of coronal brain slices, we found reducing pH either inhibits or stimulates putative orexin neurons similar to other chemosensitive neurons; however, a significantly greater percentage of putative orexin neurons from Kcna1-/-mice were stimulated and the magnitude of stimulation was increased resulting in augmentation of the calculated chemosensitivity index relative to WT. Collectively, our data suggest that increased chemosensitive activity of orexin neurons may be pathologic in the Kcna1-/- mouse model of SUDEP, and contribute to elevated ventilatory responses. Our preclinical data suggest that those at high risk for SUDEP may be more sensitive to HH challenges, whether induced by seizures or other means; and the depth and length of the HH exposure could dictate the probability of survival.
Collapse
Affiliation(s)
- Shruthi H Iyer
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE 68178, USA
| | - Jillian E Hinman
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE 68178, USA
| | - Ted Warren
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE 68178, USA
| | - Stephanie A Matthews
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE 68178, USA
| | - Timothy A Simeone
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE 68178, USA
| | - Kristina A Simeone
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE 68178, USA.
| |
Collapse
|
4
|
Kukkonen JP, Jacobson LH, Hoyer D, Rinne MK, Borgland SL. International Union of Basic and Clinical Pharmacology CXIV: Orexin Receptor Function, Nomenclature and Pharmacology. Pharmacol Rev 2024; 76:625-688. [PMID: 38902035 DOI: 10.1124/pharmrev.123.000953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 06/02/2024] [Accepted: 06/06/2024] [Indexed: 06/22/2024] Open
Abstract
The orexin system consists of the peptide transmitters orexin-A and -B and the G protein-coupled orexin receptors OX1 and OX2 Orexin receptors are capable of coupling to all four families of heterotrimeric G proteins, and there are also other complex features of the orexin receptor signaling. The system was discovered 25 years ago and was immediately identified as a central regulator of sleep and wakefulness; this is exemplified by the symptomatology of the disorder narcolepsy with cataplexy, in which orexinergic neurons degenerate. Subsequent translation of these findings into drug discovery and development has resulted to date in three clinically used orexin receptor antagonists to treat insomnia. In addition to sleep and wakefulness, the orexin system appears to be a central player at least in addiction and reward, and has a role in depression, anxiety and pain gating. Additional antagonists and agonists are in development to treat, for instance, insomnia, narcolepsy with or without cataplexy and other disorders with excessive daytime sleepiness, depression with insomnia, anxiety, schizophrenia, as well as eating and substance use disorders. The orexin system has thus proved an important regulator of numerous neural functions and a valuable drug target. Orexin prepro-peptide and orexin receptors are also expressed outside the central nervous system, but their potential physiological roles there remain unknown. SIGNIFICANCE STATEMENT: The orexin system was discovered 25 years ago and immediately emerged as an essential sleep-wakefulness regulator. This discovery has tremendously increased the understanding of these processes and has thus far resulted in the market approval of three orexin receptor antagonists, which promote more physiological aspects of sleep than previous hypnotics. Further, orexin receptor agonists and antagonists with different pharmacodynamic properties are in development since research has revealed additional potential therapeutic indications. Orexin receptor signaling is complex and may represent novel features.
Collapse
Affiliation(s)
- Jyrki P Kukkonen
- Department of Pharmacology, Medicum, University of Helsinki, Helsinki, Finland (J.P.K., M.K.R.); Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne (D.H., L.H.J.), The Florey (D.H., L.H.J.), Parkville, Victoria, Australia; Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California (D.H.); and Department of Physiology and Pharmacology, University of Calgary, Calgary Canada (S.L.B.)
| | - Laura H Jacobson
- Department of Pharmacology, Medicum, University of Helsinki, Helsinki, Finland (J.P.K., M.K.R.); Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne (D.H., L.H.J.), The Florey (D.H., L.H.J.), Parkville, Victoria, Australia; Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California (D.H.); and Department of Physiology and Pharmacology, University of Calgary, Calgary Canada (S.L.B.)
| | - Daniel Hoyer
- Department of Pharmacology, Medicum, University of Helsinki, Helsinki, Finland (J.P.K., M.K.R.); Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne (D.H., L.H.J.), The Florey (D.H., L.H.J.), Parkville, Victoria, Australia; Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California (D.H.); and Department of Physiology and Pharmacology, University of Calgary, Calgary Canada (S.L.B.)
| | - Maiju K Rinne
- Department of Pharmacology, Medicum, University of Helsinki, Helsinki, Finland (J.P.K., M.K.R.); Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne (D.H., L.H.J.), The Florey (D.H., L.H.J.), Parkville, Victoria, Australia; Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California (D.H.); and Department of Physiology and Pharmacology, University of Calgary, Calgary Canada (S.L.B.)
| | - Stephanie L Borgland
- Department of Pharmacology, Medicum, University of Helsinki, Helsinki, Finland (J.P.K., M.K.R.); Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne (D.H., L.H.J.), The Florey (D.H., L.H.J.), Parkville, Victoria, Australia; Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California (D.H.); and Department of Physiology and Pharmacology, University of Calgary, Calgary Canada (S.L.B.)
| |
Collapse
|
5
|
Ben Musa R, Cornelius-Green J, Zhang H, Li DP, Kline DD, Hasser EM, Cummings KJ. Orexin Facilitates the Peripheral Chemoreflex via Corticotropin-Releasing Hormone Neurons Projecting to the Nucleus of the Solitary Tract. J Neurosci 2024; 44:e2383232024. [PMID: 38789262 PMCID: PMC11223477 DOI: 10.1523/jneurosci.2383-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 04/14/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024] Open
Abstract
We previously showed that orexin neurons are activated by hypoxia and facilitate the peripheral chemoreflex (PCR)-mediated hypoxic ventilatory response (HVR), mostly by promoting the respiratory frequency response. Orexin neurons project to the nucleus of the solitary tract (nTS) and the paraventricular nucleus of the hypothalamus (PVN). The PVN contributes significantly to the PCR and contains nTS-projecting corticotropin-releasing hormone (CRH) neurons. We hypothesized that in male rats, orexin neurons contribute to the PCR by activating nTS-projecting CRH neurons. We used neuronal tract tracing and immunohistochemistry (IHC) to quantify the degree that hypoxia activates PVN-projecting orexin neurons. We coupled this with orexin receptor (OxR) blockade with suvorexant (Suvo, 20 mg/kg, i.p.) to assess the degree that orexin facilitates the hypoxia-induced activation of CRH neurons in the PVN, including those projecting to the nTS. In separate groups of rats, we measured the PCR following systemic orexin 1 receptor (Ox1R) blockade (SB-334867; 1 mg/kg) and specific Ox1R knockdown in PVN. OxR blockade with Suvo reduced the number of nTS and PVN neurons activated by hypoxia, including those CRH neurons projecting to nTS. Hypoxia increased the number of activated PVN-projecting orexin neurons but had no effect on the number of activated nTS-projecting orexin neurons. Global Ox1R blockade and partial Ox1R knockdown in the PVN significantly reduced the PCR. Ox1R knockdown also reduced the number of activated PVN neurons and the number of activated tyrosine hydroxylase neurons in the nTS. Our findings suggest orexin facilitates the PCR via nTS-projecting CRH neurons expressing Ox1R.
Collapse
Affiliation(s)
- Ruwaida Ben Musa
- Department of Biomedical Sciences, College of Veterinary Medicine, Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri 65211
| | - Jennifer Cornelius-Green
- Department of Biomedical Sciences, College of Veterinary Medicine, Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri 65211
| | - Hua Zhang
- Department of Medicine, Center for Precision Medicine, School of Medicine, University of Missouri, Columbia, Missouri 65212
| | - De-Pei Li
- Department of Medicine, Center for Precision Medicine, School of Medicine, University of Missouri, Columbia, Missouri 65212
| | - David D Kline
- Department of Biomedical Sciences, College of Veterinary Medicine, Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri 65211
| | - Eileen M Hasser
- Department of Biomedical Sciences, College of Veterinary Medicine, Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri 65211
| | - Kevin J Cummings
- Department of Biomedical Sciences, College of Veterinary Medicine, Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri 65211
| |
Collapse
|
6
|
Wang Y, Deng T, Zhao X, Shao L, Chen J, Fu C, He W, Wang X, Wang H, Yuan F, Wang S. Control of breathing by orexinergic signaling in the nucleus tractus solitarii. Sci Rep 2024; 14:7473. [PMID: 38553555 PMCID: PMC10980752 DOI: 10.1038/s41598-024-58075-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 03/25/2024] [Indexed: 04/02/2024] Open
Abstract
Orexin signaling plays a facilitatory role in respiration. Abnormalities in orexin levels correlate with disordered breathing patterns and impaired central respiratory chemoreception. Nucleus tractus solitarii (NTS) neurons expressing the transcription factor Phox2b contribute to the chemoreceptive regulation of respiration. However, the extent to which orexinergic signaling modulates respiratory activity in these Phox2b-expressing NTS neurons remains unclear. In the present study, the injection of orexin A into the NTS significantly increased the firing rate of the phrenic nerve. Further analysis using fluorescence in situ hybridization and immunohistochemistry revealed that orexin 1 receptors (OX1Rs) were primarily located in the ventrolateral subdivision of the NTS and expressed in 25% of Phox2b-expressing neurons. Additionally, electrophysiological recordings showed that exposure to orexin A increased the spontaneous firing rate of Phox2b-expressing neurons. Immunostaining experiments with cFos revealed that the OX1R-residing Phox2b-expressing neurons were activated by an 8% CO2 stimulus. Crucially, OX1R knockdown in these NTS neurons notably blunted the ventilatory response to 8% CO2, alongside an increase in sigh-related apneas. In conclusion, orexinergic signaling in the NTS facilitates breathing through the activation of OX1Rs, which induces the depolarization of Phox2b-expressing neurons. OX1Rs are essential for the involvement of Phox2b-expressing NTS neurons in the hypercapnic ventilatory response.
Collapse
Affiliation(s)
- Yakun Wang
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, Hebei, China
- Department of Sleep Medicine, Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Tianjiao Deng
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xue Zhao
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Liuqi Shao
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jinting Chen
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Congrui Fu
- Nursing School, Hebei Medical University, Shijiazhuang, China
| | - Wei He
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xiaoyi Wang
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Hanqiao Wang
- Department of Sleep Medicine, Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Fang Yuan
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Neurophysiology, Shijiazhuang, China
| | - Sheng Wang
- Department of Neurobiology, Hebei Medical University, Shijiazhuang, Hebei, China.
- Hebei Key Laboratory of Neurophysiology, Shijiazhuang, China.
| |
Collapse
|
7
|
Abstract
Substantial advances have been made recently into the discovery of fundamental mechanisms underlying the neural control of breathing and even some inroads into translating these findings to treating breathing disorders. Here, we review several of these advances, starting with an appreciation of the importance of V̇A:V̇CO2:PaCO2 relationships, then summarizing our current understanding of the mechanisms and neural pathways for central rhythm generation, chemoreception, exercise hyperpnea, plasticity, and sleep-state effects on ventilatory control. We apply these fundamental principles to consider the pathophysiology of ventilatory control attending hypersensitized chemoreception in select cardiorespiratory diseases, the pathogenesis of sleep-disordered breathing, and the exertional hyperventilation and dyspnea associated with aging and chronic diseases. These examples underscore the critical importance that many ventilatory control issues play in disease pathogenesis, diagnosis, and treatment.
Collapse
Affiliation(s)
- Jerome A Dempsey
- John Rankin Laboratory of Pulmonary Medicine, Department of Population Health Sciences, University of Wisconsin, Madison, Wisconsin
| | - Joseph F Welch
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| |
Collapse
|
8
|
Gonye EC, Bayliss DA. Criteria for central respiratory chemoreceptors: experimental evidence supporting current candidate cell groups. Front Physiol 2023; 14:1241662. [PMID: 37719465 PMCID: PMC10502317 DOI: 10.3389/fphys.2023.1241662] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 08/16/2023] [Indexed: 09/19/2023] Open
Abstract
An interoceptive homeostatic system monitors levels of CO2/H+ and provides a proportionate drive to respiratory control networks that adjust lung ventilation to maintain physiologically appropriate levels of CO2 and rapidly regulate tissue acid-base balance. It has long been suspected that the sensory cells responsible for the major CNS contribution to this so-called respiratory CO2/H+ chemoreception are located in the brainstem-but there is still substantial debate in the field as to which specific cells subserve the sensory function. Indeed, at the present time, several cell types have been championed as potential respiratory chemoreceptors, including neurons and astrocytes. In this review, we advance a set of criteria that are necessary and sufficient for definitive acceptance of any cell type as a respiratory chemoreceptor. We examine the extant evidence supporting consideration of the different putative chemoreceptor candidate cell types in the context of these criteria and also note for each where the criteria have not yet been fulfilled. By enumerating these specific criteria we hope to provide a useful heuristic that can be employed both to evaluate the various existing respiratory chemoreceptor candidates, and also to focus effort on specific experimental tests that can satisfy the remaining requirements for definitive acceptance.
Collapse
Affiliation(s)
- Elizabeth C. Gonye
- Department of Pharmacology, University of Virginia, Charlottesville, VA, United States
| | | |
Collapse
|
9
|
Kim LJ, Pho H, Anokye-Danso F, Ahima RS, Pham LV, Polotsky VY. The effect of diet-induced obesity on sleep and breathing in female mice. Sleep 2023; 46:zsad158. [PMID: 37262435 PMCID: PMC10424169 DOI: 10.1093/sleep/zsad158] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/24/2023] [Indexed: 06/03/2023] Open
Abstract
Obesity and male sex are main risk factors for sleep-disordered breathing (SDB). We have shown that male diet-induced obesity (DIO) mice develop hypoventilation, sleep apnea, and sleep fragmentation. The effects of DIO on breathing and sleep architecture in females have not been investigated. We hypothesized that female mice are less susceptible to the detrimental effects of DIO on sleep and SDB compared to males. Female DIO-C57BL/6J and lean C57BL/6J mice underwent 24-hour metabolic studies and were exposed to 8% CO2 to measure the hypercapnic ventilatory response (HCVR), and sleep studies. Ventilatory response to arousals was calculated as ratio of the average and peak minute ventilation (VE) during each arousal relative to the baseline VE. Breathing stability was measured with Poincaré plots of VE. Female obesity was associated with decreased metabolism, indicated by reduced oxygen consumption (VO2) and CO2 production (VCO2). VE in 8% CO2 and HCVR were significantly attenuated during wakefulness. NREM sleep duration was reduced in DIO mice, but REM sleep was preserved. Ventilation during NREM and REM sleep was augmented compared to lean mice. Arousal frequency was similar between groups. Obesity increased the frequency of spontaneous arousals, whereas the apnea index was 4-fold reduced in DIO compared to lean mice. Obesity decreased pre- and post-apnea arousals. Obese mice had more stable breathing with reduced ventilatory response to arousals, compared to lean females. We conclude that obese female mice are protected against SDB, which appears to be related to an attenuated CO2 responsiveness, compared to the lean state.
Collapse
Affiliation(s)
- Lenise J Kim
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Anesthesiology and Critical Care Medicine, School of Medicine and Health Sciences, George Washington University, Washington, DC, USA
| | - Huy Pho
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Frederick Anokye-Danso
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Rexford S Ahima
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Luu V Pham
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Vsevolod Y Polotsky
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Anesthesiology and Critical Care Medicine, School of Medicine and Health Sciences, George Washington University, Washington, DC, USA
- Department of Pharmacology and Physiology, School of Medicine and Health Sciences, George Washington University, Washington, DC, USA
| |
Collapse
|
10
|
Alvente S, Matteoli G, Miglioranza E, Zoccoli G, Bastianini S. How to study sleep apneas in mouse models of human pathology. J Neurosci Methods 2023; 395:109923. [PMID: 37459897 DOI: 10.1016/j.jneumeth.2023.109923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/28/2023] [Accepted: 07/13/2023] [Indexed: 07/22/2023]
Abstract
Sleep apnea, the most widespread sleep-related breathing disorder (SBD), consists of recurrent episodes of breathing cessation during sleep. This condition can be classified as either central (CSA) or obstructive (OSA) sleep apnea, with the latest being the most common and toxic. Due to the complexity of living organisms, animal models and, particularly, mice still represent an essential tool for the study of SBD. In the present review we first discuss the methodological pros and cons in the use of whole-body plethysmography to coupling respiratory and sleep measurements and to characterize CSA and OSA in mice; then, we draw an updated and objective picture of the methods used so far in the study of sleep apnea in mice. Most of the studies present in the literature used intermittent hypoxia to mimic OSA in mice and to investigate consequent pathological correlates. On the contrary, few studies using genetic manipulation or high-fat diets investigated the pathogenesis or potential treatments of sleep apnea. To date, mice lacking orexins, hemeoxygenase-2, monoamine oxidase A, Phox2b or Cdkl5 can be considered validated mouse models of sleep apnea. Moreover, genetically- or diet-induced obese mice, and mice recapitulating Down syndrome were proposed as OSA models. In conclusion, our review shows that despite the growing interest in the field and the need of new therapeutical approaches, technical complexity and inter-study variability strongly limit the availability of validated mouse of sleep apnea, which are essential in biomedical research.
Collapse
Affiliation(s)
- Sara Alvente
- PRISM Lab, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Gabriele Matteoli
- PRISM Lab, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Elena Miglioranza
- PRISM Lab, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Giovanna Zoccoli
- PRISM Lab, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Stefano Bastianini
- PRISM Lab, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy.
| |
Collapse
|
11
|
Kinkead R, Ambrozio-Marques D, Fournier S, Gagnon M, Guay LM. Estrogens, age, and, neonatal stress: panic disorders and novel views on the contribution of non-medullary structures to respiratory control and CO 2 responses. Front Physiol 2023; 14:1183933. [PMID: 37265841 PMCID: PMC10229816 DOI: 10.3389/fphys.2023.1183933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 04/21/2023] [Indexed: 06/03/2023] Open
Abstract
CO2 is a fundamental component of living matter. This chemical signal requires close monitoring to ensure proper match between metabolic production and elimination by lung ventilation. Besides ventilatory adjustments, CO2 can also trigger innate behavioral and physiological responses associated with fear and escape but the changes in brain CO2/pH required to induce ventilatory adjustments are generally lower than those evoking fear and escape. However, for patients suffering from panic disorder (PD), the thresholds for CO2-evoked hyperventilation, fear and escape are reduced and the magnitude of those reactions are excessive. To explain these clinical observations, Klein proposed the false suffocation alarm hypothesis which states that many spontaneous panics occur when the brain's suffocation monitor erroneously signals a lack of useful air, thereby maladaptively triggering an evolved suffocation alarm system. After 30 years of basic and clinical research, it is now well established that anomalies in respiratory control (including the CO2 sensing system) are key to PD. Here, we explore how a stress-related affective disorder such as PD can disrupt respiratory control. We discuss rodent models of PD as the concepts emerging from this research has influenced our comprehension of the CO2 chemosensitivity network, especially structure that are not located in the medulla, and how factors such as stress and biological sex modulate its functionality. Thus, elucidating why hormonal fluctuations can lead to excessive responsiveness to CO2 offers a unique opportunity to gain insights into the neuroendocrine mechanisms regulating this key aspect of respiratory control and the pathophysiology of respiratory manifestations of PD.
Collapse
|
12
|
Ben Musa R, Cornelius-Green J, Hasser EM, Cummings KJ. The effect of orexin on the hypoxic ventilatory response of female rats is greatest in the active phase during diestrus. J Appl Physiol (1985) 2023; 134:638-648. [PMID: 36656978 PMCID: PMC10010922 DOI: 10.1152/japplphysiol.00661.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 01/21/2023] Open
Abstract
We recently showed that in male rats, orexin contributes to the hypoxic ventilatory response (HVR), with a stronger effect in the active phase. The effect of orexin on the HVR in females has not been investigated. As estrogen can inhibit orexin neurons, here we hypothesized that orexin neurons are activated by hypoxia and facilitate the HVR only in diestrus, when estrogen is low. We exposed female rats (n = 10) to near-isocapnic hypoxia ([Formula: see text] from 0.21 to 0.09) over ∼5 min, after vehicle and again after suvorexant (a dual OxR antagonist; 20 mg/kg ip), with ventilation measured using whole body plethysmography. Each rat was tested in proestrus or estrus (p/estrus), and again in diestrus, during both inactive and active phases. We also performed immunohistochemistry (IHC) to determine the proportion of orexin neurons activated by acute hypoxia during diestrus (n = 6) or proestrus/estrus (n = 6) in the active phase. In the inactive phase, the HVR was unaffected by OxR blockade, irrespective of estrus stage. In the active phase, the effect of OxR blockade depended on stage: the slope of the HVR was significantly reduced by OxR blockade only during diestrus. IHC revealed that hypoxia activated more orexin neurons during diestrus compared with p/estrus. We conclude that in females, orexin neurons are activated by hypoxia and contribute to the HVR only in diestrus when estrogen levels are low. Stage of the estrus cycle should be considered when examining the physiological function of orexin neurons in females.NEW & NOTEWORTHY We previously showed that orexin facilitates the hypoxic ventilatory response (HVR) of adult male rats during the active phase. Others have shown that estrogen inhibits orexin neurons. Here we show that orexin neurons are activated by hypoxia and facilitate the HVR of adult female rats during the active phase, but only in diestrus. These data suggest that orexin neurons facilitate the HVR in females when they are free from the inhibitory effects of estrogen.
Collapse
Affiliation(s)
- Ruwaida Ben Musa
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, United States
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, United States
| | - Jennifer Cornelius-Green
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, United States
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, United States
| | - Eileen M Hasser
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, United States
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, United States
| | - Kevin J Cummings
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, United States
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, United States
| |
Collapse
|
13
|
Mogavero MP, Silvani A, Lanza G, DelRosso LM, Ferini-Strambi L, Ferri R. Targeting Orexin Receptors for the Treatment of Insomnia: From Physiological Mechanisms to Current Clinical Evidence and Recommendations. Nat Sci Sleep 2023; 15:17-38. [PMID: 36713640 PMCID: PMC9879039 DOI: 10.2147/nss.s201994] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 01/08/2023] [Indexed: 01/23/2023] Open
Abstract
After a detailed description of orexins and their roles in sleep and other medical disorders, we discuss here the current clinical evidence on the effects of dual (DORAs) or selective (SORAs) orexin receptor antagonists on insomnia with the aim to provide recommendations for their further assessment in a context of personalized and precision medicine. In the last decade, many trials have been conducted with orexin receptor antagonists, which represent an innovative and valid therapeutic option based on the multiple mechanisms of action of orexins on different biological circuits, both centrally and peripherally, and their role in a wide range of medical conditions which are often associated with insomnia. A very interesting aspect of this new category of drugs is that they have limited abuse liability and their discontinuation does not seem associated with significant rebound effects. Further studies on the efficacy of DORAs are required, especially on children and adolescents and in particular conditions, such as menopause. Which DORA is most suitable for each patient, based on comorbidities and/or concomitant treatments, should be the focus of further careful research. On the contrary, studies on SORAs, some of which seem to be appropriate also in insomnia in patients with psychiatric diseases, are still at an early stage and, therefore, do not allow to draw definite conclusions.
Collapse
Affiliation(s)
- Maria P Mogavero
- Vita-Salute San Raffaele University, Milan, Italy
- Sleep Disorders Center, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Alessandro Silvani
- PRISM Lab, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Giuseppe Lanza
- Sleep Research Centre, Oasi Research Institute - IRCCS, Troina, Italy
- Department of Surgery and Medical-Surgical Specialties, University of Catania, Catania, Italy
| | - Lourdes M DelRosso
- Pulmonary and Sleep Medicine, University of California San Francisco-Fresno, Fresno, CA, USA
| | - Luigi Ferini-Strambi
- Vita-Salute San Raffaele University, Milan, Italy
- Sleep Disorders Center, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Raffaele Ferri
- Sleep Research Centre, Oasi Research Institute - IRCCS, Troina, Italy
| |
Collapse
|
14
|
Sithirungson S, Sonsuwan N, Chattipakorn SC, Chattipakorn N, Shinlapawittayatorn K. Functional roles of orexin in obstructive sleep apnea: From clinical observation to mechanistic insights. Sleep Med 2023; 101:40-49. [PMID: 36334500 DOI: 10.1016/j.sleep.2022.10.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 09/23/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022]
Abstract
Obstructive sleep apnea is the most common sleep-related breathing disorder. Repetitive episodes of the obstructive respiratory events lead to arousal, sleep fragmentation, and excessive daytime sleepiness. Orexin, also known as hypocretin, is one of the most important neurotransmitters responsible for sleep and arousal regulation. Deficiency of orexin has been shown to be involved in the pathogenesis of narcolepsy, which shares cardinal symptoms of sleep apnea and excessive daytime sleep with obstructive sleep apnea. However, the relationship between orexin and obstructive sleep apnea is not well defined. In this review, we summarize the current evidence, from in vitro, in vivo, and clinical data, regarding the association between orexin and obstructive sleep apnea. The effects of orexin on sleep apnea, as well as how the consequences of obstructive sleep apnea affect the orexin system function are also discussed. Additionally, the contrary findings are also included and discussed.
Collapse
Affiliation(s)
- Suchanya Sithirungson
- Department of Otolaryngology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Nuntigar Sonsuwan
- Department of Otolaryngology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Siriporn C Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Krekwit Shinlapawittayatorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.
| |
Collapse
|
15
|
Varga AG, Whitaker-Fornek JR, Maletz SN, Levitt ES. Activation of orexin-2 receptors in the Kӧlliker-Fuse nucleus of anesthetized mice leads to transient slowing of respiratory rate. Front Physiol 2022; 13:977569. [PMID: 36406987 PMCID: PMC9667107 DOI: 10.3389/fphys.2022.977569] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 10/11/2022] [Indexed: 11/05/2022] Open
Abstract
Orexins are neuropeptides originating from the hypothalamus that serve broad physiological roles, including the regulation of autonomic function, sleep-wake states, arousal and breathing. Lack of orexins may lead to narcolepsy and sleep disordered breathing. Orexinergic hypothalamic neurons send fibers to Kӧlliker-Fuse (KF) neurons that directly project to the rostroventral respiratory group, and phrenic and hypoglossal motor neurons. These connections indicate a potential role of orexin-modulated KF neurons in functionally linking the control of wakefulness/arousal and respiration. In a reduced preparation of juvenile rats Orexin B microinjected into the KF led to a transient increase in respiratory rate and hypoglossal output, however Orexin B modulation of the KF in intact preparations has not been explored. Here, we performed microinjections of the Orexin B mouse peptide and the synthetic Orexin 2 receptor agonist, MDK 5220, in the KF of spontaneously breathing, isoflurane anesthetized wild type mice. Microinjection of Orexin-2 receptor agonists into the KF led to transient slowing of respiratory rate, which was more exaggerated in response to Orexin-B than MDK 5220 injections. Our data suggest that Orexin B signaling in the KF may contribute to arousal-mediated respiratory responses.
Collapse
Affiliation(s)
- Adrienn G. Varga
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, United States
- Breathing Research and Therapeutics Center, University of Florida, Gainesville, FL, United States
| | - Jessica R. Whitaker-Fornek
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, United States
- Breathing Research and Therapeutics Center, University of Florida, Gainesville, FL, United States
| | - Sebastian N. Maletz
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, United States
| | - Erica S. Levitt
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, United States
- Breathing Research and Therapeutics Center, University of Florida, Gainesville, FL, United States
| |
Collapse
|
16
|
CO 2 exposure enhances Fos expression in hypothalamic neurons in rats during the light and dark phases of the diurnal cycle. Brain Struct Funct 2022; 227:2667-2679. [PMID: 36109371 DOI: 10.1007/s00429-022-02562-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 08/29/2022] [Indexed: 12/30/2022]
Abstract
Orexinergic (OX) neurons in the lateral hypothalamus (LH), perifornical area (PFA) and dorsomedial hypothalamus (DMH) play a role in the hypercapnic ventilatory response, presumably through direct inputs to central pattern generator sites and/or through interactions with other chemosensitive regions. OX neurons can produce and release orexins, excitatory neuropeptides involved in many functions, including physiological responses to changes in CO2/pH. Thus, in the present study, we tested the hypothesis that different nuclei (LH, PFA and DMH) where the orexinergic neurons are located, show distinct activation by CO2 during the light-dark cycle phases. For this purpose, we evaluated the Fos and OXA expression by immunohistochemistry to identify neurons that co-localize Fos + OXA in the LH, LPeF, MPeF and DMH in the light-inactive and dark-active phase in Wistar rats subjected to 3 h of normocapnia or hypercapnia (7% CO2). Quantitative analyses of immunoreactive neurons show that hypercapnia caused an increase in the number of neurons expressing Fos in the LH, LPeF, MPeF and DMH in the light and dark phases. In addition, the number of Fos + OXA neurons increased in the LPeF and DMH independently of the phases of the diurnal cycle; whereas in the MPeF, this increase was observed exclusively in the light phase. Thus, we suggest that OX neurons are selectively activated by hypercapnia throughout the diurnal cycle, reinforcing the differential role of nuclei in the hypothalamus during central chemosensitivity.
Collapse
|
17
|
Amorim MR, Aung O, Mokhlesi B, Polotsky VY. Leptin-mediated neural targets in obesity hypoventilation syndrome. Sleep 2022; 45:zsac153. [PMID: 35778900 PMCID: PMC9453616 DOI: 10.1093/sleep/zsac153] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 06/20/2022] [Indexed: 07/30/2023] Open
Abstract
Obesity hypoventilation syndrome (OHS) is defined as daytime hypercapnia in obese individuals in the absence of other underlying causes. In the United States, OHS is present in 10%-20% of obese patients with obstructive sleep apnea and is linked to hypoventilation during sleep. OHS leads to high cardiorespiratory morbidity and mortality, and there is no effective pharmacotherapy. The depressed hypercapnic ventilatory response plays a key role in OHS. The pathogenesis of OHS has been linked to resistance to an adipocyte-produced hormone, leptin, a major regulator of metabolism and control of breathing. Mechanisms by which leptin modulates the control of breathing are potential targets for novel therapeutic strategies in OHS. Recent advances shed light on the molecular pathways related to the central chemoreceptor function in health and disease. Leptin signaling in the nucleus of the solitary tract, retrotrapezoid nucleus, hypoglossal nucleus, and dorsomedial hypothalamus, and anatomical projections from these nuclei to the respiratory control centers, may contribute to OHS. In this review, we describe current views on leptin-mediated mechanisms that regulate breathing and CO2 homeostasis with a focus on potential therapeutics for the treatment of OHS.
Collapse
Affiliation(s)
- Mateus R Amorim
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - O Aung
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Babak Mokhlesi
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Vsevolod Y Polotsky
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
18
|
Fukushi I, Yokota S, Takeda K, Terada J, Umeda A, Yoshizawa M, Kono Y, Hasebe Y, Onimaru H, Pokorski M, Okada Y. Dual orexin receptor blocker suvorexant attenuates hypercapnic ventilatory augmentation in mice. Brain Res 2022; 1795:148061. [PMID: 36037880 DOI: 10.1016/j.brainres.2022.148061] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/14/2022] [Accepted: 08/23/2022] [Indexed: 11/17/2022]
Abstract
Suvorexant (Belsomra(R)), a dual orexin receptor antagonist widely used in the treatment of insomnia, inhibits the arousal system in the brain. However, the drug's ventilatory effects have not been fully explored. This study aims to investigate the expression of orexin receptors in respiratory neurons and the effects of suvorexant on ventilation. Immunohistology of brainstem orexin receptor OX2R expression was performed in adult mice (n=4) in (1) rostral ventral respiratory group (rVRG) neurons projecting to the phrenic nucleus (PhN) retrogradely labeled by Fluoro-Gold (FG) tracer, (2) neurons immunoreactive for paired like homeobox 2b (Phox2b) in the parafacial respiratory group/retrotrapezoid nucleus (pFRG/RTN), and (3) neurons immunoreactive for neurokinin 1 receptor (NK1R) and somatostatin (SST) in the preBötzinger complex (preBötC). Additionally, we measured in vivo ventilatory responses to hyperoxic hypercapnia (5% CO2) and hypoxia (10% O2) before and after suvorexant pretreatment (10 and cumulative 100 mg/kg) in unrestrained mice (n=10) in a body plethysmograph. We found the OX2R immunoreactive materials in pFRG/RTN Phox2b and preBötC NK1R/SST immunoreactive neurons but not in FG-labeled rVRG neurons, which suggests the involvement of orexin in respiratory control. Further, suvorexant expressly suppressed the hypercapnic ventilatory augmentation, otherwise unaffecting ventilation. Central orexin is involved in shaping the hypercapnic ventilatory chemosensitivity. Suppression of hypercapnic ventilatory augmentation by the orexin receptor antagonist suvorexant calls for caution in its use in pathologies that may progress to hypercapnic respiratory failure, or sleep-disordered breathing. Clinical trials are required to explore the role of targeted pharmacological inhibition of orexin in ventilatory pathologies.
Collapse
Affiliation(s)
- Isato Fukushi
- Faculty of Health Sciences, Aomori University of Health and Welfare, Aomori, Japan; Clinical Research Center, Murayama Medical Center, Musashimurayama, Japan.
| | - Shigefumi Yokota
- Department of Anatomy and Neuroscience, Shimane University School of Medicine, Izumo, Japan
| | - Kotaro Takeda
- Clinical Research Center, Murayama Medical Center, Musashimurayama, Japan; Faculty of Rehabilitation, School of Health Sciences, Fujita Health University, Toyoake, Japan
| | - Jiro Terada
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Akira Umeda
- Department of Respiratory Medicine, International University of Health and Welfare Shioya Hospital, Yaita, Japan
| | - Masashi Yoshizawa
- Clinical Research Center, Murayama Medical Center, Musashimurayama, Japan; Department of Pediatrics, Faculty of Medicine, University of Yamanashi, Chuo, Japan
| | - Yosuke Kono
- Clinical Research Center, Murayama Medical Center, Musashimurayama, Japan; Department of Pediatrics, Faculty of Medicine, University of Yamanashi, Chuo, Japan
| | - Yohei Hasebe
- Clinical Research Center, Murayama Medical Center, Musashimurayama, Japan; Department of Pediatrics, Faculty of Medicine, University of Yamanashi, Chuo, Japan
| | - Hiroshi Onimaru
- Department of Physiology, Showa University School of Medicine, Tokyo, Japan
| | | | - Yasumasa Okada
- Clinical Research Center, Murayama Medical Center, Musashimurayama, Japan
| |
Collapse
|
19
|
Gu L, Yu Q, Shen Y, Wang Y, Xu Q, Zhang H. The role of monoaminergic neurons in modulating respiration during sleep and the connection with SUDEP. Biomed Pharmacother 2022; 150:112983. [PMID: 35453009 DOI: 10.1016/j.biopha.2022.112983] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 04/04/2022] [Accepted: 04/14/2022] [Indexed: 11/25/2022] Open
Abstract
Sudden unexpected death in epilepsy (SUDEP) is the leading cause of death among epilepsy patients, occurring even more frequently in cases with anti-epileptic drug resistance. Despite some advancements in characterizing SUDEP, the underlying mechanism remains incompletely understood. This review summarizes the latest advances in our understanding of the pathogenic mechanisms of SUDEP, in order to identify possible targets for the development of new strategies to prevent SUDEP. Based on our previous research along with the current literature, we focus on the role of sleep-disordered breathing (SDB) and its related neural mechanisms to consider the possible roles of monoaminergic neurons in the modulation of respiration during sleep and the occurrence of SUDEP. Overall, this review suggests that targeting the monoaminergic neurons is a promising approach to preventing SUDEP. The proposed roles of SDB and related monoaminergic neural mechanisms in SUDEP provide new insights for explaining the pathogenesis of SUDEP.
Collapse
Affiliation(s)
- LeYuan Gu
- Department of Anesthesiology, The Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou 310006, China; Department of Anesthesiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Qian Yu
- Department of Anesthesiology, The Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou 310006, China; Department of Anesthesiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Yue Shen
- Department of Anesthesiology, The Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou 310006, China; Department of Anesthesiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - YuLing Wang
- Department of Anesthesiology, The Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou 310006, China; Department of Anesthesiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Qing Xu
- Department of Anesthesiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - HongHai Zhang
- Department of Anesthesiology, The Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou 310006, China; Department of Anesthesiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310006, China.
| |
Collapse
|
20
|
Spinieli RL, Ben Musa R, Cornelius-Green J, Hasser EM, Cummings KJ. Orexin facilitates the ventilatory and behavioral responses of rats to hypoxia. Am J Physiol Regul Integr Comp Physiol 2022; 322:R581-R596. [PMID: 35380477 PMCID: PMC9109809 DOI: 10.1152/ajpregu.00334.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 03/22/2022] [Accepted: 03/30/2022] [Indexed: 02/03/2023]
Abstract
Orexin neurons are sensitive to CO2 and contribute to cardiorespiratory homeostasis as well as sensorimotor control. Whether orexin facilitates respiratory and behavioral responses to acute hypoxia is unclear. We hypothesized that orexin neurons are activated by acute hypoxia and that orexin facilitates the hypoxic ventilatory response (HVR), as well as the arterial blood pressure (ABP) and behavioral (movement) responses to acute hypoxia. We further hypothesized that orexin has greater effects in the active phase of the rat circadian cycle, when orexin neurons have high activity. Using whole body plethysmography with EEG, EMG, and the dual-orexin receptor (OxR) antagonist suvorexant (20 mg/kg ip), we determined the effect of OxR blockade on the respiratory, ABP, and behavioral responses of adult rats to acute, graded hypoxia ([Formula: see text]= 0.15, 0.13, 0.11, and 0.09) and hyperoxic hypercapnia ([Formula: see text]= 0.05; [Formula: see text]= 0.95). OxR blockade had no effect on eupnea. OxR blockade significantly reduced the HVR in both inactive and active phases, with a stronger effect in the active phase. OxR blockade reduced the behavioral response to acute hypoxia in the active phase. The central component of the ventilatory and the ABP responses to hypercapnia were reduced by OxR blockade solely in the inactive phase. In the inactive phase, hypoxia activated ∼10% of orexin neurons in the perifornical hypothalamus. These data suggest that orexin neurons participate in the peripheral chemoreflex to facilitate the ventilatory and behavioral responses to acute hypoxia in rats, particularly in the active phase. Orexin also facilitates central chemoreflex responses to CO2 in the inactive phase.
Collapse
Affiliation(s)
- Richard L Spinieli
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, Missouri
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Ruwaida Ben Musa
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, Missouri
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Jennifer Cornelius-Green
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, Missouri
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Eileen M Hasser
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, Missouri
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Kevin J Cummings
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, Missouri
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| |
Collapse
|
21
|
Severs L, Vlemincx E, Ramirez JM. The psychophysiology of the sigh: I: The sigh from the physiological perspective. Biol Psychol 2022; 170:108313. [DOI: 10.1016/j.biopsycho.2022.108313] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 03/08/2022] [Accepted: 03/09/2022] [Indexed: 12/30/2022]
|
22
|
Spinieli RL, Ben Musa R, Kielhofner J, Cornelius-Green J, Cummings KJ. Orexin contributes to eupnea within a critical period of postnatal development. Am J Physiol Regul Integr Comp Physiol 2021; 321:R558-R571. [PMID: 34405704 PMCID: PMC8560369 DOI: 10.1152/ajpregu.00156.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 08/10/2021] [Accepted: 08/10/2021] [Indexed: 02/03/2023]
Abstract
Orexin neurons are active in wakefulness and mostly silent in sleep. In adult rats and humans, orexin facilitates the hypercapnic ventilatory response but has little effect on resting ventilation. The influence of orexin on breathing in the early postnatal period, and across states of vigilance, have not been investigated. This is relevant as the orexin system may be impaired in Sudden Infant Death Syndrome (SIDS) cases. We addressed three hypotheses: 1) orexin provides a drive to breathe in infancy; 2) the effect of orexin depends on stage of postnatal development; and 3) orexin has a greater influence on breathing in wakefulness compared with sleep. Whole body plethysmography was used to monitor breathing of infant rats at three ages: postnatal days (P) 7-8, 12-14, and 17-19. Respiratory variables were analyzed in wakefulness (W), quiet sleep (QS), and active sleep (AS), following suvorexant (5 mg/kg ip), a dual orexin receptor antagonist, or vehicle (DMSO). Effects of suvorexant on ventilatory responses to graded hypercapnia ([Formula: see text] = 0.02, 0.04, 0.06), hypoxia ([Formula: see text] = 0.10), and hyperoxia ([Formula: see text] = 1.0) at P12-14 were also tested. At P12-14, but not at other ages, suvorexant significantly reduced respiratory frequency in all states, reduced the ventilatory equivalent in QW and QS, and increased [Formula: see text] to ∼5 mmHg. Suvorexant had no effect on ventilatory responses to graded hypercapnia or hypoxia. Hyperoxia eliminated the effects of suvorexant on respiratory frequency at P12-14. Our data suggest that orexin preserves eupneic frequency and ventilation in rats, specifically at ∼2 wk of age, perhaps by facilitating tonic peripheral chemoreflex activity.
Collapse
Affiliation(s)
- Richard L Spinieli
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, Missouri
| | - Ruwaida Ben Musa
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, Missouri
| | - Jane Kielhofner
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, Missouri
| | | | - Kevin J Cummings
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| |
Collapse
|
23
|
Liu Q, Duan H, Lian A, Zhuang M, Zhao X, Liu X. Rehabilitation Effects of Acupuncture on the Diaphragm Dysfunction in Chronic Obstructive Pulmonary Disease: A Systematic Review. Int J Chron Obstruct Pulmon Dis 2021; 16:2023-2037. [PMID: 34262271 PMCID: PMC8275099 DOI: 10.2147/copd.s313439] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/01/2021] [Indexed: 12/25/2022] Open
Abstract
Introduction Diaphragm dysfunction is a significant extrapulmonary effect in chronic obstructive pulmonary disease (COPD), which is manifested by changes in diaphragm structure and reduced diaphragm strength. Acupuncture is a traditional rehabilitation technique in China, which has been used in rehabilitation for COPD. But whether acupuncture can improve the diaphragm function of COPD patients remains to be verified. Objective The objective of this study was to evaluate the rehabilitation effects of acupuncture on diaphragm dysfunction in patients with COPD. Methods The authors retrieved in CNKI, VIP, SinoMed, PubMed, Ebsco, Web of Science, from inception to November 2020, for relevant randomized control trials. Two researchers independently screened the articles and extracted the data. The quality of the included studies was evaluated by Physiotherapy Evidence Database scale. The primary outcome measures were maximal inspiratory pressure and the scale for accessory respiratory muscle mobilization, the secondary outcome measures were pulmonary function-related indicators and arterial blood gas indicators. Results Nine articles were finally obtained. Seven studies added acupuncture to standard treatment for patients with diaphragm dysfunction in COPD and found statistically significant changes in the maximum inspiratory pressure and the scale for accessory respiratory muscle mobilization. Two studies have proved that use acupuncture combined with other Traditional Chinese Medicine methods in the rehabilitation for COPD can effectively improve the diaphragm strength and diaphragmatic motor performance. Seven studies showed that acupuncture has obvious improvement in pulmonary ventilation function. Seven studies reported significant differences in arterial blood gas pre- to post-intervention. Conclusion This systematic review found that acupuncture can effectively enhance the diaphragm strength, relieve respiratory muscle fatigue, it can also play a promoting role in improving lung function, hypoxia, and carbon dioxide retention, as well as preventing and alleviating respiratory failure. The generalizability of these results is limited by the design of the included studies.
Collapse
Affiliation(s)
- Qinxin Liu
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Hongxia Duan
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Anbei Lian
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Min Zhuang
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Department of Rehabilitation, Huashan Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Xianli Zhao
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Xiaodan Liu
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Engineering Research Center of Intelligent Rehabilitation of Traditional Chinese Medicine, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| |
Collapse
|
24
|
Fonseca EM, Janes TA, Fournier S, Gargaglioni LH, Kinkead R. Orexin-A inhibits fictive air breathing responses to respiratory stimuli in the bullfrog tadpole (Lithobates catesbeianus). J Exp Biol 2021; 224:239725. [PMID: 33914034 DOI: 10.1242/jeb.240804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 02/25/2021] [Indexed: 11/20/2022]
Abstract
In pre-metamorphic tadpoles, the neural network generating lung ventilation is present but actively inhibited; the mechanisms leading to the onset of air breathing are not well understood. Orexin (ORX) is a hypothalamic neuropeptide that regulates several homeostatic functions, including breathing. While ORX has limited effects on breathing at rest, it potentiates reflexive responses to respiratory stimuli mainly via ORX receptor 1 (OX1R). Here, we tested the hypothesis that OX1Rs facilitate the expression of the motor command associated with air breathing in pre-metamorphic bullfrog tadpoles (Lithobates catesbeianus). To do so, we used an isolated diencephalic brainstem preparation to determine the contributions of OX1Rs to respiratory motor output during baseline breathing, hypercapnia and hypoxia. A selective OX1R antagonist (SB-334867; 5-25 µmol l-1) or agonist (ORX-A; 200 nmol l-1 to 1 µmol l-1) was added to the superfusion media. Experiments were performed under basal conditions (media equilibrated with 98.2% O2 and 1.8% CO2), hypercapnia (5% CO2) or hypoxia (5-7% O2). Under resting conditions gill, but not lung, motor output was enhanced by the OX1R antagonist and ORX-A. Hypercapnia alone did not stimulate respiratory motor output, but its combination with SB-334867 increased lung burst frequency and amplitude, lung burst episodes, and the number of bursts per episode. Hypoxia alone increased lung burst frequency and its combination with SB-334867 enhanced this effect. Inactivation of OX1Rs during hypoxia also increased gill burst amplitude, but not frequency. In contrast with our initial hypothesis, we conclude that ORX neurons provide inhibitory modulation of the CO2 and O2 chemoreflexes in pre-metamorphic tadpoles.
Collapse
Affiliation(s)
- Elisa M Fonseca
- Department of Animal Morphology and Physiology, College of Agricultural and Veterinary Sciences, São Paulo State University, Unesp. Jaboticabal, SP 14884-900, Brazil.,Department of Pediatrics, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, QC, Canada, G1V 4G5
| | - Tara A Janes
- Department of Pediatrics, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, QC, Canada, G1V 4G5
| | - Stéphanie Fournier
- Department of Pediatrics, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, QC, Canada, G1V 4G5
| | - Luciane H Gargaglioni
- Department of Animal Morphology and Physiology, College of Agricultural and Veterinary Sciences, São Paulo State University, Unesp. Jaboticabal, SP 14884-900, Brazil
| | - Richard Kinkead
- Department of Pediatrics, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, QC, Canada, G1V 4G5
| |
Collapse
|
25
|
Berteotti C, Liguori C, Pace M. Dysregulation of the orexin/hypocretin system is not limited to narcolepsy but has far-reaching implications for neurological disorders. Eur J Neurosci 2020; 53:1136-1154. [PMID: 33290595 DOI: 10.1111/ejn.15077] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 10/05/2020] [Accepted: 10/05/2020] [Indexed: 12/23/2022]
Abstract
Neuropeptides orexin A and B (OX-A/B, also called hypocretin 1 and 2) are released selectively by a population of neurons which projects widely into the entire central nervous system but is localized in a restricted area of the tuberal region of the hypothalamus, caudal to the paraventricular nucleus. The OX system prominently targets brain structures involved in the regulation of wake-sleep state switching, and also orchestrates multiple physiological functions. The degeneration and dysregulation of the OX system promotes narcoleptic phenotypes both in humans and animals. Hence, this review begins with the already proven involvement of OX in narcolepsy, but it mainly discusses the new pre-clinical and clinical insights of the role of OX in three major neurological disorders characterized by sleep impairment which have been recently associated with OX dysfunction, such as Alzheimer's disease, stroke and Prader Willi syndrome, and have been emerged over the past 10 years to be strongly associated with the OX dysfunction and should be more considered in the future. In the light of the impairment of the OX system in these neurological disorders, it is conceivable to speculate that the integrity of the OX system is necessary for a healthy functioning body.
Collapse
Affiliation(s)
- Chiara Berteotti
- PRISM Lab, Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - Claudio Liguori
- Sleep Medicine Centre, Neurology Unit, University Hospital Tor Vergata, Rome, Italy.,Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Marta Pace
- Genetics and Epigenetics of Behaviour Laboratory, Istituto Italiano di Tecnologia, Genova, Italy
| |
Collapse
|
26
|
Fonseca EM, Vicente MC, Fournier S, Kinkead R, Bícego KC, Gargaglioni LH. Influence of light/dark cycle and orexins on breathing control in green iguanas (Iguana iguana). Sci Rep 2020; 10:22105. [PMID: 33328521 PMCID: PMC7744544 DOI: 10.1038/s41598-020-79107-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 12/01/2020] [Indexed: 11/17/2022] Open
Abstract
Light/dark cycle affects the physiology of vertebrates and hypothalamic orexin neurons (ORX) are involved in this function. The breathing pattern of the green iguana changes from continuous to episodic across the light/dark phases. Since the stimulatory actions of ORX on breathing are most important during arousal, we hypothesized that ORX regulates changes of breathing pattern in iguanas. Thus, we: (1) Localized ORX neurons with immunohistochemistry; (2) Quantified cyclic changes in plasma orexin-A levels by ELISA; (3) Compared breathing pattern at rest and during hypoxia and hypercarbia; (4) Evaluated the participation of the ORX receptors in ventilation with intracerebroventricular microinjections of ORX antagonists during light and dark phases. We show that the ORX neurons of I. iguana are located in the periventricular hypothalamic nucleus. Orexin-A peaks during the light/active phase and breathing parallels these cyclic changes: ventilation is higher during the light phase than during the dark phase. However, inactivation of ORX-receptors does not affect the breathing pattern. Iguanas increase ventilation during hypoxia only during the light phase. Conversely, CO2 promotes post-hypercarbic hyperpnea during both phases. We conclude that ORXs potentiate the post-hypercarbic (but not the hypoxic)-drive to breathe and are not involved in light/dark changes in the breathing pattern.
Collapse
Affiliation(s)
- Elisa M Fonseca
- Department of Animal Morphology and Physiology, College of Agricultural and Veterinary Sciences, São Paulo State University, Unesp, Via de Acesso Prof. Paulo Donato Castellane s/n, Jaboticabal, SP, CEP 14884-900, Brazil
| | - Mariane C Vicente
- Department of Animal Morphology and Physiology, College of Agricultural and Veterinary Sciences, São Paulo State University, Unesp, Via de Acesso Prof. Paulo Donato Castellane s/n, Jaboticabal, SP, CEP 14884-900, Brazil
| | - Stephanie Fournier
- Department of Pediatrics, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Quebec, QC, Canada
| | - Richard Kinkead
- Department of Pediatrics, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Quebec, QC, Canada
| | - Kênia C Bícego
- Department of Animal Morphology and Physiology, College of Agricultural and Veterinary Sciences, São Paulo State University, Unesp, Via de Acesso Prof. Paulo Donato Castellane s/n, Jaboticabal, SP, CEP 14884-900, Brazil
| | - Luciane H Gargaglioni
- Department of Animal Morphology and Physiology, College of Agricultural and Veterinary Sciences, São Paulo State University, Unesp, Via de Acesso Prof. Paulo Donato Castellane s/n, Jaboticabal, SP, CEP 14884-900, Brazil.
| |
Collapse
|
27
|
Berteotti C, Lo Martire V, Alvente S, Bastianini S, Matteoli G, Silvani A, Zoccoli G. Effect of ambient temperature on sleep breathing phenotype in mice: the role of orexins. J Exp Biol 2020; 223:jeb219485. [PMID: 32457059 DOI: 10.1242/jeb.219485] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 05/19/2020] [Indexed: 11/20/2022]
Abstract
The loss of orexinergic neurons, which release orexins, results in narcolepsy. Orexins participate in the regulation of many physiological functions, and their role as wake-promoting molecules has been widely described. Less is known about the involvement of orexins in body temperature and respiratory regulation. The aim of this study was to investigate if orexin peptides modulate respiratory regulation as a function of ambient temperature (Ta) during different sleep stages. Respiratory phenotype of male orexin knockout (KO-ORX, N=9) and wild-type (WT, N=8) mice was studied at thermoneutrality (Ta=30°C) or during mild cold exposure (Ta=20°C) inside a whole-body plethysmography chamber. The states of wakefulness (W), non-rapid eye movement sleep (NREMS) and rapid eye movement sleep (REMS) were scored non-invasively, using a previously validated technique. In both WT and KO-ORX mice, Ta strongly and significantly affected ventilatory period and minute ventilation values during NREMS and REMS; moreover, the occurrence rate of sleep apneas in NREMS was significantly reduced at Ta=20°C compared with Ta=30°C. Overall, there were no differences in respiratory regulation during sleep between WT and KO-ORX mice, except for sigh occurrence rate, which was significantly increased at Ta=20°C compared with Ta=30°C in WT mice, but not in KO-ORX mice. These results do not support a main role for orexin peptides in the temperature-dependent modulation of respiratory regulation during sleep. However, we showed that the occurrence rate of sleep apneas critically depends on Ta, without any significant effect of orexin peptides.
Collapse
Affiliation(s)
- Chiara Berteotti
- PRISM Lab, Department of Biomedical and Neuromotor Sciences, and Center for Applied Biomedical Research, Department of Medical and Surgical Sciences, Sant'Orsola University Hospital, Alma Mater Studiorum - University of Bologna, Bologna 40126, Italy
| | - Viviana Lo Martire
- PRISM Lab, Department of Biomedical and Neuromotor Sciences, and Center for Applied Biomedical Research, Department of Medical and Surgical Sciences, Sant'Orsola University Hospital, Alma Mater Studiorum - University of Bologna, Bologna 40126, Italy
| | - Sara Alvente
- PRISM Lab, Department of Biomedical and Neuromotor Sciences, and Center for Applied Biomedical Research, Department of Medical and Surgical Sciences, Sant'Orsola University Hospital, Alma Mater Studiorum - University of Bologna, Bologna 40126, Italy
| | - Stefano Bastianini
- PRISM Lab, Department of Biomedical and Neuromotor Sciences, and Center for Applied Biomedical Research, Department of Medical and Surgical Sciences, Sant'Orsola University Hospital, Alma Mater Studiorum - University of Bologna, Bologna 40126, Italy
| | - Gabriele Matteoli
- PRISM Lab, Department of Biomedical and Neuromotor Sciences, and Center for Applied Biomedical Research, Department of Medical and Surgical Sciences, Sant'Orsola University Hospital, Alma Mater Studiorum - University of Bologna, Bologna 40126, Italy
| | - Alessandro Silvani
- PRISM Lab, Department of Biomedical and Neuromotor Sciences, and Center for Applied Biomedical Research, Department of Medical and Surgical Sciences, Sant'Orsola University Hospital, Alma Mater Studiorum - University of Bologna, Bologna 40126, Italy
| | - Giovanna Zoccoli
- PRISM Lab, Department of Biomedical and Neuromotor Sciences, and Center for Applied Biomedical Research, Department of Medical and Surgical Sciences, Sant'Orsola University Hospital, Alma Mater Studiorum - University of Bologna, Bologna 40126, Italy
| |
Collapse
|
28
|
Varga AG, Maletz SN, Bateman JT, Reid BT, Levitt ES. Neurochemistry of the Kölliker-Fuse nucleus from a respiratory perspective. J Neurochem 2020; 156:16-37. [PMID: 32396650 DOI: 10.1111/jnc.15041] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 04/27/2020] [Accepted: 05/04/2020] [Indexed: 12/11/2022]
Abstract
The Kölliker-Fuse nucleus (KF) is a functionally distinct component of the parabrachial complex, located in the dorsolateral pons of mammals. The KF has a major role in respiration and upper airway control. A comprehensive understanding of the KF and its contributions to respiratory function and dysfunction requires an appreciation for its neurochemical characteristics. The goal of this review is to summarize the diverse neurochemical composition of the KF, focusing on the neurotransmitters, neuromodulators, and neuropeptides present. We also include a description of the receptors expressed on KF neurons and transporters involved in each system, as well as their putative roles in respiratory physiology. Finally, we provide a short section reviewing the literature regarding neurochemical changes in the KF in the context of respiratory dysfunction observed in SIDS and Rett syndrome. By over-viewing the current literature on the neurochemical composition of the KF, this review will serve to aid a wide range of topics in the future research into the neural control of respiration in health and disease.
Collapse
Affiliation(s)
- Adrienn G Varga
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, USA.,Department of Physical Therapy, Center for Respiratory Research and Rehabilitation, University of Florida, Gainesville, FL, USA
| | - Sebastian N Maletz
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, USA
| | - Jordan T Bateman
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, USA.,Department of Physical Therapy, Center for Respiratory Research and Rehabilitation, University of Florida, Gainesville, FL, USA
| | - Brandon T Reid
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, USA
| | - Erica S Levitt
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, USA.,Department of Physical Therapy, Center for Respiratory Research and Rehabilitation, University of Florida, Gainesville, FL, USA
| |
Collapse
|
29
|
Lo Martire V, Berteotti C, Bastianini S, Alvente S, Valli A, Cerri M, Amici R, Silvani A, Swoap SJ, Zoccoli G. The physiological signature of daily torpor is not orexin dependent. J Comp Physiol B 2020; 190:493-507. [DOI: 10.1007/s00360-020-01281-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 04/01/2020] [Accepted: 04/27/2020] [Indexed: 12/25/2022]
|
30
|
Barnett S, Li A. Orexin in Respiratory and Autonomic Regulation, Health and Diseases. Compr Physiol 2020; 10:345-363. [DOI: 10.1002/cphy.c190013] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
31
|
Barros D, García-Río F. Obstructive sleep apnea and dyslipidemia: from animal models to clinical evidence. Sleep 2020; 42:5204276. [PMID: 30476296 DOI: 10.1093/sleep/zsy236] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 10/02/2018] [Accepted: 11/22/2018] [Indexed: 01/11/2023] Open
Abstract
Lipid metabolism deregulation constitutes the pathogenic basis for the development of atherosclerosis and justifies a high incidence of cardiovascular-related morbidity and mortality. Some data suggest that dyslipidemia may be associated with sleep-disordered breathing, mainly obstructive sleep apnea (OSA), due to alterations in fundamental biochemical processes, such as intermittent hypoxia (IH). The aim of this systematic review was to identify and critically evaluate the current evidence supporting the existence of a possible relationship between OSA and alterations in lipid metabolism. Much evidence shows that, during the fasting state, OSA and IH increase lipid delivery from the adipose tissue to the liver through an up-regulation of the sterol regulatory element-binding protein-1 and stearoyl-CoA desaturase-1, increasing the synthesis of cholesterol esters and triglycerides. In the postprandial state, lipoprotein clearance is delayed due to lower lipoprotein lipase activity, probably secondary to IH-up-regulation of angiopoietin-like protein 4 and decreased activity of the peroxisome proliferator-activated receptor alpha. Moreover, oxidative stress can generate dysfunctional oxidized lipids and reduce the capacity of high-density lipoproteins (HDL) to prevent low-density lipoprotein (LDL) oxidation. In the clinical field, several observational studies and a meta-regression analysis support the existence of a link between OSA and dyslipidemia. Although there is evidence of improved lipid profile after apnea-hypopnea suppression with continuous positive airway pressure (CPAP), the majority of the data come from observational studies. In contrast, randomized controlled trials evaluating the effects of CPAP on lipid metabolism present inconclusive results and two meta-analyses provide contradictory evidence.
Collapse
Affiliation(s)
- David Barros
- Servicio de Neumología, Hospital Montecelo, Pontevedra, Spain
| | - Francisco García-Río
- Servicio de Neumología, Hospital Universitario La Paz-IdiPAZ, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
32
|
Furuya WI, Bassi M, Menani JV, Colombari E, Zoccal DB, Colombari DSA. Modulation of hypercapnic respiratory response by cholinergic transmission in the commissural nucleus of the solitary tract. Pflugers Arch 2019; 472:49-60. [PMID: 31884528 DOI: 10.1007/s00424-019-02341-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 11/28/2019] [Accepted: 12/11/2019] [Indexed: 01/16/2023]
Abstract
The nucleus of the solitary tract (NTS) is an important area of the brainstem that receives and integrates afferent cardiorespiratory sensorial information, including those from arterial chemoreceptors and baroreceptors. It was described that acetylcholine (ACh) in the commissural subnucleus of the NTS (cNTS) promotes an increase in the phrenic nerve activity (PNA) and antagonism of nicotinic receptors in the same region reduces the magnitude of tachypneic response to peripheral chemoreceptor stimulation, suggesting a functional role of cholinergic transmission within the cNTS in the chemosensory control of respiratory activity. In the present study, we investigated whether cholinergic receptor antagonism in the cNTS modifies the sympathetic and respiratory reflex responses to hypercapnia. Using an arterially perfused in situ preparation of juvenile male Holtzman rats, we found that the nicotinic antagonist (mecamylamine, 5 mM), but not the muscarinic antagonist (atropine, 5 mM), into the cNTS attenuated the hypercapnia-induced increase of hypoglossal activity. Furthermore, mecamylamine in the cNTS potentiated the generation of late-expiratory (late-E) activity in abdominal nerve induced by hypercapnia. None of the cholinergic antagonists microinjected in the cNTS changed either the sympathetic or the phrenic nerve responses to hypercapnia. Our data provide evidence for the role of cholinergic transmission in the cNTS, acting on nicotinic receptors, modulating the hypoglossal and abdominal responses to hypercapnia.
Collapse
Affiliation(s)
- Werner I Furuya
- Department of Physiology and Pathology, School of Dentistry, UNESP - São Paulo State University, Araraquara, SP, Brazil
| | - Mirian Bassi
- Department of Physiology and Pathology, School of Dentistry, UNESP - São Paulo State University, Araraquara, SP, Brazil
| | - José V Menani
- Department of Physiology and Pathology, School of Dentistry, UNESP - São Paulo State University, Araraquara, SP, Brazil
| | - Eduardo Colombari
- Department of Physiology and Pathology, School of Dentistry, UNESP - São Paulo State University, Araraquara, SP, Brazil
| | - Daniel B Zoccal
- Department of Physiology and Pathology, School of Dentistry, UNESP - São Paulo State University, Araraquara, SP, Brazil
| | - Débora S A Colombari
- Department of Physiology and Pathology, School of Dentistry, UNESP - São Paulo State University, Araraquara, SP, Brazil.
| |
Collapse
|
33
|
Chen Y, Guo Y, Yan X, Zeng M, Chen H, Qiu D, Wang J. Orexin-A Excites Airway Vagal Preganglionic Neurons via Activation of Orexin Receptor Type 1 and Type 2 in Rats. Front Cell Neurosci 2019; 13:478. [PMID: 31708749 PMCID: PMC6819310 DOI: 10.3389/fncel.2019.00478] [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: 06/14/2019] [Accepted: 10/10/2019] [Indexed: 11/19/2022] Open
Abstract
Airway vagal nerves play a predominant role in the neural control of the airway, and augmented airway vagal activity is known to play important roles in the pathogenesis of some chronic inflammatory airway diseases. Several lines of evidence indicate that dysfunctional central orexinergic system is closely related to the severity of airway diseases, however, whether orexins affect airway vagal activity is unknown. This study investigates whether and how orexin-A regulates the activity of medullary airway vagal preganglionic neurons (AVPNs). The expression of orexin receptor type 1 (OX1R) and type 2 (OX2R) was examined using immunofluorescent staining. The effects of orexin-A on functionally identified inspiratory-activated AVPNs (IA-AVPNs), which are critical in the control of airway smooth muscle, were examined using patch-clamp in medullary slices of neonatal rats. Airway vagal response to injection of orexin-A into the magna cisterna was examined using plethysmography in juvenile rats. The results show that retrogradely labeled AVPNs were immunoreactive to anti-OX1R antibody and anti-OX2R antibody. Orexin-A dose-dependently depolarized IA-AVPNs and increased their firing rate. In synaptically isolated IA-AVPNs, the depolarization induced by orexin-A was blocked partially by OX1R antagonist SB-334867 or OX2R antagonist TCS OX2 29 alone, and completely by co-application of both antagonists. The orexin-A-induced depolarization was also mostly blocked by Na+/Ca2+ exchanger inhibitor KB-R7943. Orexin-A facilitated the glutamatergic, glycinergic and GABAergic inputs to IA-AVPNs, and the facilitation of each type of input was blocked partially by SB-334867 or TCS OX2 29 alone, and completely by co-application of both antagonists. Injection of orexin-A into the magna cisterna of juvenile rats significantly increased the inspiratory and expiratory resistance of the airway and consequently decreased the dynamic compliance of the lungs, all of which were prevented by atropine sulfate or bilateral vagotomy. These results demonstrate that orexin-A excites IA-AVPNs via activation of both OX1R and OX2R, and suggest that increased central synthesis/release of orexins might participate in the pathogenesis of airway diseases via over-activation of AVPNs.
Collapse
Affiliation(s)
- Yonghua Chen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yuhong Guo
- Department of Neurobiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xianxia Yan
- Department of Neurobiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ming Zeng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Hong Chen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Dongying Qiu
- Department of Gerontology, Fudan University Affiliated Zhongshan Hospital, Shanghai, China
| | - Jijiang Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
34
|
Hoshino T, Sasanabe R, Mano M, Nomura A, Kato C, Sato M, Imai M, Murotani K, Guilleminault C, Shiomi T. Prevalence of Rapid Eye Movement-related Obstructive Sleep Apnea in Adult Narcolepsy. Intern Med 2019; 58:2151-2157. [PMID: 30996185 PMCID: PMC6709340 DOI: 10.2169/internalmedicine.2601-18] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Objective The association between narcolepsy and rapid eye movement (REM)-related obstructive sleep apnea (OSA) has not been reported. This study aimed to examine the prevalence of REM-related OSA in narcolepsy patients. Methods From January 2013 to April 2018, 141 adult patients were diagnosed with narcolepsy using nocturnal polysomnography and the multiple sleep latency test. The prevalence of REM-related OSA in narcolepsy patients was retrospectively reviewed. Three criteria were used to determine REM-related OSA: Definition #1, an overall apnea-hypopnea index (AHI) ≥5 and AHI during REM (AHIREM)/AHI during non-rapid eye movement (NREM) (AHINREM) ≥2; Definition #2, an overall AHI ≥5 and AHIREM/AHINREM≥2 and AHINREM <15; and Definition #3, an overall AHI ≥5 and AHIREM/AHINREM≥2 and AHINREM <8 plus an REM sleep duration >10.5 minutes. Results Of the 141 narcolepsy patients, 26 were diagnosed with narcolepsy with cataplexy (NA-CA) and 115 with narcolepsy without cataplexy (NA w/o CA). Seventeen patients with NA-CA and 39 with NA w/o CA had OSA. According to Definition #1, the prevalence of REM-related OSA was 47.1% and 41.0%, respectively, in OSA patients with NA-CA and NA w/o CA; according to Definition #2, the respective prevalence was 47.1% and 38.5%, while that according to Definition #3 was 41.2% and 25.6%. No significant differences were found in the prevalence of REM-related OSA for each definition. Conclusion A high prevalence of REM-related OSA was confirmed in adult narcolepsy patients with OSA. Compared to previous reports, we noted a high frequency of REM-related OSA satisfying the relatively strict Definition #3. These results might reflect the pathophysiological characteristics of narcolepsy.
Collapse
Affiliation(s)
- Tetsuro Hoshino
- Department of Sleep Medicine and Sleep Disorders Center, Aichi Medical University Hospital, Japan
| | - Ryujiro Sasanabe
- Department of Sleep Medicine and Sleep Disorders Center, Aichi Medical University Hospital, Japan
| | - Mamiko Mano
- Department of Sleep Medicine and Sleep Disorders Center, Aichi Medical University Hospital, Japan
| | - Atsuhiko Nomura
- Department of Sleep Medicine and Sleep Disorders Center, Aichi Medical University Hospital, Japan
| | - Chihiro Kato
- Department of Sleep Medicine and Sleep Disorders Center, Aichi Medical University Hospital, Japan
| | - Masako Sato
- Department of Sleep Medicine and Sleep Disorders Center, Aichi Medical University Hospital, Japan
| | - Masato Imai
- Department of Sleep Medicine and Sleep Disorders Center, Aichi Medical University Hospital, Japan
| | - Kenta Murotani
- Center for Clinical Research, Aichi Medical University Hospital, Japan
| | | | - Toshiaki Shiomi
- Department of Sleep Medicine and Sleep Disorders Center, Aichi Medical University Hospital, Japan
| |
Collapse
|
35
|
Dhaibar H, Gautier NM, Chernyshev OY, Dominic P, Glasscock E. Cardiorespiratory profiling reveals primary breathing dysfunction in Kcna1-null mice: Implications for sudden unexpected death in epilepsy. Neurobiol Dis 2019; 127:502-511. [PMID: 30974168 PMCID: PMC6588471 DOI: 10.1016/j.nbd.2019.04.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 03/14/2019] [Accepted: 04/05/2019] [Indexed: 12/20/2022] Open
Abstract
Sudden unexpected death in epilepsy (SUDEP) is the leading cause of epilepsy-related mortality, but the relative importance of underlying cardiac and respiratory mechanisms remains unclear. To illuminate the interactions between seizures, respiration, cardiac function, and sleep that contribute to SUDEP risk, here we developed a mouse epilepsy monitoring unit (EMU) to simultaneously record video, electroencephalography (EEG), electromyography (EMG), plethysmography, and electrocardiography (ECG) in a commonly used genetic model of SUDEP, the Kcna1 knockout (Kcna1-/-) mouse. During interictal periods, Kcna1-/- mice exhibited an abnormal absence of post-sigh apneas and a 3-fold increase in respiratory variability. During spontaneous convulsive seizures, Kcna1-/- mice displayed an array of aberrant breathing patterns that always preceded cardiac abnormalities. These findings support respiratory dysfunction as a primary risk factor for susceptibility to deleterious cardiorespiratory sequelae in epilepsy and reveal a new role for Kcna1-encoded Kv1.1 channels in the regulation of basal respiratory physiology.
Collapse
Affiliation(s)
- Hemangini Dhaibar
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center Shreveport, LA 71103, USA.
| | - Nicole M Gautier
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center Shreveport, LA 71103, USA.
| | - Oleg Y Chernyshev
- Department of Neurology, Division of Sleep Medicine, Louisiana State University Health Sciences Center Shreveport, LA 71103, USA.
| | - Paari Dominic
- Department of Internal Medicine, Section of Cardiology, Louisiana State University Health Sciences Center Shreveport, LA 71103, USA.
| | - Edward Glasscock
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center Shreveport, LA 71103, USA.
| |
Collapse
|
36
|
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
|
37
|
Bastianini S, Alvente S, Berteotti C, Bosi M, Lo Martire V, Silvani A, Valli A, Zoccoli G. Post-sigh sleep apneas in mice: Systematic review and data-driven definition. J Sleep Res 2019; 28:e12845. [PMID: 30920081 DOI: 10.1111/jsr.12845] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 02/01/2019] [Accepted: 02/18/2019] [Indexed: 11/26/2022]
Abstract
Sleep apneas can be categorized as post-sigh (prevailing in non-rapid eye movement sleep) or spontaneous (prevailing in rapid eye movement sleep) according to whether or not they are preceded by an augmented breath (sigh). Notably, the occurrence of these apnea subtypes changes differently in hypoxic/hypercapnic environments and in some genetic diseases, highlighting the importance of an objective discrimination. We aim to: (a) systematically review the literature comparing the criteria used in categorizing mouse sleep apneas; and (b) provide data-driven criteria for this categorization, with the final goal of reducing experimental variability in future studies. Twenty-two wild-type mice, instrumented with electroencephalographic/electromyographic electrodes, were placed inside a whole-body plethysmographic chamber to quantify sleep apneas and sighs. Wake-sleep states were scored on 4-s epochs based on electroencephalographic/electromyographic signals. Literature revision showed that highly different criteria were used for post-sigh apnea definition, the intervals for apnea occurrence after sigh ranging from 1 breath up to 20 s. In our data, the apnea occurrence rate during non-rapid eye movement sleep was significantly higher than that calculated before the sigh only in the 1st and 2nd 4-s epochs following a sigh. These data suggest that, in mice, apneas should be categorized as post-sigh only if they start within 8 s from a sigh; the choice of shorter or longer time windows might underestimate or slightly overestimate their occurrence rate, respectively.
Collapse
Affiliation(s)
- Stefano Bastianini
- PRISM Lab, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Sara Alvente
- PRISM Lab, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Chiara Berteotti
- PRISM Lab, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Marcello Bosi
- ASL of Romagna, Department Thoracic Diseases, Pulmonary Operative Unit, Morgagni-Pierantoni Hospital, Forlì, Italy
| | - Viviana Lo Martire
- PRISM Lab, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Alessandro Silvani
- PRISM Lab, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Alice Valli
- PRISM Lab, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Giovanna Zoccoli
- PRISM Lab, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
38
|
Summers CH, Yaeger JDW, Staton CD, Arendt DH, Summers TR. Orexin/hypocretin receptor modulation of anxiolytic and antidepressive responses during social stress and decision-making: Potential for therapy. Brain Res 2018; 1731:146085. [PMID: 30590027 DOI: 10.1016/j.brainres.2018.12.036] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 12/15/2018] [Accepted: 12/21/2018] [Indexed: 12/12/2022]
Abstract
Hypothalmic orexin/hypocretin (Orx) neurons in the lateral and dorsomedial perifornical region (LH-DMH/PeF) innervate broadly throughout the brain, and receive similar inputs. This wide distribution, as well as two Orx peptides (OrxA and OrxB) and two Orx receptors (Orx1 and Orx2) allow for functionally related but distinctive behavioral outcomes, that include arousal, sleep-wake regulation, food seeking, metabolism, feeding, reward, addiction, and learning. These are all motivational functions, and tie the orexin systems to anxiety and depression as well. We present evidence, that for affective behavior, Orx1 and Orx2 receptors appear to have opposing functions. The majority of research on anxiety- and depression-related outcomes has focused on Orx1 receptors, which appear to have primarily anxiogenic and pro-depressive actions. Although there is significant research suggesting contrary findings, the primary potential for pharmacotherapies linked to the Orx1 receptor is via antagonists to block anxious and depressive behavior. Dual orexin receptor antagonists have been approved for treatment of sleep disorders, and are likely candidates for adaptation for affect disorder treatments. However, we present evidence here that demonstrates the Orx2 receptors are anxiolytic and antidepressive. Using a new experimental pre-clinical model of anxious and depressive behavior stimulated by social stress and decision-making that produces two stable behavioral phenotypes, Escape/Resilient and Stay/Susceptible, we tested the effects of intracerebroventricular injections of Orx2 agonist and antagonist drugs. Over ten behavioral measures, we have demonstrated that Orx2 agonists promote resilience, as well as anxiolytic and antidepressive behavior. In contrast, Orx2 antagonists or knockdown kindle anxious and pro-depressive behavior plus increase susceptibility. The results suggest that the Orx2 receptor may be a useful target for pharmacotherapies to treat anxiety and depression.
Collapse
Affiliation(s)
- Cliff H Summers
- Department of Biology, University of South Dakota, Vermillion, SD 57069 USA; Neuroscience Group, Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD 57069 USA; Veterans Affairs Research Service, Sioux Falls VA Health Care System, Sioux Falls, SD 57105 USA.
| | - Jazmine D W Yaeger
- Department of Biology, University of South Dakota, Vermillion, SD 57069 USA; Neuroscience Group, Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD 57069 USA; Veterans Affairs Research Service, Sioux Falls VA Health Care System, Sioux Falls, SD 57105 USA
| | - Clarissa D Staton
- Department of Biology, University of South Dakota, Vermillion, SD 57069 USA; Neuroscience Group, Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD 57069 USA; Veterans Affairs Research Service, Sioux Falls VA Health Care System, Sioux Falls, SD 57105 USA
| | - David H Arendt
- Neuroscience Group, Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD 57069 USA
| | - Tangi R Summers
- Department of Biology, University of South Dakota, Vermillion, SD 57069 USA; Neuroscience Group, Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD 57069 USA; Veterans Affairs Research Service, Sioux Falls VA Health Care System, Sioux Falls, SD 57105 USA
| |
Collapse
|
39
|
Lo Martire V, Alvente S, Bastianini S, Berteotti C, Valli A, Manconi M, Zoccoli G, Silvani A. Sleep and Tibialis Anterior Muscle Activity in Mice With Mild Hypoxia and Iron Deficiency: Implications for the Restless Legs Syndrome. Front Physiol 2018; 9:1818. [PMID: 30618828 PMCID: PMC6304696 DOI: 10.3389/fphys.2018.01818] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 12/05/2018] [Indexed: 12/14/2022] Open
Abstract
Restless legs syndrome (RLS) is a neurological disorder that entails an urge to move with a circadian pattern during the evening/night. RLS may be accompanied by decreased sleep time and increased occurrence of periodic leg movements during sleep (PLMS), which involve bursts of tibialis anterior (TA) muscle electromyogram (EMG). Mild hypoxia and non-anemic iron deficiency, a highly prevalent nutritional deficiency, are relatively unexplored factors in RLS pathophysiology. We tested whether mice exposed to mild hypoxia, alone or in combination with non-anemic iron deficiency, show decreased sleep time particularly in the light (rest) period and increased occurrence of TA EMG phasic events similar to human PLMS. Female C57BL/6J mice were fed diets with low or normal iron for 6 months from weaning and instrumented with electrodes to record the electroencephalogram and the EMG of both TA muscles. Mice were recorded in a whole-body plethysmograph while breathing a normoxic or mildly hypoxic (15% O2) gas mixture for 48 h. Hypoxia increased minute ventilation during sleep. The low-iron diet decreased liver and serum iron, leaving blood hemoglobin and brainstem iron levels unaffected. Hypoxia, either alone or in combination with non-anemic iron deficiency, decreased non-rapid-eye-movement (non-REM) sleep time, but this occurred irrespective of the light/dark period and was not associated with increased occurrence of TA EMG events during non-REM sleep. These results do not support the hypothesis that mild hypoxia is sufficient to cause signs of RLS, either alone or in combination with non-anemic iron deficiency, pointing to the necessity of further susceptibility factors.
Collapse
Affiliation(s)
- Viviana Lo Martire
- Laboratory of Physiological Regulations in Sleeping Mice, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Sara Alvente
- Laboratory of Physiological Regulations in Sleeping Mice, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Stefano Bastianini
- Laboratory of Physiological Regulations in Sleeping Mice, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Chiara Berteotti
- Laboratory of Physiological Regulations in Sleeping Mice, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Alice Valli
- Laboratory of Physiological Regulations in Sleeping Mice, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Mauro Manconi
- Sleep and Epilepsy Center, Neurocenter of Southern Switzerland, Civic Hospital (EOC) of Lugano, Lugano, Switzerland.,Department of Neurology, Bern University Hospital, Bern, Switzerland
| | - Giovanna Zoccoli
- Laboratory of Physiological Regulations in Sleeping Mice, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Alessandro Silvani
- Laboratory of Physiological Regulations in Sleeping Mice, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
40
|
da Silva EN, Horta-Júnior JDAC, Gargaglioni LH, Dias MB. ATP in the lateral hypothalamus/perifornical area enhances the CO 2 chemoreflex control of breathing. Exp Physiol 2018; 103:1679-1691. [PMID: 30242927 DOI: 10.1113/ep087182] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 09/20/2018] [Indexed: 12/24/2022]
Abstract
NEW FINDINGS What is the central question of this study? ATP is known to modulate the chemosensitivity of some brain areas. However, whether the ATP contributes specifically to the mechanism of chemoreception in the lateral hypothalamus/perifornical area (LH/PFA) remains to be determined. What is the main finding and its importance? ATP, acting on the LH/PFA, enhances the hypercapnic ventilatory response in rats during wakefulness, in the dark period. Our results highlight the importance of ATP as a modulator of central chemoreception and provide new insight regarding the mechanisms involved in LH/PFA chemosensitivity and the sleep-wake differences in the CO2 /H+ -dependent drive to breathe. ABSTRACT The lateral hypothalamus/perifornical area (LH/PFA) is a central chemoreceptor site, which acts in an arousal state-dependent manner. It has been shown that purinergic signalling through ATP influences the CO2 /H+ responsiveness of other chemosensitive regions, but it is unknown whether ATP is also involved in the mechanisms that underlie LH/PFA chemoreception. Here, we studied the effects of microdialysis of a P2X-receptor agonist [α,β-methylene ATP (α,β-meATP), 10 mm] and a non-selective P2-receptor antagonist [pyridoxal-phosphate-6-azophenyl-2',4'-disulfonate (PPADS), 1 mm] into the LH/PFA of conscious rats on ventilation in room air and in 7% CO2 . In the dark (active) phase, but not in the light, microdialysis of α,β-meATP caused an augmented hypercapnic ventilatory response during wakefulness, but not during non-REM sleep (P < 0.001). PPADS caused no change in CO2 ventilatory responses in either the dark period or the light period. Our data suggest that ATP in LH/PFA contributes to the hypercapnic ventilatory response in conscious rats during wakefulness in the dark phase of the diurnal cycle.
Collapse
Affiliation(s)
- Eliandra N da Silva
- Department of Physiology, Institute of Bioscience, Sao Paulo State University-UNESP, Botucatu, SP, Brazil
| | | | - Luciane H Gargaglioni
- Department of Animal Morphology and Physiology, Sao Paulo State University-FCAV, Jaboticabal, SP, Brazil
| | - Mirela B Dias
- Department of Physiology, Institute of Bioscience, Sao Paulo State University-UNESP, Botucatu, SP, Brazil
| |
Collapse
|
41
|
Zhang XF, Qin Q, Geng WY, Jiang CW, Liu Y, Liu XL, Li J, Liu ZB. Electroacupuncture reduces hypothalamic and medullary expression of orexins and their receptors in a rat model of chronic obstructive pulmonary disease. Acupunct Med 2018; 36:312-318. [PMID: 29669795 DOI: 10.1136/acupmed-2017-011391] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/22/2017] [Indexed: 01/05/2023]
Abstract
OBJECTIVES Decreased lung function in chronic obstructive pulmonary disease (COPD) is correlated with abnormal excitability of the respiratory centre where orexin neuropeptides from the hypothalamus are responsible for regulating respiration. We hypothesised that improvements in pulmonary function with electroacupuncture (EA) may be related to orexins in a rat model of COPD. METHODS The COPD model was established by cigarette smoke exposure and lipopolysaccharide injection. Modelled rats received EA at BL13 and ST36 for two weeks, after which lung function was tested. Orexin levels in the hypothalamus and medulla were detected by ELISA, while mRNA/protein expression and localisation of orexins and their receptors were investigated using real time PCR, Western blotting and immunohistochemistry, respectively. RESULTS The decrease in lung function observed in COPD rats was improved after EA treatment. Orexin levels in the hypothalamus and medulla were significantly higher in COPD rats than in normal rats, but were significantly reduced in the EA-treated group. There was a negative correlation between orexin content and lung function. In the hypothalamus, mRNA and protein expression and immunoreactivity of orexins were significantly higher in the COPD group than in the normal group, but a significant decrease was observed after EA. In the medulla, the expression and immunoreactivity of orexin receptors were significantly higher in the COPD group than in the normal group, but a significant decrease was observed after EA. CONCLUSIONS The positive effect of EA on pulmonary function in COPD rats may be related to downregulation of orexins and their receptors in the medulla.
Collapse
Affiliation(s)
- Xin-Fang Zhang
- Department of Physiology, College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Qin Qin
- College of Nursing, Anhui University of Chinese Medicine, Hefei, China
| | - Wen-Ye Geng
- Department of Pharmacology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Chuan-Wei Jiang
- Department of Physiology, College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Yong Liu
- Department of Physiology, College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Xiao-Li Liu
- Institute of Acu-Moxibustion and Meridian, College of Acupuncture and Massage, Anhui University of Chinese Medicine, Hefei, China
| | - Jing Li
- Institute of Acu-Moxibustion and Meridian, College of Acupuncture and Massage, Anhui University of Chinese Medicine, Hefei, China
| | - Zi-Bing Liu
- Department of Physiology, College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China.,Institute of Acu-Moxibustion and Meridian, College of Acupuncture and Massage, Anhui University of Chinese Medicine, Hefei, China
| |
Collapse
|
42
|
Yang CF, Feldman JL. Efferent projections of excitatory and inhibitory preBötzinger Complex neurons. J Comp Neurol 2018; 526:1389-1402. [PMID: 29473167 DOI: 10.1002/cne.24415] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 02/04/2018] [Accepted: 02/09/2018] [Indexed: 02/01/2023]
Abstract
The preBötzinger Complex (preBötC), a compact medullary region essential for generating normal breathing rhythm and pattern, is the kernel of the breathing central pattern generator (CPG). Excitatory preBötC neurons in rats project to major breathing-related brainstem regions. Here, we provide a brainstem connectivity map in mice for both excitatory and inhibitory preBötC neurons. Using a genetic strategy to label preBötC neurons, we confirmed extensive projections of preBötC excitatory neurons within the brainstem breathing CPG including the contralateral preBötC, Bötzinger Complex (BötC), ventral respiratory group, nucleus of the solitary tract, parahypoglossal nucleus, parafacial region (RTN/pFRG or alternatively, pFL /pFV ), parabrachial and Kölliker-Füse nuclei, as well as major projections to the midbrain periaqueductal gray. Interestingly, preBötC inhibitory projections paralleled the excitatory projections. Moreover, we examined overlapping projections in the pons in detail and found that they targeted the same neurons. We further explored the direct anatomical link between the preBötC and suprapontine brain regions that may govern emotion and other complex behaviors that can affect or be affected by breathing. Forebrain efferent projections were sparse and restricted to specific nuclei within the thalamus and hypothalamus, with processes rarely observed in cortex, basal ganglia, or other limbic regions, e.g., amygdala or hippocampus. We conclude that the preBötC sends direct, presumably inspiratory-modulated, excitatory and inhibitory projections in parallel to distinct targets throughout the brain that generate and modulate breathing pattern and/or coordinate breathing with other behaviors, physiology, cognition, or emotional state.
Collapse
Affiliation(s)
- Cindy F Yang
- Department of Neurobiology, David Geffen School of Medicine, UCLA, Los Angeles, California, 90095-1763
| | - Jack L Feldman
- Department of Neurobiology, David Geffen School of Medicine, UCLA, Los Angeles, California, 90095-1763
| |
Collapse
|
43
|
Huber MJ, Chen QH, Shan Z. The Orexin System and Hypertension. Cell Mol Neurobiol 2018; 38:385-391. [PMID: 28349223 PMCID: PMC5617754 DOI: 10.1007/s10571-017-0487-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 03/18/2017] [Indexed: 12/18/2022]
Abstract
In this review, we focus on the role of orexin signaling in blood pressure control and its potential link to hypertension by summarizing evidence from several experimental animal models of hypertension. Studies using the spontaneously hypertensive rat (SHR) animal model of human essential hypertension show that pharmacological blockade of orexin receptors reduces blood pressure in SHRs but not in Wistar-Kyoto rats. In addition, increased activity of the orexin system contributes to elevated blood pressure and sympathetic nerve activity (SNA) in dark-active period Schlager hypertensive (BPH/2J) mice, another genetic model of neurogenic hypertension. Similar to these two models, Sprague-Dawley rats with stress-induced hypertension display an overactive central orexin system. Furthermore, upregulation of the orexin receptor 1 increases firing of hypothalamic paraventricular nucleus neurons, augments SNA, and contributes to hypertension in the obese Zucker rat, an animal model of obesity-related hypertension. Finally, we propose a hypothesis for the implication of the orexin system in salt-sensitive hypertension. All of this evidence, coupled with the important role of elevated SNA in increasing blood pressure, strongly suggests that hyperactivity of the orexin system contributes to hypertension.
Collapse
Affiliation(s)
- Michael J Huber
- Department of Kinesiology and Integrative Physiology, Michigan Technological University, SDC 231, 1400 Townsend Drive, Houghton, MI, 49931, USA
| | - Qing-Hui Chen
- Department of Kinesiology and Integrative Physiology, Michigan Technological University, SDC 231, 1400 Townsend Drive, Houghton, MI, 49931, USA
| | - Zhiying Shan
- Department of Kinesiology and Integrative Physiology, Michigan Technological University, SDC 231, 1400 Townsend Drive, Houghton, MI, 49931, USA.
| |
Collapse
|
44
|
Tarasiuk A, Segev Y. Abnormal Growth and Feeding Behavior in Upper Airway Obstruction in Rats. Front Endocrinol (Lausanne) 2018; 9:298. [PMID: 29915561 PMCID: PMC5994397 DOI: 10.3389/fendo.2018.00298] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 05/18/2018] [Indexed: 12/18/2022] Open
Abstract
Pediatric obstructive sleep apnea (OSA) is a syndrome manifesting with snoring and increased respiratory effort due to increased upper airway resistance. In addition to cause the abnormal sleep, this syndrome has been shown to elicit either growth retardation or metabolic syndrome and obesity. Treating OSA by adenotonsillectomy is usually associated with increased risk for obesity, despite near complete restoration of breathing and sleep. However, the underlying mechanism linking upper airways obstruction (AO) to persistent change in food intake, metabolism, and growth remains unclear. Rodent models have examined the impact of intermittent hypoxia on metabolism. However, an additional defining feature of OSA that is not related to intermittent hypoxia is enhanced respiratory loading leading to increased respiratory effort and abnormal sleep. The focus of this mini review is on recent evidence indicating the persistent abnormalities in endocrine regulation of feeding and growth that are not fully restored by the chronic upper AO removal in rats. Here, we highlight important aspects related to abnormal regulation of metabolism that are not related to intermittent hypoxia per se, in an animal model that mimics many of the clinical features of pediatric OSA. Our evidence from the AO model indicates that obstruction removal may not be sufficient to prevent the post-removal tendency for abnormal growth.
Collapse
Affiliation(s)
- Ariel Tarasiuk
- Sleep-Wake Disorders Unit, Soroka University Medical Center, Beer-Sheva, Israel
- Department of Physiology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- *Correspondence: Ariel Tarasiuk,
| | - Yael Segev
- Shraga Segal Department of Microbiology and Immunology, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| |
Collapse
|
45
|
Mishima T, Kasanuki K, Koga S, Castanedes-Casey M, Wszolek ZK, Tsuboi Y, Dickson DW. Reduced orexin immunoreactivity in Perry syndrome and multiple system atrophy. Parkinsonism Relat Disord 2017; 42:85-89. [PMID: 28651750 DOI: 10.1016/j.parkreldis.2017.06.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 06/01/2017] [Accepted: 06/10/2017] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Orexin is a neuropeptide that plays a key role in maintaining a state of arousal, and possibly associates with sleep apnea syndrome (SAS). Reduced orexin immunoreactivity has been reported in various neurologic conditions such as narcolepsy, Alzheimer's disease, Lewy body disease and multiple system atrophy (MSA); however, there has been no report investigating orexin in Perry syndrome, a rare hereditary neurodegenerative disease characterized by four clinical cardinal signs (parkinsonism, depression/apathy, weight loss, and central hypoventilation). Perry syndrome patients frequently have sleep disturbances, including SAS and insomnia. METHODS We evaluated orexin immunoreactivity in Perry syndrome. Using imaging analysis, we quantitatively assessed orexin immunoreactivity in the nucleus basalis of Meynert in three Perry syndrome cases, as well as five cases of frontotemporal lobar degeneration with motor neuron disease, five cases of MSA and five age-matched controls. For these cases, antemortem clinical information on sleep disturbances has been reviewed. RESULTS In Perry syndrome and MSA, there was reduction of orexin immunoreactivity compared with controls (Perry syndrome: p = 0.020, MSA: p < 0.001). In contrast, FTLD-MND did not have significant reduction of orexin immunoreactivity. Two out of three cases of Perry syndrome had SAS confirmed by polysomnography. CONCLUSIONS This is the first report assessing orexin immunoreactivity in Perry syndrome, and it showed significant reduction, similar to select neurodegenerative diseases, such as MSA. Further analysis with more cases will be needed to elucidate the specific mechanism of orexin loss in these disorders.
Collapse
Affiliation(s)
- Takayasu Mishima
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, United States; Department of Neurology, Fukuoka University, Fukuoka 8140180, Japan
| | - Koji Kasanuki
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, United States
| | - Shunsuke Koga
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, United States
| | | | - Zbigniew K Wszolek
- Department of Neurology, Mayo Clinic, Jacksonville, FL 32224, United States
| | - Yoshio Tsuboi
- Department of Neurology, Fukuoka University, Fukuoka 8140180, Japan
| | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, United States.
| |
Collapse
|
46
|
Narwade SC, Mallick BN, Deobagkar DD. Transcriptome Analysis Reveals Altered Expression of Memory and Neurotransmission Associated Genes in the REM Sleep Deprived Rat Brain. Front Mol Neurosci 2017; 10:67. [PMID: 28367113 PMCID: PMC5355427 DOI: 10.3389/fnmol.2017.00067] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 02/28/2017] [Indexed: 12/21/2022] Open
Abstract
Sleep disorders are associated with cognitive impairment. Selective rapid eye movement sleep (REMS) deprivation (REMSD) alters several physiological processes and behaviors. By employing NGS platform we carried out transcriptomic analysis in brain samples of control rats and those exposed to REMSD. The expression of genes involved in chromatin assembly, methylation, learning, memory, regulation of synaptic transmission, neuronal plasticity and neurohypophysial hormone synthesis were altered. Increased transcription of BMP4, DBH and ATP1B2 genes after REMSD supports our earlier findings and hypothesis. Alteration in the transcripts encoding histone subtypes and important players in chromatin remodeling was observed. The mRNAs which transcribe neurotransmitters such as OXT, AVP, PMCH and LNPEP and two small non-coding RNAs, namely RMRP and BC1 were down regulated. At least some of these changes are likely to regulate REMS and may participate in the consequences of REMS loss. Thus, the findings of this study have identified key epigenetic regulators and neuronal plasticity genes associated to REMS and its loss. This analysis provides a background and opens up avenues for unraveling their specific roles in the complex behavioral network particularly in relation to sustained REMS-loss associated changes.
Collapse
Affiliation(s)
- Santosh C Narwade
- Molecular Biology Research Laboratory, Center of Advanced Studies, Department of Zoology, Savitribai Phule Pune University Pune, India
| | | | - Deepti D Deobagkar
- Molecular Biology Research Laboratory, Center of Advanced Studies, Department of Zoology, Savitribai Phule Pune UniversityPune, India; Bioinformatics Center, Savitribai Phule Pune UniversityPune, India
| |
Collapse
|
47
|
Bastianini S, Alvente S, Berteotti C, Lo Martire V, Silvani A, Swoap SJ, Valli A, Zoccoli G, Cohen G. Accurate discrimination of the wake-sleep states of mice using non-invasive whole-body plethysmography. Sci Rep 2017; 7:41698. [PMID: 28139776 PMCID: PMC5282481 DOI: 10.1038/srep41698] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 12/29/2016] [Indexed: 01/11/2023] Open
Abstract
A major limitation in the study of sleep breathing disorders in mouse models of pathology is the need to combine whole-body plethysmography (WBP) to measure respiration with electroencephalography/electromyography (EEG/EMG) to discriminate wake-sleep states. However, murine wake-sleep states may be discriminated from breathing and body movements registered by the WBP signal alone. Our goal was to compare the EEG/EMG-based and the WBP-based scoring of wake-sleep states of mice, and provide formal guidelines for the latter. EEG, EMG, blood pressure and WBP signals were simultaneously recorded from 20 mice. Wake-sleep states were scored based either on EEG/EMG or on WBP signals and sleep-dependent respiratory and cardiovascular estimates were calculated. We found that the overall agreement between the 2 methods was 90%, with a high Cohen's Kappa index (0.82). The inter-rater agreement between 2 experts and between 1 expert and 1 naïve sleep investigators gave similar results. Sleep-dependent respiratory and cardiovascular estimates did not depend on the scoring method. We show that non-invasive discrimination of the wake-sleep states of mice based on visual inspection of the WBP signal is accurate, reliable and reproducible. This work may set the stage for non-invasive high-throughput experiments evaluating sleep and breathing patterns on mouse models of pathophysiology.
Collapse
Affiliation(s)
- Stefano Bastianini
- Prism Lab, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, I-40126, Italy
| | - Sara Alvente
- Prism Lab, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, I-40126, Italy
| | - Chiara Berteotti
- Prism Lab, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, I-40126, Italy
| | - Viviana Lo Martire
- Prism Lab, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, I-40126, Italy
| | - Alessandro Silvani
- Prism Lab, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, I-40126, Italy
| | - Steven J Swoap
- Department of Biology, Williams College, Williamstown, Massachusetts, MA 01267, USA
| | - Alice Valli
- Prism Lab, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, I-40126, Italy
| | - Giovanna Zoccoli
- Prism Lab, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, I-40126, Italy
| | - Gary Cohen
- Department of Women's and Children's Health, Neonatal Unit, Karolinska Institutet, Stockholm, 17176, Sweden
| |
Collapse
|
48
|
Activity of Tachykinin1-Expressing Pet1 Raphe Neurons Modulates the Respiratory Chemoreflex. J Neurosci 2017; 37:1807-1819. [PMID: 28073937 DOI: 10.1523/jneurosci.2316-16.2016] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 11/30/2016] [Accepted: 12/30/2016] [Indexed: 11/21/2022] Open
Abstract
Homeostatic control of breathing, heart rate, and body temperature relies on circuits within the brainstem modulated by the neurotransmitter serotonin (5-HT). Mounting evidence points to specialized neuronal subtypes within the serotonergic neuronal system, borne out in functional studies, for the modulation of distinct facets of homeostasis. Such functional differences, read out at the organismal level, are likely subserved by differences among 5-HT neuron subtypes at the cellular and molecular levels, including differences in the capacity to coexpress other neurotransmitters such as glutamate, GABA, thyrotropin releasing hormone, and substance P encoded by the Tachykinin-1 (Tac1) gene. Here, we characterize in mice a 5-HT neuron subtype identified by expression of Tac1 and the serotonergic transcription factor gene Pet1, referred to as the Tac1-Pet1 neuron subtype. Transgenic cell labeling showed Tac1-Pet1 soma resident largely in the caudal medulla. Chemogenetic [clozapine-N-oxide (CNO)-hM4Di] perturbation of Tac1-Pet1 neuron activity blunted the ventilatory response of the respiratory CO2 chemoreflex, which normally augments ventilation in response to hypercapnic acidosis to restore normal pH and PCO2Tac1-Pet1 axonal boutons were found localized to brainstem areas implicated in respiratory modulation, with highest density in motor regions. These findings demonstrate that the activity of a Pet1 neuron subtype with the potential to release both 5-HT and substance P is necessary for normal respiratory dynamics, perhaps via motor outputs that engage muscles of respiration and maintain airway patency. These Tac1-Pet1 neurons may act downstream of Egr2-Pet1 serotonergic neurons, which were previously established in respiratory chemoreception, but do not innervate respiratory motor nuclei.SIGNIFICANCE STATEMENT Serotonin (5-HT) neurons modulate physiological processes and behaviors as diverse as body temperature, respiration, aggression, and mood. Using genetic tools, we characterize a 5-HT neuron subtype defined by expression of Tachykinin1 and Pet1 (Tac1-Pet1 neurons), mapping soma localization to the caudal medulla primarily and axonal projections to brainstem motor nuclei most prominently, and, when silenced, observed blunting of the ventilatory response to inhaled CO2Tac1-Pet1 neurons thus appear distinct from and contrast previously described Egr2-Pet1 neurons, which project primarily to chemosensory integration centers and are themselves chemosensitive.
Collapse
|
49
|
Carrive P, Kuwaki T. Orexin and Central Modulation of Cardiovascular and Respiratory Function. Curr Top Behav Neurosci 2017; 33:157-196. [PMID: 27909989 DOI: 10.1007/7854_2016_46] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Orexin makes an important contribution to the regulation of cardiorespiratory function. When injected centrally under anesthesia, orexin increases blood pressure, heart rate, sympathetic nerve activity, and the amplitude and frequency of respiration. This is consistent with the location of orexin neurons in the hypothalamus and the distribution of orexin terminals at all levels of the central autonomic and respiratory network. These cardiorespiratory responses are components of arousal and are necessary to allow the expression of motivated behaviors. Thus, orexin contributes to the cardiorespiratory response to acute stressors, especially those of a psychogenic nature. Consequently, upregulation of orexin signaling, whether it is spontaneous or environmentally induced, can increase blood pressure and lead to hypertension, as is the case for the spontaneously hypertensive rat and the hypertensive BPH/2J Schlager mouse. Blockade of orexin receptors will reduce blood pressure in these animals, which could be a new pharmacological approach for the treatment of some forms of hypertension. Orexin can also magnify the respiratory reflex to hypercapnia in order to maintain respiratory homeostasis, and this may be in part why it is upregulated during obstructive sleep apnea. In this pathological condition, blockade of orexin receptors would make the apnea worse. To summarize, orexin is an important modulator of cardiorespiratory function. Acting on orexin signaling may help in the treatment of some cardiovascular and respiratory disorders.
Collapse
Affiliation(s)
- Pascal Carrive
- School of Medical Sciences, University of New South Wales, Sydney, NSW, 2052, Australia.
| | - Tomoyuki Kuwaki
- Department of Physiology, Graduate School of Medical & Dental Sciences, Kagoshima University, Kagoshima, Japan
| |
Collapse
|
50
|
Kubin L. Neural Control of the Upper Airway: Respiratory and State-Dependent Mechanisms. Compr Physiol 2016; 6:1801-1850. [PMID: 27783860 DOI: 10.1002/cphy.c160002] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Upper airway muscles subserve many essential for survival orofacial behaviors, including their important role as accessory respiratory muscles. In the face of certain predisposition of craniofacial anatomy, both tonic and phasic inspiratory activation of upper airway muscles is necessary to protect the upper airway against collapse. This protective action is adequate during wakefulness, but fails during sleep which results in recurrent episodes of hypopneas and apneas, a condition known as the obstructive sleep apnea syndrome (OSA). Although OSA is almost exclusively a human disorder, animal models help unveil the basic principles governing the impact of sleep on breathing and upper airway muscle activity. This article discusses the neuroanatomy, neurochemistry, and neurophysiology of the different neuronal systems whose activity changes with sleep-wake states, such as the noradrenergic, serotonergic, cholinergic, orexinergic, histaminergic, GABAergic and glycinergic, and their impact on central respiratory neurons and upper airway motoneurons. Observations of the interactions between sleep-wake states and upper airway muscles in healthy humans and OSA patients are related to findings from animal models with normal upper airway, and various animal models of OSA, including the chronic-intermittent hypoxia model. Using a framework of upper airway motoneurons being under concurrent influence of central respiratory, reflex and state-dependent inputs, different neurotransmitters, and neuropeptides are considered as either causing a sleep-dependent withdrawal of excitation from motoneurons or mediating an active, sleep-related inhibition of motoneurons. Information about the neurochemistry of state-dependent control of upper airway muscles accumulated to date reveals fundamental principles and may help understand and treat OSA. © 2016 American Physiological Society. Compr Physiol 6:1801-1850, 2016.
Collapse
Affiliation(s)
- Leszek Kubin
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|