1
|
Moe AAK. Cracking the Code of the Jugular Vagal Sensory Neurons in Allergic Airway Responsiveness. Am J Respir Cell Mol Biol 2025; 72:346-348. [PMID: 39471332 PMCID: PMC12005020 DOI: 10.1165/rcmb.2024-0445ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 10/23/2024] [Indexed: 11/01/2024] Open
Affiliation(s)
- Aung Aung Kywe Moe
- Department of Medical Imaging and Radiation Sciences Monash University School of Primary and Allied Health Care Clayton, Victoria, Australia
- Department of Anatomy and Physiology University of Melbourne School of Biomedical Sciences Parkville, Victoria, Australia
| |
Collapse
|
2
|
Xing Y, Nho Y, Lawson K, Zhu Y, Ellison AE, Chang MY, Hancock W, Han L. MrgprC11 + Jugular Neurons Control Airway Hyperresponsiveness in Allergic Airway Inflammation. Am J Respir Cell Mol Biol 2025; 72:393-407. [PMID: 39405479 PMCID: PMC12005045 DOI: 10.1165/rcmb.2024-0153oc] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 10/15/2024] [Indexed: 04/02/2025] Open
Abstract
The lung is densely innervated by sensory nerves, the majority of which are derived from the vagal sensory neurons. Vagal ganglia consist of two different ganglia, termed nodose and jugular ganglia, with distinct embryonic origins, innervation patterns, and physiological functions in the periphery. Because nodose neurons constitute the majority of the vagal ganglia, our understanding of the function of jugular nerves in the lung is very limited. This study aims to investigate the role of MrgprC11+ jugular sensory neurons in a mouse allergic asthma model. Our previous study has shown that MrgprC11+ jugular neurons mediate cholinergic bronchoconstriction. In this study, we found that, in addition to MrgprC11, several other Mrgpr family members, including MrgprA3, MrgprB4, and MrgprD, are also specifically expressed in the jugular sensory neurons. MrgprC11+ jugular neurons exhibit dense innervation in the respiratory tract, including the larynx, trachea, proximal bronchus, and distal bronchus. We also found that receptors for IL-4 and oncostatin M, two critical cytokines promoting allergic airway inflammation, are mainly expressed in jugular sensory neurons. Both IL-4 and oncostatin M can sensitize the neuronal responses of MrgprC11+ jugular neurons. Moreover, ablation of MrgprC11+ neurons significantly inhibited airway hyperresponsiveness in the asthmatic lung, demonstrating the critical role of MrgprC11+ neurons in controlling airway constriction. Our results emphasize the critical role of jugular sensory neurons in respiratory diseases and present MrgprC11+ neurons as a potential therapeutic target for treating airway hyperresponsiveness.
Collapse
Affiliation(s)
- Yanyan Xing
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia
| | - Yeseul Nho
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia
| | - Katy Lawson
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia
| | - Yuyan Zhu
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia
| | - Alexandra E Ellison
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia
| | - Margaret Y Chang
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia
| | - William Hancock
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia
| | - Liang Han
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia
| |
Collapse
|
3
|
Li J, Liu Y. Vagal sensory circuits of the lower airway in respiratory physiology: Insights from neuronal diversity. Curr Opin Neurobiol 2025; 92:103000. [PMID: 40101474 DOI: 10.1016/j.conb.2025.103000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 01/22/2025] [Accepted: 02/20/2025] [Indexed: 03/20/2025]
Abstract
Sensory neurons innervating the lower airway provide essential feedback information that regulates respiratory physiology. These neurons synapse with second-order neurons in the central nervous system, which project directly or indirectly to the respiratory and autonomic centers. Both primary sensory neurons and second-order neurons within these circuits exhibit significant heterogeneity, and the precise roles of individual neuronal subtypes in coding the airway's internal states and modulating respiratory and autonomic outputs remain incompletely understood. In this review, we summarize recent advances in understanding the neuronal diversity along sensory circuits of the lower airway and their physiological functions. We also highlight the challenges in elucidating the roles of specific neuronal subtypes due to the extensive molecular and anatomical diversity among these neurons. Improving targeting specificity for neuronal manipulation, combined with the development of a comprehensive connectivity map, will be critical for revealing the coding and wiring logics that underlie the precise control of respiratory physiology.
Collapse
Affiliation(s)
- Jie Li
- HHMI/Janelia Research Campus, Ashburn, VA 20147, USA
| | - Yin Liu
- HHMI/Janelia Research Campus, Ashburn, VA 20147, USA.
| |
Collapse
|
4
|
Jiang Y, Huang Z, Sun W, Huang J, Xu Y, Liao Y, Jin T, Li Q, Ho IHT, Zou Y, Zhu W, Li Q, Qin F, Zhang X, Shi S, Zhang N, Yang S, Xie W, Wu S, Tan L, Zhang L, Chen H, Gin T, Chan MTV, Wu WKK, Xiao L, Liu X. Roseburia intestinalis-derived butyrate alleviates neuropathic pain. Cell Host Microbe 2025; 33:104-118.e7. [PMID: 39706182 DOI: 10.1016/j.chom.2024.11.013] [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: 04/02/2024] [Revised: 10/30/2024] [Accepted: 11/25/2024] [Indexed: 12/23/2024]
Abstract
Approximately 20% of patients with shingles develop postherpetic neuralgia (PHN). We investigated the role of gut microbiota in shingle- and PHN-related pain. Patients with shingles or PHN exhibited significant alterations in their gut microbiota with microbial markers predicting PHN development among patients with shingles. Functionally, fecal microbiota transplantation from patients with PHN to mice heightened pain sensitivity. Administration of Roseburia intestinalis, a bacterium both depleted in patients with shingles and PHN, alleviated peripheral nerve injury-induced pain in mice. R. intestinalis enhanced vagal neurotransmission to the nucleus tractus solitarius (NTS) to suppress the central amygdala (CeA), a brain region involved in pain perception. R. intestinalis-generated butyrate activated vagal neurons through the receptor, G protein-coupled receptor 41 (GPR41). Vagal knockout of Gpr41 abolished the effects of R. intestinalis on the NTS-CeA circuit and reduced pain behaviors. Overall, we established a microbiota-based model for PHN risk assessment and identified R. intestinalis as a potential pain-alleviating probiotic.
Collapse
Affiliation(s)
- Yanjun Jiang
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ziheng Huang
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Wuping Sun
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, Shenzhen Nanshan People's Hospital and the 6th Affiliated Hospital of Shenzhen University Medical School, National Key Clinical Pain Medicine of China, Shenzhen 518060, China
| | - Jiabin Huang
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, Shenzhen Nanshan People's Hospital and the 6th Affiliated Hospital of Shenzhen University Medical School, National Key Clinical Pain Medicine of China, Shenzhen 518060, China
| | - Yunlong Xu
- Shenzhen Key Laboratory of Drug Addiction, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Department of Neonatology, Shenzhen Maternity & Child Healthcare Hospital, The First School of Clinical Medicine, Southern Medical University, Shenzhen 518055, China
| | - Yuliang Liao
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, Shenzhen Nanshan People's Hospital and the 6th Affiliated Hospital of Shenzhen University Medical School, National Key Clinical Pain Medicine of China, Shenzhen 518060, China
| | - Tingting Jin
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Qing Li
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China; Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Idy Hiu Ting Ho
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yidan Zou
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Wenyi Zhu
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Qian Li
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Fenfen Qin
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Xinyi Zhang
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Shuqi Shi
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, Shenzhen Nanshan People's Hospital and the 6th Affiliated Hospital of Shenzhen University Medical School, National Key Clinical Pain Medicine of China, Shenzhen 518060, China
| | - Na Zhang
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Shaomin Yang
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, Shenzhen Nanshan People's Hospital and the 6th Affiliated Hospital of Shenzhen University Medical School, National Key Clinical Pain Medicine of China, Shenzhen 518060, China
| | - Wenhui Xie
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, Shenzhen Nanshan People's Hospital and the 6th Affiliated Hospital of Shenzhen University Medical School, National Key Clinical Pain Medicine of China, Shenzhen 518060, China
| | - Songbin Wu
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, Shenzhen Nanshan People's Hospital and the 6th Affiliated Hospital of Shenzhen University Medical School, National Key Clinical Pain Medicine of China, Shenzhen 518060, China
| | - Likai Tan
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Lin Zhang
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Microbiota I-Center (MagIC), The Chinese University of Hong Kong, Hong Kong SAR, China; Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Huarong Chen
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Tony Gin
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Matthew Tak Vai Chan
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - William Ka Kei Wu
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China; State Key Laboratory of Digestive Disease Institute of Digestive Disease and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Lizu Xiao
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, Shenzhen Nanshan People's Hospital and the 6th Affiliated Hospital of Shenzhen University Medical School, National Key Clinical Pain Medicine of China, Shenzhen 518060, China.
| | - Xiaodong Liu
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong SAR, China; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
5
|
Lu H, Chen G, Zhao M, Gu H, Zheng W, Li X, Huang M, Geng D, Yu M, Guan X, Zhang L, Song H, Li Y, Wu M, Zhang F, Li D, Wu Q, Shang C, Xie Z, Cao P. Brainstem opioid peptidergic neurons regulate cough reflexes in mice. Innovation (N Y) 2024; 5:100721. [PMID: 39529953 PMCID: PMC11551472 DOI: 10.1016/j.xinn.2024.100721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024] Open
Abstract
Cough is a vital defensive reflex for expelling harmful substances from the airway. The sensory afferents for the cough reflex have been intensively studied. However, the brain mechanisms underlying the cough reflex remain poorly understood. Here, we developed a paradigm to quantitatively measure cough-like reflexes in mice. Using this paradigm, we found that prodynorphin-expressing (Pdyn+) neurons in the nucleus of the solitary tract (NTS) are critical for capsaicin-induced cough-like reflexes. These neurons receive cough-related neural signals from Trpv1+ vagal sensory neurons. The activation of Pdyn+ NTS neurons triggered respiratory responses resembling cough-like reflexes. Among the divergent projections of Pdyn+ NTS neurons, a glutamatergic pathway projecting to the caudal ventral respiratory group (cVRG), the canonical cough center, was necessary and sufficient for capsaicin-induced cough-like reflexes. These results reveal that Pdyn+ NTS neurons, as a key neuronal population at the entry point of the vagus nerve to the brainstem, initiate cough-like reflexes in mice.
Collapse
Affiliation(s)
- Haicheng Lu
- National Institute of Biological Sciences, Beijing 102206, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing 100084, China
| | - Guoqing Chen
- National Institute of Biological Sciences, Beijing 102206, China
| | - Miao Zhao
- National Institute of Biological Sciences, Beijing 102206, China
| | - Huating Gu
- National Institute of Biological Sciences, Beijing 102206, China
| | - Wenxuan Zheng
- National Institute of Biological Sciences, Beijing 102206, China
- Peking University–Tsinghua University–NIBS Joint Graduate Program, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Xiating Li
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China
| | - Meizhu Huang
- National Institute of Biological Sciences, Beijing 102206, China
| | - Dandan Geng
- Key Laboratory of Neural and Vascular Biology, Ministry of Education, Department of Biochemistry and Molecular Biology, Hebei Medical University, Shijiazhuang 050011, China
| | - Minhui Yu
- National Institute of Biological Sciences, Beijing 102206, China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Xuyan Guan
- National Institute of Biological Sciences, Beijing 102206, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing 100084, China
| | - Li Zhang
- National Institute of Biological Sciences, Beijing 102206, China
| | - Huimeng Song
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China
| | - Yaning Li
- Key Laboratory of Neural and Vascular Biology, Ministry of Education, Department of Biochemistry and Molecular Biology, Hebei Medical University, Shijiazhuang 050011, China
| | - Menghua Wu
- National Institute of Biological Sciences, Beijing 102206, China
| | - Fan Zhang
- Key Laboratory of Neural and Vascular Biology, Ministry of Education, Department of Biochemistry and Molecular Biology, Hebei Medical University, Shijiazhuang 050011, China
| | - Dapeng Li
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China
| | - Qingfeng Wu
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Congping Shang
- School of Basic Medical Sciences, Guangzhou National Laboratory, Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 510799, China
| | - Zhiyong Xie
- Department of Psychological Medicine, Zhongshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200433, China
| | - Peng Cao
- National Institute of Biological Sciences, Beijing 102206, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing 100084, China
| |
Collapse
|
6
|
Verzele NAJ, Chua BY, Short KR, Moe AAK, Edwards IN, Bielefeldt-Ohmann H, Hulme KD, Noye EC, Tong MZW, Reading PC, Trewella MW, Mazzone SB, McGovern AE. Evidence for vagal sensory neural involvement in influenza pathogenesis and disease. PLoS Pathog 2024; 20:e1011635. [PMID: 38626267 PMCID: PMC11051609 DOI: 10.1371/journal.ppat.1011635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 04/26/2024] [Accepted: 04/01/2024] [Indexed: 04/18/2024] Open
Abstract
Influenza A virus (IAV) is a common respiratory pathogen and a global cause of significant and often severe morbidity. Although inflammatory immune responses to IAV infections are well described, little is known about how neuroimmune processes contribute to IAV pathogenesis. In the present study, we employed surgical, genetic, and pharmacological approaches to manipulate pulmonary vagal sensory neuron innervation and activity in the lungs to explore potential crosstalk between pulmonary sensory neurons and immune processes. Intranasal inoculation of mice with H1N1 strains of IAV resulted in stereotypical antiviral lung inflammation and tissue pathology, changes in breathing, loss of body weight and other clinical signs of severe IAV disease. Unilateral cervical vagotomy and genetic ablation of pulmonary vagal sensory neurons had a moderate effect on the pulmonary inflammation induced by IAV infection, but significantly worsened clinical disease presentation. Inhibition of pulmonary vagal sensory neuron activity via inhalation of the charged sodium channel blocker, QX-314, resulted in a moderate decrease in lung pathology, but again this was accompanied by a paradoxical worsening of clinical signs. Notably, vagal sensory ganglia neuroinflammation was induced by IAV infection and this was significantly potentiated by QX-314 administration. This vagal ganglia hyperinflammation was characterized by alterations in IAV-induced host defense gene expression, increased neuropeptide gene and protein expression, and an increase in the number of inflammatory cells present within the ganglia. These data suggest that pulmonary vagal sensory neurons play a role in the regulation of the inflammatory process during IAV infection and suggest that vagal neuroinflammation may be an important contributor to IAV pathogenesis and clinical presentation. Targeting these pathways could offer therapeutic opportunities to treat IAV-induced morbidity and mortality.
Collapse
Affiliation(s)
- Nathalie A. J. Verzele
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia Queensland, Australia
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, Victoria, Australia
| | - Brendon Y. Chua
- The Peter Doherty Institute for Infection and Immunity, Department of Microbiology and Immunology, University of Melbourne, Melbourne, Victoria, Australia
| | - Kirsty R. Short
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia Queensland, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, St Lucia, Queensland, Australia
| | - Aung Aung Kywe Moe
- Department of Medical Imaging and Radiation Sciences, Monash University, Clayton, Victoria, Australia
| | - Isaac N. Edwards
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia Queensland, Australia
| | - Helle Bielefeldt-Ohmann
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia Queensland, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, St Lucia, Queensland, Australia
| | - Katina D. Hulme
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia Queensland, Australia
| | - Ellesandra C. Noye
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia Queensland, Australia
| | - Marcus Z. W. Tong
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia Queensland, Australia
| | - Patrick C. Reading
- The Peter Doherty Institute for Infection and Immunity, Department of Microbiology and Immunology, University of Melbourne, Melbourne, Victoria, Australia
- WHO Collaborating Centre for Reference and Research on Influenza, Victorian Infectious Disease Reference Laboratory, Peter Doherty Institute for Infection, and Immunity, 792 Elizabeth St., Melbourne, Victoria, Australia
| | - Matthew W. Trewella
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, Victoria, Australia
| | - Stuart B. Mazzone
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, Victoria, Australia
| | - Alice E. McGovern
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
7
|
Moe AAK, Bautista TG, Trewella MW, Korim WS, Yao ST, Behrens R, Driessen AK, McGovern AE, Mazzone SB. Investigation of vagal sensory neurons in mice using optical vagal stimulation and tracheal neuroanatomy. iScience 2024; 27:109182. [PMID: 38414860 PMCID: PMC10897902 DOI: 10.1016/j.isci.2024.109182] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 12/28/2023] [Accepted: 02/06/2024] [Indexed: 02/29/2024] Open
Abstract
In rats and guinea pigs, sensory innervation of the airways is derived largely from the vagus nerve, with the extrapulmonary airways innervated by Wnt1+ jugular neurons and the intrapulmonary airways and lungs by Phox2b+ nodose neurons; however, our knowledge of airway innervation in mice is limited. We used genetically targeted expression of enhanced yellow fluorescent protein-channelrhodopsin-2 (EYFP-ChR2) in Wnt1+ or Phox2b+ tissues to characterize jugular and nodose-mediated physiological responses and airway innervation in mice. With optical stimulation, Phox2b+ vagal fibers modulated cardiorespiratory function in a frequency-dependent manner while right Wnt1+ vagal fibers induced a small increase in respiratory rate. Mouse tracheae contained sparse Phox2b-EYFP fibers but dense networks of Wnt1-EYFP fibers. Retrograde tracing from the airways showed limited tracheal innervation by the jugular sensory neurons, distinct from other species. These differences in physiology and vagal sensory distribution have important implications when using mice for studying airway neurobiology.
Collapse
Affiliation(s)
- Aung Aung Kywe Moe
- Department of Anatomy and Physiology, School of Biomedical Sciences, The University of Melbourne, Parkville, VIC 3010, Australia
- Department of Medical Imaging and Radiation Sciences, School of Primary and Allied Health Care, Monash University, Clayton Campus, Clayton, VIC 3800, Australia
| | - Tara G Bautista
- Department of Anatomy and Physiology, School of Biomedical Sciences, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Matthew W Trewella
- Department of Anatomy and Physiology, School of Biomedical Sciences, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Willian S Korim
- Department of Anatomy and Physiology, School of Biomedical Sciences, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Song T Yao
- Department of Anatomy and Physiology, School of Biomedical Sciences, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Robert Behrens
- Department of Anatomy and Physiology, School of Biomedical Sciences, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Alexandria K Driessen
- Department of Anatomy and Physiology, School of Biomedical Sciences, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Alice E McGovern
- Department of Anatomy and Physiology, School of Biomedical Sciences, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Stuart B Mazzone
- Department of Anatomy and Physiology, School of Biomedical Sciences, The University of Melbourne, Parkville, VIC 3010, Australia
| |
Collapse
|
8
|
González-García M, Carrillo-Franco L, Morales-Luque C, Dawid-Milner MS, López-González MV. Central Autonomic Mechanisms Involved in the Control of Laryngeal Activity and Vocalization. BIOLOGY 2024; 13:118. [PMID: 38392336 PMCID: PMC10886357 DOI: 10.3390/biology13020118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 02/07/2024] [Accepted: 02/10/2024] [Indexed: 02/24/2024]
Abstract
In humans, speech is a complex process that requires the coordinated involvement of various components of the phonatory system, which are monitored by the central nervous system. The larynx in particular plays a crucial role, as it enables the vocal folds to meet and converts the exhaled air from our lungs into audible sounds. Voice production requires precise and sustained exhalation, which generates an air pressure/flow that creates the pressure in the glottis required for voice production. Voluntary vocal production begins in the laryngeal motor cortex (LMC), a structure found in all mammals, although the specific location in the cortex varies in humans. The LMC interfaces with various structures of the central autonomic network associated with cardiorespiratory regulation to allow the perfect coordination between breathing and vocalization. The main subcortical structure involved in this relationship is the mesencephalic periaqueductal grey matter (PAG). The PAG is the perfect link to the autonomic pontomedullary structures such as the parabrachial complex (PBc), the Kölliker-Fuse nucleus (KF), the nucleus tractus solitarius (NTS), and the nucleus retroambiguus (nRA), which modulate cardiovascular autonomic function activity in the vasomotor centers and respiratory activity at the level of the generators of the laryngeal-respiratory motor patterns that are essential for vocalization. These cores of autonomic structures are not only involved in the generation and modulation of cardiorespiratory responses to various stressors but also help to shape the cardiorespiratory motor patterns that are important for vocal production. Clinical studies show increased activity in the central circuits responsible for vocalization in certain speech disorders, such as spasmodic dysphonia because of laryngeal dystonia.
Collapse
Affiliation(s)
- Marta González-García
- Department of Human Physiology, Faculty of Medicine, University of Málaga, 29010 Málaga, Spain
- Unit of Neurophysiology of the Autonomic Nervous System (CIMES), University of Málaga, 29010 Málaga, Spain
- Biomedical Research Institute of Málaga (IBIMA Plataforma BIONAND), 29010 Málaga, Spain
| | - Laura Carrillo-Franco
- Department of Human Physiology, Faculty of Medicine, University of Málaga, 29010 Málaga, Spain
- Unit of Neurophysiology of the Autonomic Nervous System (CIMES), University of Málaga, 29010 Málaga, Spain
- Biomedical Research Institute of Málaga (IBIMA Plataforma BIONAND), 29010 Málaga, Spain
| | - Carmen Morales-Luque
- Department of Human Physiology, Faculty of Medicine, University of Málaga, 29010 Málaga, Spain
| | - Marc Stefan Dawid-Milner
- Department of Human Physiology, Faculty of Medicine, University of Málaga, 29010 Málaga, Spain
- Unit of Neurophysiology of the Autonomic Nervous System (CIMES), University of Málaga, 29010 Málaga, Spain
- Biomedical Research Institute of Málaga (IBIMA Plataforma BIONAND), 29010 Málaga, Spain
| | - Manuel Víctor López-González
- Department of Human Physiology, Faculty of Medicine, University of Málaga, 29010 Málaga, Spain
- Unit of Neurophysiology of the Autonomic Nervous System (CIMES), University of Málaga, 29010 Málaga, Spain
- Biomedical Research Institute of Málaga (IBIMA Plataforma BIONAND), 29010 Málaga, Spain
| |
Collapse
|
9
|
Huo L, Ye Z, Liu M, He Z, Huang M, Li D, Wu Q, Wang Q, Wang X, Cao P, Dong J, Shang C. Brain circuits for retching-like behavior. Natl Sci Rev 2024; 11:nwad256. [PMID: 38288368 PMCID: PMC10824557 DOI: 10.1093/nsr/nwad256] [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: 05/26/2023] [Revised: 09/06/2023] [Accepted: 09/24/2023] [Indexed: 01/31/2024] Open
Abstract
Nausea and vomiting are important defensive responses to cope with pathogens and toxins that invade the body. The nucleus of the solitary tract (NTS) is important for initiating these responses. However, the molecular heterogeneities and cellular diversities of the NTS occlude a better understanding of these defensive responses. Here, we constructed the single-nucleus transcriptomic atlas of NTS cells and found multiple populations of NTS neurons that may be involved in these defensive responses. Among these, we identified Calbindin1-positive (Calb1+) NTS neurons that are molecularly distinct from Tac1+ neurons. These Calb1+ neurons are critical for nausea and retching induced by cereulide; an emetic toxin secreted by Bacillus Cereus. Strikingly, we found that cereulide can directly modulate vagal sensory neurons that innervate Calb1+ NTS neurons, a novel mechanism distinct from that for nausea and retching induced by Staphylococcal enterotoxin A. Together, our transcriptomic atlas of NTS neurons and the functional analyses revealed the neural mechanism for cereulide-induced retching-like behavior. These results demonstrate the molecular and cellular complexities in the brain that underlie defensive responses to the diversities of pathogens and toxins.
Collapse
Affiliation(s)
- Lifang Huo
- School of Basic Medical Sciences, Guangzhou National Laboratory, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 510799, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou 510320, China
| | - Zhimin Ye
- School of Basic Medical Sciences, Guangzhou National Laboratory, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 510799, China
| | - Meiling Liu
- School of Basic Medical Sciences, Guangzhou National Laboratory, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 510799, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou 510320, China
| | - Ziqing He
- School of Basic Medical Sciences, Guangzhou National Laboratory, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 510799, China
| | - Meizhu Huang
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou 510320, China
| | - Dapeng Li
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China
| | - Qian Wu
- State Key Laboratory of Cognitive Neuroscience and Learning, IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China
| | - Qian Wang
- Changping Life Science Laboratory, Beijing 102299, China
| | - Xiaoqun Wang
- State Key Laboratory of Cognitive Neuroscience and Learning, IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China
| | - Peng Cao
- National Institute of Biological Sciences, Beijing 102206, China
| | - Ji Dong
- School of Basic Medical Sciences, Guangzhou National Laboratory, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 510799, China
| | - Congping Shang
- School of Basic Medical Sciences, Guangzhou National Laboratory, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 510799, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou 510320, China
| |
Collapse
|
10
|
Trevizan-Bau P, Mazzone SB. Neuroimmune pathways regulating airway inflammation. Ann Allergy Asthma Immunol 2023; 131:550-560. [PMID: 37517657 DOI: 10.1016/j.anai.2023.07.020] [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: 04/24/2023] [Revised: 05/24/2023] [Accepted: 07/20/2023] [Indexed: 08/01/2023]
Abstract
Airways diseases are typically accompanied by inflammation, which has long been known to contribute to obstruction, mucus hypersecretion, dyspnea, cough, and other characteristic symptoms displayed in patients. Clinical interventions, therefore, often target inflammation to reverse lung pathology and reduce morbidity. The airways and lungs are densely innervated by subsets of nerve fibers, which are not only impacted by pulmonary inflammation but, in addition, likely serve as important regulators of immune cell function. This bidirectional neuroimmune crosstalk is supported by close spatial relationships between immune cells and airway nerve fibers, complementary neural and immune signaling pathways, local specialized airway chemosensory cells, and dedicated reflex circuits. In this article, we review the recent literature on this topic and present state-of-the-art evidence supporting the role of neuroimmune interactions in airway inflammation. In addition, we extend this evidence to synthesize considerations for the clinical translation of these discoveries to improve the management of patients with airway disease.
Collapse
Affiliation(s)
- Pedro Trevizan-Bau
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Victoria, Australia; Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Stuart B Mazzone
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia.
| |
Collapse
|
11
|
Behrens R, Dutschmann M, Trewella M, Mazzone SB, Moe AAK. Regulation of vagally-evoked respiratory responses by the lateral parabrachial nucleus in the mouse. Respir Physiol Neurobiol 2023; 316:104141. [PMID: 37597796 DOI: 10.1016/j.resp.2023.104141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/04/2023] [Accepted: 08/14/2023] [Indexed: 08/21/2023]
Abstract
Vagal sensory inputs to the brainstem can alter breathing through the modulation of pontomedullary respiratory circuits. In this study, we set out to investigate the localised effects of modulating lateral parabrachial nucleus (LPB) activity on vagally-evoked changes in breathing pattern. In isoflurane-anaesthetised and instrumented mice, electrical stimulation of the vagus nerve (eVNS) produced stimulation frequency-dependent changes in diaphragm electromyograph (dEMG) activity with an evoked tachypnoea and apnoea at low and high stimulation frequencies, respectively. Muscimol microinjections into the LPB significantly attenuated eVNS-evoked respiratory rate responses. Notably, muscimol injections reaching the caudal LPB, previously unrecognised for respiratory modulation, potently modulated eVNS-evoked apnoea, whilst muscimol injections reaching the intermediate LPB selectively modulated the eVNS-evoked tachypnoea. The effects of muscimol on eVNS-evoked breathing rate changes occurred without altering basal eupneic breathing. These results highlight novel roles for the LPB in regulating vagally-evoked respiratory reflexes.
Collapse
Affiliation(s)
- Robert Behrens
- Department of Anatomy and Physiology, University of Melbourne, VIC, Australia
| | - Mathias Dutschmann
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Matthew Trewella
- Department of Anatomy and Physiology, University of Melbourne, VIC, Australia
| | - Stuart B Mazzone
- Department of Anatomy and Physiology, University of Melbourne, VIC, Australia.
| | - Aung Aung Kywe Moe
- Department of Anatomy and Physiology, University of Melbourne, VIC, Australia; Department of Medical Imaging and Radiation Sciences, Monash University, Clayton, Australia
| |
Collapse
|
12
|
Fischer KB, Collins HK, Pang Y, Roy DS, Zhang Y, Feng G, Li SJ, Kepecs A, Callaway EM. Monosynaptic restriction of the anterograde herpes simplex virus strain H129 for neural circuit tracing. J Comp Neurol 2023; 531:584-595. [PMID: 36606699 PMCID: PMC10040246 DOI: 10.1002/cne.25451] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 11/09/2022] [Accepted: 12/13/2022] [Indexed: 01/07/2023]
Abstract
Identification of synaptic partners is a fundamental task for systems neuroscience. To date, few reliable techniques exist for whole brain labeling of downstream synaptic partners in a cell-type-dependent and monosynaptic manner. Herein, we describe a novel monosynaptic anterograde tracing system based on the deletion of the gene UL6 from the genome of a cre-dependent version of the anterograde Herpes Simplex Virus 1 strain H129. Given that this knockout blocks viral genome packaging and thus viral spread, we reasoned that co-infection of a HSV H129 ΔUL6 virus with a recombinant adeno-associated virus expressing UL6 in a cre-dependent manner would result in monosynaptic spread from target cre-expressing neuronal populations. Application of this system to five nonreciprocal neural circuits resulted in labeling of neurons in expected projection areas. While some caveats may preclude certain applications, this system provides a reliable method to label postsynaptic partners in a brain-wide fashion.
Collapse
Affiliation(s)
- Kyle B Fischer
- Systems Neurobiology Laboratories, Salk Institute for Biological Studies, La Jolla, California, USA
| | - Hannah K Collins
- Systems Neurobiology Laboratories, Salk Institute for Biological Studies, La Jolla, California, USA
| | - Yan Pang
- Systems Neurobiology Laboratories, Salk Institute for Biological Studies, La Jolla, California, USA
| | - Dheeraj S Roy
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Ying Zhang
- Department of Brain and Cognitive Sciences, McGovern Institute for Brain Research at MIT, Cambridge, Massachusetts, USA
| | - Guoping Feng
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Brain and Cognitive Sciences, McGovern Institute for Brain Research at MIT, Cambridge, Massachusetts, USA
| | - Shu-Jing Li
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, USA
| | - Adam Kepecs
- Departments of Neuroscience and Psychiatry, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Edward M Callaway
- Systems Neurobiology Laboratories, Salk Institute for Biological Studies, La Jolla, California, USA
| |
Collapse
|
13
|
Krohn F, Novello M, van der Giessen RS, De Zeeuw CI, Pel JJM, Bosman LWJ. The integrated brain network that controls respiration. eLife 2023; 12:83654. [PMID: 36884287 PMCID: PMC9995121 DOI: 10.7554/elife.83654] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 01/29/2023] [Indexed: 03/09/2023] Open
Abstract
Respiration is a brain function on which our lives essentially depend. Control of respiration ensures that the frequency and depth of breathing adapt continuously to metabolic needs. In addition, the respiratory control network of the brain has to organize muscular synergies that integrate ventilation with posture and body movement. Finally, respiration is coupled to cardiovascular function and emotion. Here, we argue that the brain can handle this all by integrating a brainstem central pattern generator circuit in a larger network that also comprises the cerebellum. Although currently not generally recognized as a respiratory control center, the cerebellum is well known for its coordinating and modulating role in motor behavior, as well as for its role in the autonomic nervous system. In this review, we discuss the role of brain regions involved in the control of respiration, and their anatomical and functional interactions. We discuss how sensory feedback can result in adaptation of respiration, and how these mechanisms can be compromised by various neurological and psychological disorders. Finally, we demonstrate how the respiratory pattern generators are part of a larger and integrated network of respiratory brain regions.
Collapse
Affiliation(s)
- Friedrich Krohn
- Department of Neuroscience, Erasmus MC, Rotterdam, Netherlands
| | - Manuele Novello
- Department of Neuroscience, Erasmus MC, Rotterdam, Netherlands
| | | | - Chris I De Zeeuw
- Department of Neuroscience, Erasmus MC, Rotterdam, Netherlands.,Netherlands Institute for Neuroscience, Royal Academy of Arts and Sciences, Amsterdam, Netherlands
| | - Johan J M Pel
- Department of Neuroscience, Erasmus MC, Rotterdam, Netherlands
| | | |
Collapse
|
14
|
Bin NR, Prescott SL, Horio N, Wang Y, Chiu IM, Liberles SD. An airway-to-brain sensory pathway mediates influenza-induced sickness. Nature 2023; 615:660-667. [PMID: 36890237 PMCID: PMC10033449 DOI: 10.1038/s41586-023-05796-0] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 02/03/2023] [Indexed: 03/10/2023]
Abstract
Pathogen infection causes a stereotyped state of sickness that involves neuronally orchestrated behavioural and physiological changes1,2. On infection, immune cells release a 'storm' of cytokines and other mediators, many of which are detected by neurons3,4; yet, the responding neural circuits and neuro-immune interaction mechanisms that evoke sickness behaviour during naturalistic infections remain unclear. Over-the-counter medications such as aspirin and ibuprofen are widely used to alleviate sickness and act by blocking prostaglandin E2 (PGE2) synthesis5. A leading model is that PGE2 crosses the blood-brain barrier and directly engages hypothalamic neurons2. Here, using genetic tools that broadly cover a peripheral sensory neuron atlas, we instead identified a small population of PGE2-detecting glossopharyngeal sensory neurons (petrosal GABRA1 neurons) that are essential for influenza-induced sickness behaviour in mice. Ablating petrosal GABRA1 neurons or targeted knockout of PGE2 receptor 3 (EP3) in these neurons eliminates influenza-induced decreases in food intake, water intake and mobility during early-stage infection and improves survival. Genetically guided anatomical mapping revealed that petrosal GABRA1 neurons project to mucosal regions of the nasopharynx with increased expression of cyclooxygenase-2 after infection, and also display a specific axonal targeting pattern in the brainstem. Together, these findings reveal a primary airway-to-brain sensory pathway that detects locally produced prostaglandins and mediates systemic sickness responses to respiratory virus infection.
Collapse
Affiliation(s)
- Na-Ryum Bin
- Howard Hughes Medical Institute, Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Sara L Prescott
- Howard Hughes Medical Institute, Department of Cell Biology, Harvard Medical School, Boston, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Nao Horio
- Howard Hughes Medical Institute, Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Yandan Wang
- Howard Hughes Medical Institute, Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Isaac M Chiu
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Stephen D Liberles
- Howard Hughes Medical Institute, Department of Cell Biology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
15
|
An NTS-CeA projection modulates depression-like behaviors in a mouse model of chronic pain. Neurobiol Dis 2022; 174:105893. [DOI: 10.1016/j.nbd.2022.105893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 09/28/2022] [Accepted: 10/08/2022] [Indexed: 11/22/2022] Open
|
16
|
O'Dell DE, Smith-Bell CA, Enquist LW, Engel EA, Schreurs BG. Eyeblink tract tracing with two strains of herpes simplex virus 1. Brain Res 2022; 1793:148040. [PMID: 35932812 DOI: 10.1016/j.brainres.2022.148040] [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: 03/31/2022] [Revised: 07/20/2022] [Accepted: 08/01/2022] [Indexed: 11/18/2022]
Abstract
BACKGROUND Neuroinvasive herpes simplex-1 (HSV-1) isolates including H129 and McIntyre cross at or near synapses labeling higher-order neurons directly connected to infected cells. H129 spreads predominately in the anterograde direction while McIntyre strains spread only in the retrograde direction. However, it is unknown if neurons are functional once infected with derivatives of H129 or McIntyre. NEW METHOD We describe a previously unpublished HSV-1 recombinant derived from H129 (HSV-373) expressing mCherry fluorescent reporters and one new McIntyre recombinant (HSV-780) expressing the mCherry fluorophore and demonstrate how infections affect neuron viability. RESULTS AND COMPARISON WITH EXISTING METHODS Each recombinant virus behaved similarly and spread to the target 4 days post-infection. We tested H129 recombinant infected neurons for neurodegeneration using Fluoro-jade C and found them to be necrotic as a result of viral infection. We performed dual inoculations with both HSV-772 and HSV-780 to identify cells comprising both the anterograde pathway and the retrograde pathway, respectively, of our circuit of study. We examined the presence of postsynaptic marker PSD-95, which plays a role in synaptic plasticity, in HSV-772 infected and in dual-infected rats (HSV-772 and HSV-780). PSD-95 reactivity decreased in HSV-772-infected neurons and dual-infected tissue had no PSD-95 reactivity. CONCLUSIONS Infection by these new recombinant viruses traced the circuit of interest but functional studies of the cells comprising the pathway were not possible because viral-infected neurons died as a result of necrosis or were stripped of PSD-95 by the time the viral labels reached the target.
Collapse
Affiliation(s)
- Deidre E O'Dell
- Department of Neuroscience, Rockefeller Neuroscience Institute, United States; West Virginia University, Morgantown, WV 26505, United States.
| | - Carrie A Smith-Bell
- Department of Neuroscience, Rockefeller Neuroscience Institute, United States; West Virginia University, Morgantown, WV 26505, United States
| | - Lynn W Enquist
- Department of Molecular Biology, United States; Princeton Neuroscience Institute, United States; Princeton University, Princeton, NJ 08544, United States
| | - Esteban A Engel
- Princeton Neuroscience Institute, United States; Princeton University, Princeton, NJ 08544, United States
| | - Bernard G Schreurs
- Department of Neuroscience, Rockefeller Neuroscience Institute, United States; West Virginia University, Morgantown, WV 26505, United States.
| |
Collapse
|
17
|
Tsujimura T, Nakajima Y, Chotirungsan T, Kawada S, Tsutsui Y, Yoshihara M, Suzuki T, Nagoya K, Magara J, Inoue M. Inhibition of Water-Evoked Swallowing During Noxious Mechanical Stimulation of Tongue in Anesthetized Rats. Dysphagia 2022; 38:965-972. [PMID: 36127446 DOI: 10.1007/s00455-022-10522-5] [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: 01/11/2022] [Accepted: 09/11/2022] [Indexed: 11/30/2022]
Abstract
Dysphagia is sometimes accompanied by pain. Because orofacial structures subserve mastication and swallowing, orofacial pain might impair both functions. Tongue biting can occur not only accidentally while eating but also in some pathological conditions. However, it remains unclear whether noxious mechanical stimulation of the tongue affects swallowing. To explore this question, we evaluated the effects of lingual pinch stimulation on the initiation of swallowing evoked by distilled water (DW) infusion with a flow rate of 5.0 µL/s for 20 s into the pharyngolaryngeal region in anesthetized rats. The swallowing reflex was identified by electromyographic (EMG) bursts in the suprahyoid muscles which include the anterior belly of the digastric muscle, mylohyoid and geniohyoid muscles, and laryngeal elevation by visual inspection. The number of DW-evoked swallows during pinch stimulation was significantly smaller than that in a control condition or during pressure stimulation. The onset latency of the first swallow during pinch stimulation was significantly longer than that in the control condition. DW-evoked swallowing was almost abolished following bilateral transection of the superior laryngeal nerve (SLN) compared with the control condition, suggesting that the SLN plays a crucial role in the initiation of DW-evoked swallowing. Finally, electrophysiological data indicated that some SLN-responsive neurons in the nucleus tractus solitarii (nTS) exhibited delayed latency from a single SLN stimulation during lingual pinch stimulation. These results suggest that noxious mechanical stimulation of the tongue inhibits the initiation of swallowing and modulates neuronal activity in the nTS.
Collapse
Affiliation(s)
- Takanori Tsujimura
- Division of Dysphagia Rehabilitation, Niigata University Graduate School of Medical and Dental Sciences, Chuo-ku, Niigata, 951-8514, Japan.
| | - Yuta Nakajima
- Division of Dysphagia Rehabilitation, Niigata University Graduate School of Medical and Dental Sciences, Chuo-ku, Niigata, 951-8514, Japan
| | - Titi Chotirungsan
- Division of Dysphagia Rehabilitation, Niigata University Graduate School of Medical and Dental Sciences, Chuo-ku, Niigata, 951-8514, Japan
| | - Satomi Kawada
- Division of Dysphagia Rehabilitation, Niigata University Graduate School of Medical and Dental Sciences, Chuo-ku, Niigata, 951-8514, Japan
| | - Yuhei Tsutsui
- Division of Dysphagia Rehabilitation, Niigata University Graduate School of Medical and Dental Sciences, Chuo-ku, Niigata, 951-8514, Japan
| | - Midori Yoshihara
- Division of Dysphagia Rehabilitation, Niigata University Graduate School of Medical and Dental Sciences, Chuo-ku, Niigata, 951-8514, Japan
| | - Taku Suzuki
- Division of Dysphagia Rehabilitation, Niigata University Graduate School of Medical and Dental Sciences, Chuo-ku, Niigata, 951-8514, Japan
| | - Kouta Nagoya
- Division of Dysphagia Rehabilitation, Niigata University Graduate School of Medical and Dental Sciences, Chuo-ku, Niigata, 951-8514, Japan
| | - Jin Magara
- Division of Dysphagia Rehabilitation, Niigata University Graduate School of Medical and Dental Sciences, Chuo-ku, Niigata, 951-8514, Japan
| | - Makoto Inoue
- Division of Dysphagia Rehabilitation, Niigata University Graduate School of Medical and Dental Sciences, Chuo-ku, Niigata, 951-8514, Japan
| |
Collapse
|
18
|
Distinct neural networks derived from galanin-containing nociceptors and neurotensin-expressing pruriceptors. Proc Natl Acad Sci U S A 2022; 119:e2118501119. [PMID: 35943985 PMCID: PMC9388111 DOI: 10.1073/pnas.2118501119] [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] [Indexed: 11/18/2022] Open
Abstract
Pain and itch are distinct sensations arousing evasion and compulsive desire for scratching, respectively. It's unclear whether they could invoke different neural networks in the brain. Here, we use the type 1 herpes simplex virus H129 strain to trace the neural networks derived from two types of dorsal root ganglia (DRG) neurons: one kind of polymodal nociceptors containing galanin (Gal) and one type of pruriceptors expressing neurotensin (Nts). The DRG microinjection and immunosuppression were performed in transgenic mice to achieve a successful tracing from specific types of DRG neurons to the primary sensory cortex. About one-third of nuclei in the brain were labeled. More than half of them were differentially labeled in two networks. For the ascending pathways, the spinothalamic tract was absent in the network derived from Nts-expressing pruriceptors, and the two networks shared the spinobulbar projections but occupied different subnuclei. As to the motor systems, more neurons in the primary motor cortex and red nucleus of the somatic motor system participated in the Gal-containing nociceptor-derived network, while more neurons in the nucleus of the solitary tract (NST) and the dorsal motor nucleus of vagus nerve (DMX) of the emotional motor system was found in the Nts-expressing pruriceptor-derived network. Functional validation of differentially labeled nuclei by c-Fos test and chemogenetic inhibition suggested the red nucleus in facilitating the response to noxious heat and the NST/DMX in regulating the histamine-induced scratching. Thus, we reveal the organization of neural networks in a DRG neuron type-dependent manner for processing pain and itch.
Collapse
|
19
|
Chung KF, McGarvey L, Song WJ, Chang AB, Lai K, Canning BJ, Birring SS, Smith JA, Mazzone SB. Cough hypersensitivity and chronic cough. Nat Rev Dis Primers 2022; 8:45. [PMID: 35773287 PMCID: PMC9244241 DOI: 10.1038/s41572-022-00370-w] [Citation(s) in RCA: 129] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/19/2022] [Indexed: 12/13/2022]
Abstract
Chronic cough is globally prevalent across all age groups. This disorder is challenging to treat because many pulmonary and extrapulmonary conditions can present with chronic cough, and cough can also be present without any identifiable underlying cause or be refractory to therapies that improve associated conditions. Most patients with chronic cough have cough hypersensitivity, which is characterized by increased neural responsivity to a range of stimuli that affect the airways and lungs, and other tissues innervated by common nerve supplies. Cough hypersensitivity presents as excessive coughing often in response to relatively innocuous stimuli, causing significant psychophysical morbidity and affecting patients' quality of life. Understanding of the mechanisms that contribute to cough hypersensitivity and excessive coughing in different patient populations and across the lifespan is advancing and has contributed to the development of new therapies for chronic cough in adults. Owing to differences in the pathology, the organs involved and individual patient factors, treatment of chronic cough is progressing towards a personalized approach, and, in the future, novel ways to endotype patients with cough may prove valuable in management.
Collapse
Affiliation(s)
- Kian Fan Chung
- Experimental Studies Unit, National Heart & Lung Institute, Imperial College London, London, UK
- Department of Respiratory Medicine, Royal Brompton and Harefield Hospital, London, UK
| | - Lorcan McGarvey
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, UK
| | - Woo-Jung Song
- Department of Allergy and Clinical Immunology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Anne B Chang
- Australian Centre for Health Services Innovation, Queensland's University of Technology and Department of Respiratory and Sleep Medicine, Queensland Children's Hospital, Brisbane, Queensland, Australia
- Division of Child Health, Menzies School of Health Research, Darwin, Northern Territory, Australia
| | - Kefang Lai
- The First Affiliated Hospital of Guangzhou Medical University, National Center of Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou, China
| | | | - Surinder S Birring
- Centre for Human & Applied Physiological Sciences, School of Basic & Medical Biosciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - Jaclyn A Smith
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, University of Manchester, Manchester, UK
| | - Stuart B Mazzone
- Department of Anatomy and Physiology, University of Melbourne, Victoria, Australia.
| |
Collapse
|
20
|
Mafa-dependent GABAergic activity promotes mouse neonatal apneas. Nat Commun 2022; 13:3284. [PMID: 35672398 PMCID: PMC9174494 DOI: 10.1038/s41467-022-30825-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 05/19/2022] [Indexed: 01/17/2023] Open
Abstract
While apneas are associated with multiple pathological and fatal conditions, the underlying molecular mechanisms remain elusive. We report that a mutated form of the transcription factor Mafa (Mafa4A) that prevents phosphorylation of the Mafa protein leads to an abnormally high incidence of breath holding apneas and death in newborn Mafa4A/4A mutant mice. This apneic breathing is phenocopied by restricting the mutation to central GABAergic inhibitory neurons and by activation of inhibitory Mafa neurons while reversed by inhibiting GABAergic transmission centrally. We find that Mafa activates the Gad2 promoter in vitro and that this activation is enhanced by the mutation that likely results in increased inhibitory drives onto target neurons. We also find that Mafa inhibitory neurons are absent from respiratory, sensory (primary and secondary) and pontine structures but are present in the vicinity of the hypoglossal motor nucleus including premotor neurons that innervate the geniohyoid muscle, to control upper airway patency. Altogether, our data reveal a role for Mafa phosphorylation in regulation of GABAergic drives and suggest a mechanism whereby reduced premotor drives to upper airway muscles may cause apneic breathing at birth. Apneas are associated with many pathological conditions. Here, the authors show in a mouse model that stabilization of the transcription factor Mafa in brainstem GABAergic neurons may contribute to apnea, by decreasing motor drive to muscles controlling the airways.
Collapse
|
21
|
Kurganov E, Okamoto K, Miyata S. Distribution of TRPM8-expressing trigeminal nerve fibers in the pons and medulla oblongata of the mouse brain. J Chem Neuroanat 2022; 122:102104. [PMID: 35561876 DOI: 10.1016/j.jchemneu.2022.102104] [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: 11/30/2021] [Revised: 05/05/2022] [Accepted: 05/06/2022] [Indexed: 11/16/2022]
Abstract
Transient receptor potential melastatin 8 (TRPM8), a cold-mediated ion channel, is well known to be expressed in primary sensory neurons; however, limited information is currently available on the distribution of TRPM8-expressing trigeminal nerve fibers in the brainstem. The present study showed the distribution of TRPM8-expressing fibers in the pons and medulla oblongata of the TRPM8 KO mice engineered by knocking in EGFP at the frame of the start codon of TRPM8. In addition, TRPM8-expressing fibers were also observed in the brachium pontis, middle cerebellar peduncle, the sensory root of the trigeminal nerve, and spinal trigeminal tract (sp5). Furthermore, TRPM8-expressing nerve fibers surrounded the somata of HuC/D-positive neurons in the sp5. Moreover, the distribution of TRPM8-expressing fibers from rostral to caudal was visualized in sagittal sections of the mouse brain. The present results also revealed that a high number of TRPM8-expressing fibers colocalized with CTB-labeled fibers in the sp5 following an injection of CTB into the whisker compared to mice's eye and ear. These results show the distribution pathway of TRPM8-expressing fibers in the pons and medulla oblongata and possible involvement in peripheral signaling from the trigeminal nerve.
Collapse
Affiliation(s)
- Erkin Kurganov
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan.
| | - Kaho Okamoto
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
| | - Seiji Miyata
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
| |
Collapse
|
22
|
Kim SH, Patil MJ, Hadley SH, Bahia PK, Butler SG, Madaram M, Taylor-Clark TE. Mapping of the Sensory Innervation of the Mouse Lung by Specific Vagal and Dorsal Root Ganglion Neuronal Subsets. eNeuro 2022; 9:ENEURO.0026-22.2022. [PMID: 35365503 PMCID: PMC9015009 DOI: 10.1523/eneuro.0026-22.2022] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 03/10/2022] [Accepted: 03/26/2022] [Indexed: 11/21/2022] Open
Abstract
The airways are densely innervated by sensory afferent nerves, whose activation regulates respiration and triggers defensive reflexes (e.g., cough, bronchospasm). Airway innervation is heterogeneous, and distinct afferent subsets have distinct functional responses. However, little is known of the innervation patterns of subsets within the lung. A neuroanatomical map is critical for understanding afferent activation under physiological and pathophysiological conditions. Here, we quantified the innervation of the mouse lung by vagal and dorsal root ganglion (DRG) sensory subsets defined by the expression of Pirt (all afferents), 5HT3 (vagal nodose afferents), Tac1 (tachykinergic afferents), and transient receptor potential vanilloid 1 channel (TRPV1; defensive/nociceptive afferents) using Cre-mediated reporter expression. We found that vagal afferents innervate almost all conducting airways and project into the alveolar region, whereas DRG afferents only innervate large airways. Of the two vagal ganglia, only nodose afferents project into the alveolar region, but both nodose and jugular afferents innervate conducting airways throughout the lung. Many afferents that project into the alveolar region express TRPV1. Few DRG afferents expressed TRPV1. Approximately 25% of blood vessels were innervated by vagal afferents (many were Tac1+). Approximately 10% of blood vessels had DRG afferents (some were Tac1+), but this was restricted to large vessels. Lastly, innervation of neuroepithelial bodies (NEBs) correlated with the cell number within the bodies. In conclusion, functionally distinct sensory subsets have distinct innervation patterns within the conducting airways, alveoli and blood vessels. Physiologic (e.g., stretch) and pathophysiological (e.g., inflammation, edema) stimuli likely vary throughout these regions. Our data provide a neuroanatomical basis for understanding afferent responses in vivo.
Collapse
Affiliation(s)
- Seol-Hee Kim
- Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612
| | - Mayur J Patil
- Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612
| | - Stephen H Hadley
- Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612
| | - Parmvir K Bahia
- Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612
| | - Shane G Butler
- Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612
| | - Meghana Madaram
- Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612
| | - Thomas E Taylor-Clark
- Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612
| |
Collapse
|
23
|
Prescott SL, Liberles SD. Internal senses of the vagus nerve. Neuron 2022; 110:579-599. [PMID: 35051375 PMCID: PMC8857038 DOI: 10.1016/j.neuron.2021.12.020] [Citation(s) in RCA: 117] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 11/30/2021] [Accepted: 12/11/2021] [Indexed: 12/16/2022]
Abstract
The vagus nerve is an indispensable body-brain connection that controls vital aspects of autonomic physiology like breathing, heart rate, blood pressure, and gut motility, reflexes like coughing and swallowing, and survival behaviors like feeding, drinking, and sickness responses. Classical physiological studies and recent molecular/genetic approaches have revealed a tremendous diversity of vagal sensory neuron types that innervate different internal organs, with many cell types remaining poorly understood. Here, we review the state of knowledge related to vagal sensory neurons that innervate the respiratory, cardiovascular, and digestive systems. We focus on cell types and their response properties, physiological/behavioral roles, engaged neural circuits and, when possible, sensory receptors. We are only beginning to understand the signal transduction mechanisms used by vagal sensory neurons and upstream sentinel cells, and future studies are needed to advance the field of interoception to the level of mechanistic understanding previously achieved for our external senses.
Collapse
|
24
|
Chen Z, Lin MT, Zhan C, Zhong NS, Mu D, Lai KF, Liu MJ. A descending pathway emanating from the periaqueductal gray mediates the development of cough-like hypersensitivity. iScience 2022; 25:103641. [PMID: 35028531 PMCID: PMC8741493 DOI: 10.1016/j.isci.2021.103641] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/11/2021] [Accepted: 12/13/2021] [Indexed: 01/10/2023] Open
Abstract
Chronic cough is a common refractory symptom of various respiratory diseases. However, the neural mechanisms that modulate the cough sensitivity and mediate chronic cough remain elusive. Here, we report that GABAergic neurons in the lateral/ventrolateral periaqueductal gray (l/vlPAG) suppress cough processing via a descending pathway. We found that l/vlPAG neurons are activated by coughing-like behaviors and that tussive agent-evoked coughing-like behaviors are impaired after activation of l/vlPAG neurons. In addition, we showed that l/vlPAG neurons form inhibitory synapses with the nucleus of the solitary tract (NTS) neurons. The synaptic strength of these inhibitory projections is weaker in cough hypersensitivity model mice than in naïve mice. Important, activation of l/vlPAG GABAergic neurons projecting to the NTS decreases coughing-like behaviors. In contrast, suppressing these neurons enhances cough sensitivity. These results support the notion that l/vlPAG GABAergic neurons play important roles in cough hypersensitivity and chronic cough through disinhibition of cough processing at the medullary level. GABAergic neurons in the l/vlPAG inhibit coughing-like behaviors The l/vlPAG sends predominately inhibitory projections to the NTS l/vlPAG GABAergic neurons modulate coughing-like behaviors via descending projections l/vlPAG-NTS projections mediate cough hypersensitivity via disinhibitory mechanisms
Collapse
Affiliation(s)
- Zhe Chen
- State Key Laboratory of Respiratory Disease, Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yan Jiang Xi Road, Guangzhou 510120, China.,Laboratory of Cough, Affiliated Kunshan Hospital of Jiangsu University, Suzhou, Jiangsu 215300, China
| | - Ming-Tong Lin
- State Key Laboratory of Respiratory Disease, Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yan Jiang Xi Road, Guangzhou 510120, China
| | - Chen Zhan
- State Key Laboratory of Respiratory Disease, Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yan Jiang Xi Road, Guangzhou 510120, China
| | - Nan-Shan Zhong
- State Key Laboratory of Respiratory Disease, Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yan Jiang Xi Road, Guangzhou 510120, China
| | - Di Mu
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 650 Xin Song Jiang Road, Shanghai 201620, China
| | - Ke-Fang Lai
- State Key Laboratory of Respiratory Disease, Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yan Jiang Xi Road, Guangzhou 510120, China
| | - Mingzhe J Liu
- State Key Laboratory of Respiratory Disease, Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yan Jiang Xi Road, Guangzhou 510120, China
| |
Collapse
|
25
|
Su Y, Barr J, Jaquish A, Xu J, Verheyden JM, Sun X. Identification of lung innervating sensory neurons and their target specificity. Am J Physiol Lung Cell Mol Physiol 2022; 322:L50-L63. [PMID: 34755535 PMCID: PMC8721910 DOI: 10.1152/ajplung.00376.2021] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Known as the gas exchange organ, the lung is also critical for responding to the aerosol environment in part through interaction with the nervous system. The diversity and specificity of lung innervating neurons remain poorly understood. Here, we interrogated the cell body location and molecular signature and projection pattern of lung innervating sensory neurons. Retrograde tracing from the lung coupled with whole tissue clearing highlighted neurons primarily in the vagal ganglia. Centrally, they project specifically to the nucleus of the solitary tract in the brainstem. Peripherally, they enter the lung alongside branching airways. Labeling of nociceptor Trpv1+ versus peptidergic Tac1+ vagal neurons showed shared and distinct terminal morphology and targeting to airway smooth muscles, vasculature including lymphatics, and alveoli. Notably, a small population of vagal neurons that are Calb1+ preferentially innervate pulmonary neuroendocrine cells, a demonstrated airway sensor population. This atlas of lung innervating neurons serves as a foundation for understanding their function in lung.
Collapse
Affiliation(s)
- Yujuan Su
- 1Department of Pediatrics, University of California, San Diego, California
| | - Justinn Barr
- 1Department of Pediatrics, University of California, San Diego, California
| | - Abigail Jaquish
- 1Department of Pediatrics, University of California, San Diego, California
| | - Jinhao Xu
- 1Department of Pediatrics, University of California, San Diego, California
| | - Jamie M. Verheyden
- 1Department of Pediatrics, University of California, San Diego, California
| | - Xin Sun
- 1Department of Pediatrics, University of California, San Diego, California,2Division of Biological Sciences, University of California, San Diego, California
| |
Collapse
|
26
|
Taylor-Clark TE, Undem BJ. Neural control of the lower airways: Role in cough and airway inflammatory disease. HANDBOOK OF CLINICAL NEUROLOGY 2022; 188:373-391. [PMID: 35965034 PMCID: PMC10688079 DOI: 10.1016/b978-0-323-91534-2.00013-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Airway function is under constant neurophysiological control, in order to maximize airflow and gas exchange and to protect the airways from aspiration, damage, and infection. There are multiple sensory nerve subtypes, whose disparate functions provide a wide array of sensory information into the CNS. Activation of these subtypes triggers specific reflexes, including cough and alterations in autonomic efferent control of airway smooth muscle, secretory cells, and vasculature. Importantly, every aspect of these reflex arcs can be impacted and altered by local inflammation caused by chronic lung disease such as asthma, bronchitis, and infections. Excessive and inappropriate activity in sensory and autonomic nerves within the airways is thought to contribute to the morbidity and symptoms associated with lung disease.
Collapse
Affiliation(s)
- Thomas E Taylor-Clark
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Bradley J Undem
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
27
|
Rouadi PW, Idriss SA, Bousquet J, Laidlaw TM, Azar CR, Sulaiman AL-Ahmad M, Yáñez A, AL-Nesf MAY, Nsouli TM, Bahna SL, Abou-Jaoude E, Zaitoun FH, Hadi UM, Hellings PW, Scadding GK, Smith PK, Morais-Almeida M, Gómez RM, González Díaz SN, Klimek L, Juvelekian GS, Riachy MA, Canonica GW, Peden D, Wong GW, Sublett J, Bernstein JA, Wang L, Tanno LK, Chikhladze M, Levin M, Chang YS, Martin BL, Caraballo L, Custovic A, Ortega-Martell JA, Jensen-Jarolim E, Ebisawa M, Fiocchi A, Ansotegui IJ. WAO-ARIA consensus on chronic cough - Part 1: Role of TRP channels in neurogenic inflammation of cough neuronal pathways. World Allergy Organ J 2021; 14:100617. [PMID: 34934475 PMCID: PMC8654622 DOI: 10.1016/j.waojou.2021.100617] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 09/09/2021] [Accepted: 09/14/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Cough features a complex peripheral and central neuronal network. The function of the chemosensitive and stretch (afferent) cough receptors is well described but partly understood. It is speculated that chronic cough reflects a neurogenic inflammation of the cough reflex, which becomes hypersensitive. This is mediated by neuromediators, cytokines, inflammatory cells, and a differential expression of neuronal (chemo/stretch) receptors, such as transient receptor potential (TRP) and purinergic P2X ion channels; yet the overall interaction of these mediators in neurogenic inflammation of cough pathways remains unclear. OBJECTIVES The World Allergy Organization/Allergic Rhinitis and its Impact on Asthma (WAO/ARIA) Joint Committee on Chronic Cough reviewed the current literature on neuroanatomy and pathophysiology of chronic cough. The role of TRP ion channels in pathogenic mechanisms of the hypersensitive cough reflex was also examined. OUTCOMES Chemoreceptors are better studied in cough neuronal pathways compared to stretch receptors, likely due to their anatomical overabundance in the respiratory tract, but also their distinctive functional properties. Central pathways are important in suppressive mechanisms and behavioral/affective aspects of chronic cough. Current evidence strongly suggests neurogenic inflammation induces a hypersensitive cough reflex marked by increased expression of neuromediators, mast cells, and eosinophils, among others. TRP ion channels, mainly TRP V1/A1, are important in the pathogenesis of chronic cough due to their role in mediating chemosensitivity to various endogenous and exogenous triggers, as well as a crosstalk between neurogenic and inflammatory pathways in cough-associated airways diseases.
Collapse
Affiliation(s)
- Philip W. Rouadi
- Department of Otolaryngology - Head and Neck Surgery, Eye and Ear University Hospital, Beirut, Lebanon
| | - Samar A. Idriss
- Department of Otolaryngology - Head and Neck Surgery, Eye and Ear University Hospital, Beirut, Lebanon
- Department of Audiology and Otoneurological Evaluation, Edouard Herriot Hospital, Lyon, France
| | - Jean Bousquet
- Hospital Charité, Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Dermatology and Allergy, Comprehensive Allergy Center, Berlin Institute of Health, Berlin, Germany
- Macvia France, Montpellier France
- Université Montpellier, Montpellier, France
| | - Tanya M. Laidlaw
- Department of Medicine, Harvard Medical School, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital Boston, MA, USA
| | - Cecilio R. Azar
- Department of Gastroenterology, American University of Beirut Medical Center (AUBMC), Beirut, Lebanon
- Department of Gastroenterology, Middle East Institute of Health (MEIH), Beirut, Lebanon
- Department of Gastroenterology, Clemenceau Medical Center (CMC), Beirut, Lebanon
| | | | - Anahí Yáñez
- INAER - Investigaciones en Alergia y Enfermedades Respiratorias, Buenos Aires, Argentina
| | - Maryam Ali Y. AL-Nesf
- Allergy and Immunology Section, Department of Medicine, Hamad Medical Corporation, P.O. Box 3050, Doha, Qatar
| | | | - Sami L. Bahna
- Allergy & Immunology Section, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | | | - Fares H. Zaitoun
- Department of Allergy Otolaryngology, LAU-RIZK Medical Center, Beirut, Lebanon
| | - Usamah M. Hadi
- Clinical Professor Department of Otolaryngology Head and Neck Surgery, American University of Beirut, Lebanon
| | - Peter W. Hellings
- KU Leuven Department of Microbiology, Immunology and Transplantation, Laboratory of Allergy and Clinical Immunology, Leuven, Belgium
- University Hospitals Leuven, Department of Otorhinolaryngology, Leuven, Belgium
- University Hospital Ghent, Department of Otorhinolaryngology, Laboratory of Upper Airways Research, Ghent, Belgium
- Academic Medical Center, University of Amsterdam, Department of Otorhinolaryngology, Amsterdam, the Netherlands
| | | | - Peter K. Smith
- Clinical Medicine Griffith University, Southport Qld, 4215, Australia
| | | | | | - Sandra N. González Díaz
- Universidad Autónoma de Nuevo León, Hospital Universitario and Facultad de Medicina, Monterrey, Nuevo León, Mexico
| | - Ludger Klimek
- Center for Rhinology and Allergology, Wiesbaden, Germany
| | - Georges S. Juvelekian
- Department of Pulmonary, Critical Care and Sleep Medicine at Saint George Hospital University Medical Center, Beirut, Lebanon
| | - Moussa A. Riachy
- Department of Pulmonary and Critical Care, Hôtel-Dieu de France University Hospital, Beirut, Lebanon
| | - Giorgio Walter Canonica
- Humanitas University, Personalized Medicine Asthma & Allergy Clinic-Humanitas Research Hospital-IRCCS-Milano Italy
| | - David Peden
- UNC Center for Environmental Medicine, Asthma, and Lung Biology, Division of Allergy, Immunology and Rheumatology, Department of Pediatrics UNS School of Medicine, USA
| | - Gary W.K. Wong
- Department of Pediatrics, Chinese University of Hong Kong, Hong Kong, China
| | - James Sublett
- Department of Pediatrics, Section of Allergy and Immunology, University of Louisville School of Medicine, 9800 Shelbyville Rd, Louisville, KY, USA
| | - Jonathan A. Bernstein
- University of Cincinnati College of Medicine, Department of Internal Medicine, Division of Immunology/Allergy Section, Cincinnati
| | - Lianglu Wang
- Department of Allergy, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Precision Medicine for Diagnosis and Treatment of Allergic Disease, State Key Laboratory of Complex Severe and Rare Diseases, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Beijing 100730, China
| | - Luciana Kase Tanno
- Université Montpellier, Montpellier, France
- Desbrest Institute of Epidemiology and Public Health, UMR UA-11, INSERM University of Montpellier, Montpellier, France
- WHO Collaborating Centre on Scientific Classification Support, Montpellier, France
| | - Manana Chikhladze
- Medical Faculty at Akaki Tsereteli State University, National Institute of Allergy, Asthma & Clinical Immunology, KuTaisi, Tskaltubo, Georgia
| | - Michael Levin
- Division of Paediatric Allergology, Department of Paediatrics, University of Cape Town, South Africa
| | - Yoon-Seok Chang
- Division of Allergy and Clinical Immunology, Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, South Korea
| | - Bryan L. Martin
- Department of Otolaryngology, Division of Allergy & Immunology, The Ohio State University, Columbus, OH, USA
| | - Luis Caraballo
- Institute for Immunological Research, University of Cartagena. Cartagena de Indias, Colombia
| | - Adnan Custovic
- National Heart and Lund Institute, Imperial College London, UK
| | | | - Erika Jensen-Jarolim
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University Vienna, Austria
- The Interuniversity Messerli Research Institute, Medical University Vienna and Univ, of Veterinary Medicine Vienna, Austria
| | - Motohiro Ebisawa
- Clinical Research Center for Allergy and Rheumatology,National Hospital Organization Sagamihara National Hospital, Sagamihara, Japan
| | - Alessandro Fiocchi
- Translational Pediatric Research Area, Allergic Diseases Research Unit, Bambino Gesù Children's Hospital IRCCS, Rome, Holy See
| | - Ignacio J. Ansotegui
- Department of Allergy and Immunology, Hospital Quironsalud Bizkaia, Bilbao, Spain
| |
Collapse
|
28
|
Moe AAK, McGovern AE, Mazzone SB. Jugular vagal ganglia neurons and airway nociception: A target for treating chronic cough. Int J Biochem Cell Biol 2021; 135:105981. [PMID: 33895353 DOI: 10.1016/j.biocel.2021.105981] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/28/2021] [Accepted: 04/16/2021] [Indexed: 12/12/2022]
Abstract
The airways receive a dense supply of sensory nerve fibers that are responsive to damaging or potentially injurious stimuli. These airway nociceptors are mainly derived from the jugular and nodose vagal ganglia, and when activated they induce a range of reflexes and sensations that play an essential role in airway protection. Jugular nociceptors differ from nodose nociceptors in their embryonic origins, molecular profile and termination patterns in the airways and the brain, and recent discoveries suggest that excessive activity in jugular nociceptors may be central to the development of chronic cough. For these reasons, targeting jugular airway nociceptor signaling processes at different levels of the neuraxis may be a promising target for therapeutic development. In this focused review, we present the current understanding of jugular ganglia nociceptors, how they may contribute to chronic cough and mechanisms that could be targeted to bring about cough suppression.
Collapse
Affiliation(s)
- Aung Aung Kywe Moe
- Department of Anatomy and Physiology, School of Biomedical Sciences, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Alice E McGovern
- Department of Anatomy and Physiology, School of Biomedical Sciences, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Stuart B Mazzone
- Department of Anatomy and Physiology, School of Biomedical Sciences, The University of Melbourne, Parkville, Victoria, 3010, Australia.
| |
Collapse
|
29
|
Abstract
The jugular-nodose ganglia contain the sensory peripheral neurons of the vagus nerve, linking visceral organs to the medulla oblongata. Accessing these ganglia in smaller animals without damaging the vascular and neural structures may be challenging, as ganglionic fibers imbed deeply into the carotid sheath, and vagal parasympathetic fibers cross through the interior of the ganglia. We describe a practical protocol for locating and accessing the mouse jugular-nodose ganglia in vivo, including instructions for intraganglionic injections and postperfusion dissection. For complete details on the use and execution of this protocol, please refer to Han et al. (2018). Practical approach to locate the mouse jugular-nodose ganglia Detailed instructions on how to perform intraganglionic injections Detailed description of ganglia-preserving postperfusion dissection
Collapse
|
30
|
Olsen WL, Rose M, Golder FJ, Wang C, Hammond JC, Bolser DC. Intra-Arterial, but Not Intrathecal, Baclofen and Codeine Attenuates Cough in the Cat. Front Physiol 2021; 12:640682. [PMID: 33746778 PMCID: PMC7973226 DOI: 10.3389/fphys.2021.640682] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 02/15/2021] [Indexed: 11/15/2022] Open
Abstract
Centrally-acting antitussive drugs are thought to act solely in the brainstem. However, the role of the spinal cord in the mechanism of action of these drugs is unknown. The purpose of this study was to determine if antitussive drugs act in the spinal cord to reduce the magnitude of tracheobronchial (TB) cough-related expiratory activity. Experiments were conducted in anesthetized, spontaneously breathing cats (n = 22). Electromyograms (EMG) were recorded from the parasternal (PS) and transversus abdominis (TA) or rectus abdominis muscles. Mechanical stimulation of the trachea or larynx was used to elicit TB cough. Baclofen (10 and 100 μg/kg, GABA-B receptor agonist) or codeine (30 μg/kg, opioid receptor agonist) was administered into the intrathecal (i.t.) space and also into brainstem circulation via the vertebral artery. Cumulative doses of i.t. baclofen or codeine had no effect on PS, abdominal muscle EMGs or cough number during the TB cough. Subsequent intra-arterial (i.a.) administration of baclofen or codeine significantly reduced magnitude of abdominal and PS muscles during TB cough. Furthermore, TB cough number was significantly suppressed by i.a. baclofen. The influence of these drugs on other behaviors that activate abdominal motor pathways was also assessed. The abdominal EMG response to noxious pinch of the tail was suppressed by i.t. baclofen, suggesting that the doses of baclofen that were employed were sufficient to affect spinal pathways. However, the abdominal EMG response to expiratory threshold loading was unaffected by i.t. administration of either baclofen or codeine. These results indicate that neither baclofen nor codeine suppress cough via a spinal action and support the concept that the antitussive effect of these drugs is restricted to the brainstem.
Collapse
Affiliation(s)
- Wendy L. Olsen
- Department of Physiological Sciences, University of Florida, Gainesville, FL, United States
| | | | | | | | | | | |
Collapse
|
31
|
Pincus AB, Fryer AD, Jacoby DB. Mini review: Neural mechanisms underlying airway hyperresponsiveness. Neurosci Lett 2021; 751:135795. [PMID: 33667601 DOI: 10.1016/j.neulet.2021.135795] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 02/22/2021] [Accepted: 02/23/2021] [Indexed: 12/25/2022]
Abstract
Neural changes underly hyperresponsiveness in asthma and other airway diseases. Afferent sensory nerves, nerves within the brainstem, and efferent parasympathetic nerves all contribute to airway hyperresponsiveness. Inflammation plays a critical role in these nerve changes. Chronic inflammation and pre-natal exposures lead to increased airway innervation and structural changes. Acute inflammation leads to shifts in neurotransmitter expression of afferent nerves and dysfunction of M2 muscarinic receptors on efferent nerve endings. Eosinophils and macrophages drive these changes through release of inflammatory mediators. Novel tools, including optogenetics, two photon microscopy, and optical clearing and whole mount microscopy, allow for improved studies of the structure and function of airway nerves and airway hyperresponsiveness.
Collapse
Affiliation(s)
- Alexandra B Pincus
- Oregon Health and Science University, 3181 SW Sam Jackson Park Road, BRB 440, Portland, OR, 97239, USA.
| | - Allison D Fryer
- Oregon Health and Science University, 3181 SW Sam Jackson Park Road, BRB 440, Portland, OR, 97239, USA
| | - David B Jacoby
- Oregon Health and Science University, 3181 SW Sam Jackson Park Road, BRB 440, Portland, OR, 97239, USA
| |
Collapse
|
32
|
Behrens R, McGovern AE, Farrell MJ, Moe AAK, Mazzone SB. Mini Review: Central Organization of Airway Afferent Nerve Circuits. Neurosci Lett 2020; 744:135604. [PMID: 33387662 DOI: 10.1016/j.neulet.2020.135604] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 12/16/2020] [Accepted: 12/21/2020] [Indexed: 10/22/2022]
Abstract
Airway afferents monitor the local chemical and physical micro-environments in the airway wall and lungs and send this information centrally to regulate neural circuits involved in setting autonomic tone, evoking reflex and volitional respiratory motor outflows, encoding perceivable sensations and contributing to higher order cognitive processing. In this mini-review we present a current overview of the central wiring of airway afferent circuits in the brainstem and brain, highlighting recent discoveries that augment our understanding of airway sensory processing. We additionally explore how advances in describing the molecular diversity of airway afferents may influence future research efforts aimed at defining central mesoscale connectivity of airway afferent pathways. A refined understanding of how functionally distinct airway afferent pathways are organized in the brain will provide deeper insight into the physiology of airway afferent-evoked responses and may foster opportunities for targeted modulation of specific pathways involved in disease.
Collapse
Affiliation(s)
- Robert Behrens
- Department of Anatomy and Neuroscience, School of Biomedical Science, The University of Melbourne, Parkville, Australia
| | - Alice E McGovern
- Department of Anatomy and Neuroscience, School of Biomedical Science, The University of Melbourne, Parkville, Australia
| | - Michael J Farrell
- Department of Medical Imaging and Radiation Sciences, Monash University, Clayton, Australia; Monash Biomedical Imaging, Monash University, Clayton, Australia
| | - Aung Aung Kywe Moe
- Department of Anatomy and Neuroscience, School of Biomedical Science, The University of Melbourne, Parkville, Australia
| | - Stuart B Mazzone
- Department of Anatomy and Neuroscience, School of Biomedical Science, The University of Melbourne, Parkville, Australia.
| |
Collapse
|
33
|
Taylor-Clark TE. Molecular identity, anatomy, gene expression and function of neural crest vs. placode-derived nociceptors in the lower airways. Neurosci Lett 2020; 742:135505. [PMID: 33197519 DOI: 10.1016/j.neulet.2020.135505] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 11/02/2020] [Accepted: 11/09/2020] [Indexed: 12/11/2022]
Abstract
The lower airways (larynx to alveoli) are protected by a complex array of neural networks that regulate respiration and airway function. Harmful stimuli trigger defensive responses such as apnea, cough and bronchospasm by activating a subpopulation of sensory afferent nerves (termed nociceptors) which are found throughout the airways. Airway nociceptive fibers are projected from the nodose vagal ganglia, the jugular vagal ganglia and the dorsal root ganglia, which are derived from distinct embryological sources: the former from the epibranchial placodes, the latter two from the neural crest. Embryological source determines nociceptive gene expression of receptors and neurotransmitters and recent evidence suggests that placode- and neural crest-derived nociceptors have distinct stimuli sensitivity, innervation patterns and functions. Improved understanding of the function of each subset in specific reflexes has substantial implications for therapeutic targeting of the neuronal components of airway disease such as asthma, viral infections and chronic obstructive pulmonary disease.
Collapse
Affiliation(s)
- Thomas E Taylor-Clark
- Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd., Tampa, FL 33612, USA.
| |
Collapse
|
34
|
Abstract
Air pollutants pose a serious worldwide health hazard, causing respiratory and cardiovascular morbidity and mortality. Pollutants perturb the autonomic nervous system, whose function is critical to cardiopulmonary homeostasis. Recent studies suggest that pollutants can stimulate defensive sensory nerves within the cardiopulmonary system, thus providing a possible mechanism for pollutant-induced autonomic dysfunction. A better understanding of the mechanisms involved would likely improve the management and treatment of pollution-related disease.
Collapse
Affiliation(s)
- Thomas E Taylor-Clark
- Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida
| |
Collapse
|
35
|
Descending Modulation of Laryngeal Vagal Sensory Processing in the Brainstem Orchestrated by the Submedius Thalamic Nucleus. J Neurosci 2020; 40:9426-9439. [PMID: 33115928 DOI: 10.1523/jneurosci.2430-20.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/20/2020] [Accepted: 10/21/2020] [Indexed: 11/21/2022] Open
Abstract
The nodose and jugular vagal ganglia supply sensory innervation to the airways and lungs. Jugular vagal airway sensory neurons wire into a brainstem circuit with ascending projections into the submedius thalamic nucleus (SubM) and ventrolateral orbital cortex (VLO), regions known to regulate the endogenous analgesia system. Here we investigate whether the SubM-VLO circuit exerts descending regulation over airway vagal reflexes in male and female rats using a range of neuroanatomical tracing, reflex physiology, and chemogenetic techniques. Anterograde and retrograde neuroanatomical tracing confirmed the connectivity of the SubM and VLO. Laryngeal stimulation in anesthetized rats reduced respiration, a reflex that was potently inhibited by activation of SubM. Conversely, inhibition of SubM potentiated laryngeal reflex responses, while prior lesions of VLO abolished the effects of SubM stimulation. In conscious rats, selective chemogenetic activation of SubM neurons specifically projecting to VLO significantly inhibited respiratory responses evoked by inhalation of the nociceptor stimulant capsaicin. Jugular vagal inputs to SubM via the medullary paratrigeminal nucleus were confirmed using anterograde transsynaptic conditional herpes viral tracing. Respiratory responses evoked by microinjections of capsaicin into the paratrigeminal nucleus were significantly attenuated by SubM stimulation, whereas those evoked via the nucleus of the solitary tract were unaltered. These data suggest that jugular vagal sensory pathways input to a nociceptive thalamocortical circuit capable of regulating jugular sensory processing in the medulla. This circuit organization suggests an intersection between vagal sensory pathways and the endogenous analgesia system, potentially important for understanding vagal sensory processing in health and mechanisms of hypersensitivity in disease.SIGNIFICANCE STATEMENT Jugular vagal sensory pathways are increasingly recognized for their important role in defensive respiratory responses evoked from the airways. Jugular ganglia neurons wire into a central circuit that is notable for overlapping with somatosensory processing networks in the brain rather than the viscerosensory circuits in receipt of inputs from the nodose vagal ganglia. Here we demonstrate a novel and functionally relevant example of intersection between vagal and somatosensory processing in the brain. The findings of the study offer new insights into interactions between vagal and spinal sensory processing, including the medullary targets of the endogenous analgesia system, and offer new insights into the central processes involved in airway defense in health and disease.
Collapse
|
36
|
Farrell MJ, Bautista TG, Liang E, Azzollini D, Egan GF, Mazzone SB. Evidence for multiple bulbar and higher brain circuits processing sensory inputs from the respiratory system in humans. J Physiol 2020; 598:5771-5787. [DOI: 10.1113/jp280220] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 09/02/2020] [Indexed: 12/13/2022] Open
Affiliation(s)
- Michael J. Farrell
- Departmental of Medical Imaging and Radiation Sciences Monash University Clayton VIC Australia
- Monash Biomedical Imaging Monash University Clayton VIC Australia
| | - Tara G. Bautista
- Department of Anatomy and Neuroscience University of Melbourne Parkville Victoria Australia
| | - Emma Liang
- Monash Biomedical Imaging Monash University Clayton VIC Australia
| | - Damian Azzollini
- Monash Biomedical Imaging Monash University Clayton VIC Australia
| | - Gary F. Egan
- Monash Biomedical Imaging Monash University Clayton VIC Australia
- School of Psychological Sciences Monash University Clayton VIC Australia
- ARC Centre of Excellence for Integrative Brain Function Monash University Clayton VIC Australia
| | - Stuart B. Mazzone
- Department of Anatomy and Neuroscience University of Melbourne Parkville Victoria Australia
| |
Collapse
|
37
|
Paranathala MP, Mitchell P. Neurogenic Cough Associated with Hyperintensity in Dorsal Medulla: Case Report and Anatomical Discussion. World Neurosurg 2020; 144:196-198. [PMID: 32977030 DOI: 10.1016/j.wneu.2020.09.096] [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: 06/13/2020] [Revised: 09/16/2020] [Accepted: 09/17/2020] [Indexed: 11/17/2022]
Abstract
BACKGROUND The nucleus tractus solitarius and paratrigeminal nucleus, which are implicated in the processing of airway-derived sensory information, are found in the dorsal medulla. The mechanism and localization of higher-order processing of urge to cough is poorly understood, and much of the existing anatomical localization is limited to animal studies. CASE DESCRIPTION A 44-year-old Caucasian lady underwent elective foramen magnum decompression for symptomatic Chiari I malformation; postoperatively she had resolution of Chiari symptoms but developed an intractable neurogenic cough. She has no significant medical history or premorbid respiratory issues. Postoperative magnetic resonance imaging of her head demonstrated signal change in the left dorsal medulla, corresponding with the nucleus tractus solitarius and paratrigeminal nucleus. CONCLUSIONS We suggest that this lesion explains her isolated new cough and localizes the pathway for "urge to cough" to this region of the medulla.
Collapse
Affiliation(s)
| | - Patrick Mitchell
- Department of Neurosurgery, Royal Victoria Hospital, Newcastle, UK
| |
Collapse
|
38
|
Singh N, Driessen AK, McGovern AE, Moe AAK, Farrell MJ, Mazzone SB. Peripheral and central mechanisms of cough hypersensitivity. J Thorac Dis 2020; 12:5179-5193. [PMID: 33145095 PMCID: PMC7578480 DOI: 10.21037/jtd-2020-icc-007] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Chronic cough is a difficult to treat symptom of many respiratory and some non-respiratory diseases, indicating that varied pathologies can underpin the development of chronic cough. However, clinically and experimentally it has been useful to collate these different pathological processes into the single unifying concept of cough hypersensitivity. Cough hypersensitivity syndrome is reflected by troublesome cough often precipitated by levels of stimuli that ordinarily don't cause cough in healthy people, and this appears to be a hallmark feature in many patients with chronic cough. Accordingly, a strong argument has emerged that changes in the excitability and/or normal regulation of the peripheral and central neural circuits responsible for cough are instrumental in establishing cough hypersensitivity and for causing excessive cough in disease. In this review, we explore the current peripheral and central neural mechanisms that are believed to be involved in altered cough sensitivity and present possible links to the mechanism of action of novel therapies that are currently undergoing clinical trials for chronic cough.
Collapse
Affiliation(s)
- Nabita Singh
- Department of Medical Imaging and Radiation Sciences, Monash University, Clayton, Australia
| | - Alexandria K. Driessen
- Department of Anatomy and Neuroscience, School of Biomedical Science, The University of Melbourne, Parkville, Australia
| | - Alice E. McGovern
- Department of Anatomy and Neuroscience, School of Biomedical Science, The University of Melbourne, Parkville, Australia
| | - Aung Aung Kywe Moe
- Department of Anatomy and Neuroscience, School of Biomedical Science, The University of Melbourne, Parkville, Australia
| | - Michael J. Farrell
- Department of Medical Imaging and Radiation Sciences, Monash University, Clayton, Australia
- Monash Biomedical Imaging, Monash University, Clayton, Australia
| | - Stuart B. Mazzone
- Department of Anatomy and Neuroscience, School of Biomedical Science, The University of Melbourne, Parkville, Australia
| |
Collapse
|
39
|
Development of a Mouse Reporter Strain for the Purinergic P2X 2 Receptor. eNeuro 2020; 7:ENEURO.0203-20.2020. [PMID: 32669344 PMCID: PMC7418537 DOI: 10.1523/eneuro.0203-20.2020] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/23/2020] [Accepted: 07/03/2020] [Indexed: 02/06/2023] Open
Abstract
The ATP-sensitive P2X2 ionotropic receptor plays a critical role in a number of signal processes including taste and hearing, carotid body detection of hypoxia, the exercise pressor reflex and sensory transduction of mechanical stimuli in the airways and bladder. Elucidation of the role of P2X2 has been hindered by the lack of selective tools. In particular, detection of P2X2 using established pharmacological and biochemical techniques yields dramatically different expression patterns, particularly in the peripheral and central nervous systems. Here, we have developed a knock-in P2X2-cre mouse, which we crossed with a cre-sensitive tdTomato reporter mouse to determine P2X2 expression. P2X2 was found in more than 80% of nodose vagal afferent neurons, but not in jugular vagal afferent neurons. Reporter expression correlated in vagal neurons with sensitivity to α,β methylene ATP (αβmATP). P2X2 was expressed in 75% of petrosal afferents, but only 12% and 4% of dorsal root ganglia (DRG) and trigeminal afferents, respectively. P2X2 expression was limited to very few cell types systemically. Together with the central terminals of P2X2-expressing afferents, reporter expression in the CNS was mainly found in brainstem neurons projecting mossy fibers to the cerebellum, with little expression in the hippocampus or cortex. The structure of peripheral terminals of P2X2-expressing afferents was demonstrated in the tongue (taste buds), carotid body, trachea and esophagus. P2X2 was observed in hair cells and support cells in the cochlear, but not in spiral afferent neurons. This mouse strain provides a novel approach to the identification and manipulation of P2X2-expressing cell types.
Collapse
|
40
|
Spanevello A, Beghé B, Visca D, Fabbri LM, Papi A. Chronic cough in adults. Eur J Intern Med 2020; 78:8-16. [PMID: 32434660 DOI: 10.1016/j.ejim.2020.03.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 03/17/2020] [Accepted: 03/19/2020] [Indexed: 12/12/2022]
Abstract
Cough, a defense mechanism for clearing the airways of secretions, exudate, or foreign bodies, may become a troublesome symptom. Chronic cough, one of the most frequent symptoms requiring medical attention, is often not due to identifiable causes in adults. Chronic productive cough defines chronic bronchitis, and thus is present in 100% of these patients, and frequently in patients with bronchiectasis, cystic fibrosis, and chronic infectious respiratory diseases. However, chronic cough is most frequently dry. Thus, chronic cough in adults is a difficult syndrome requiring multidisciplinary approaches, particularly to diagnose and treat the most frequent identifiable causes, but also to decide which patients may benefit by treating the central cough hypersensitivity by neuromodulatory therapy and/or non-pharmacologic treatment (speech pathology therapy). Recent guidelines provide algorithms for diagnosis and assessment of cough severity; particularly chronic cough in adults. After excluding life-threatening diseases, chronic cough due to identifiable causes (triggers and/or diseases), particularly smoking and/or the most frequent diseases (asthma, chronic bronchitis, chronic obstructive pulmonary disease, eosinophilic bronchitis, and adverse reactions to drugs [angiotensin-converting enzyme inhibitors and sitagliptin]) should be treated by avoiding triggers and/or according to guidelines for each underlying disease. In patients with troublesome chronic cough due to unknown causes or persisting even after adequate avoidance of triggers, and/or treatment of the underlying disease(s), a symptomatic approach with neuromodulators and/or speech pathology therapy should be considered. Additional novel promising neuromodulatory agents in clinical development (e.g., P2X3 inhibitors) will hopefully become available in the near future.
Collapse
Affiliation(s)
- Antonio Spanevello
- Istituti Clinici Scientifici Maugeri IRCCS, Respiratory Rehabilitation of the Institute of Tradate, Tradate, Varese, Italy; Department of Medicine and Surgery, Respiratory Diseases, University of Insubria, Varese-Como, Italy
| | - Bianca Beghé
- Department of Medicine, Section of Respiratory Diseases, University of Modena and Reggio Emilia, Modena, Italy
| | - Dina Visca
- Istituti Clinici Scientifici Maugeri IRCCS, Respiratory Rehabilitation of the Institute of Tradate, Tradate, Varese, Italy; Department of Medicine and Surgery, Respiratory Diseases, University of Insubria, Varese-Como, Italy
| | - Leonardo M Fabbri
- Azienda Ospedaliera Universitaria, Department of Medical Sciences, Section of Cardiorespiratory and Internal Medicine, University of Ferrara, Via Aldo Moro 8, 44124 Cona Ferrara, Italy.
| | - Alberto Papi
- Azienda Ospedaliera Universitaria, Department of Medical Sciences, Section of Cardiorespiratory and Internal Medicine, University of Ferrara, Via Aldo Moro 8, 44124 Cona Ferrara, Italy
| |
Collapse
|
41
|
Driessen AK, Devlin AC, Lundy FT, Martin SL, Sergeant GP, Mazzone SB, McGarvey LP. Perspectives on neuroinflammation contributing to chronic cough. Eur Respir J 2020; 56:13993003.00758-2020. [DOI: 10.1183/13993003.00758-2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 06/30/2020] [Indexed: 02/07/2023]
Abstract
Chronic cough can be a troublesome clinical problem. Current thinking is that increased activity and/or enhanced sensitivity of the peripheral and central neural pathways mediates chronic cough via processes similar to those associated with the development of chronic pain. While inflammation is widely thought to be involved in the development of chronic cough, the true mechanisms causing altered neural activity and sensitisation remain largely unknown. In this back-to-basics perspective article we explore evidence that inflammation in chronic cough may, at least in part, involve neuroinflammation orchestrated by glial cells of the nervous system. We summarise the extensive evidence for the role of both peripheral and central glial cells in chronic pain, and hypothesise that the commonalities between pain and cough pathogenesis and clinical presentation warrant investigations into the neuroinflammatory mechanisms that contribute to chronic cough. We open the debate that glial cells may represent an underappreciated therapeutic target for controlling troublesome cough in disease.
Collapse
|
42
|
Bookout AL, Gautron L. Characterization of a cell bridge variant connecting the nodose and superior cervical ganglia in the mouse: Prevalence, anatomical features, and practical implications. J Comp Neurol 2020; 529:111-128. [PMID: 32356570 DOI: 10.1002/cne.24936] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 04/08/2020] [Accepted: 04/19/2020] [Indexed: 12/22/2022]
Abstract
While autonomic ganglia have been extensively studied in rats instead of mice, there is renewed interest in the anatomy of the mouse autonomic nervous system. This study examined the prevalence and anatomical features of a cell bridge linking two autonomic ganglia of the neck, namely, the nodose ganglion (NG) and the superior cervical ganglion (SCG) in a cohort of C57BL/6J mice. We identified a cell bridge between the NG and the cranial pole of the SCG. This cell bridge was tubular shaped with an average length and width of 700 and 240 μm, respectively. The cell bridge was frequently unilateral and significantly more prevalent in the ganglionic masses from males (38%) than females (21%). On each of its extremities, it contained a mixed of vagal afferents and postganglionic sympathetic neurons. The two populations of neurons abruptly replaced each other in the middle of the cell bridge. We examined the mRNA expression for selected autonomic markers in samples of the NG with or without cell bridge. Our results indicated that the cell bridge was enriched in both markers of postganglionic sympathetic and vagal afferents neurons. Lastly, using FluoroGold microinjection into the NG, we found that the existence of a cell bridge may occasionally lead to the inadvertent contamination of the SCG. In summary, this study describes the anatomy of a cell bridge variant consisting of the fusion of the mouse NG and SCG. The practical implications of our observations are discussed with respect to studies of the mouse vagal afferents, an area of research of increasing popularity.
Collapse
Affiliation(s)
- Angie L Bookout
- Division of Hypothalamic Research and Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Laurent Gautron
- Division of Hypothalamic Research and Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
43
|
Driessen AK, McGovern AE, Behrens R, Moe AAK, Farrell MJ, Mazzone SB. A role for neurokinin 1 receptor expressing neurons in the paratrigeminal nucleus in bradykinin-evoked cough in guinea-pigs. J Physiol 2020; 598:2257-2275. [PMID: 32237239 DOI: 10.1113/jp279644] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 03/16/2020] [Indexed: 12/21/2022] Open
Abstract
KEY POINTS Airway projecting sensory neurons arising from the jugular vagal ganglia terminate centrally in the brainstem paratrigeminal nucleus, synapsing upon neurons expressing the neurokinin 1 receptor. This study aimed to assess the involvement of paratrigeminal neurokinin 1 receptor neurons in the regulation of cough, breathing and airway defensive responses. Lesioning neurokinin 1 receptor expressing paratrigeminal neurons significantly reduced cough evoked by inhaled bradykinin but not inhaled ATP or tracheal mechanical stimulation. The reduction in bradykinin-evoked cough was not accompanied by changes in baseline or evoked respiratory variables (e.g. frequency, volume or timing), animal avoidance behaviours or the laryngeal apnoea reflex. These findings warrant further investigations into targeting the jugular ganglia and paratrigeminal nucleus as a therapy for treating cough in disease. ABSTRACT Jugular vagal ganglia sensory neurons innervate the large airways and are thought to mediate cough and associated perceptions of airway irritations to a range of chemical irritants. The central terminals of jugular sensory neurons lie within the brainstem paratrigeminal nucleus, where postsynaptic neurons can be differentiated based on the absence or presence of the neurokinin 1 (NK1) receptor. Therefore, in the present study, we set out to test the hypothesis that NK1 receptor expressing paratrigeminal neurons play a role in cough evoked by inhaled chemical irritants. To test this, we performed selective neurotoxin lesions of NK1 receptor expressing neurons in the paratrigeminal nucleus in guinea-pigs using substance P conjugated to saporin (SSP-SAP). Sham lesion control or SSP-SAP lesion guinea-pigs received nebulised challenges, with the pan-nociceptor stimulant bradykinin or the nodose ganglia specific stimulant adenosine 5'-triphosphate (ATP), in conscious whole-body plethysmography to study cough and associated behaviours. Laryngeal apnoea reflexes and cough evoked by mechanical stimulation of the trachea were additionally investigated in anaesthetised guinea-pigs. SSP-SAP significantly and selectively reduced the number of NK1 receptor expressing neurons in the paratrigeminal nucleus. This was associated with a significant reduction in bradykinin-evoked cough, but not ATP-evoked cough, mechanical cough or laryngeal apnoeic responses. These data provide further evidence for a role of jugular vagal pathways in cough, and additionally suggest an involvement of NK1 receptor expressing neurons in the paratrigeminal nucleus. Therefore, this neural pathway may provide novel therapeutic opportunities to treat conditions of chronic cough.
Collapse
Affiliation(s)
- Alexandria K Driessen
- Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Alice E McGovern
- Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Robert Behrens
- Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Aung Aung Kywe Moe
- Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Michael J Farrell
- Department of Medical Imaging and Radiation Sciences, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
| | - Stuart B Mazzone
- Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, VIC, 3010, Australia
| |
Collapse
|
44
|
Mapping of Sensory Nerve Subsets within the Vagal Ganglia and the Brainstem Using Reporter Mice for Pirt, TRPV1, 5-HT3, and Tac1 Expression. eNeuro 2020; 7:ENEURO.0494-19.2020. [PMID: 32060036 PMCID: PMC7294455 DOI: 10.1523/eneuro.0494-19.2020] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 01/09/2020] [Accepted: 01/30/2020] [Indexed: 11/21/2022] Open
Abstract
Vagal afferent sensory nerves, originating in jugular and nodose ganglia, are composed of functionally distinct subsets whose activation evokes distinct thoracic and abdominal reflex responses. We used Cre-expressing mouse strains to identify specific vagal afferent populations and map their central projections within the brainstem. We show that Pirt is expressed in virtually all vagal afferents; whereas, 5-HT3 is expressed only in nodose neurons, with little expression in jugular neurons. Transient receptor potential vanilloid 1 (TRPV1), the capsaicin receptor, is expressed in a subset of small nodose and jugular neurons. Tac1, the gene for tachykinins, is expressed predominantly in jugular neurons, some of which also express TRPV1. Vagal fibers project centrally to the nucleus tractus solitarius (nTS), paratrigeminal complex, area postrema, and to a limited extent the dorsal motor nucleus of the vagus. nTS subnuclei preferentially receive projections by specific afferent subsets, with TRPV1+ fibers terminating in medial and dorsal regions predominantly caudal of obex, whereas TRPV1− fibers terminate in ventral and lateral regions throughout the rostral–caudal aspect of the medulla. Many vagal Tac1+ afferents (mostly derived from the jugular ganglion) terminate in the nTS. The paratrigeminal complex was the target of multiple vagal afferent subsets. Importantly, lung-specific TRPV1+ and Tac1+ afferent terminations were restricted to the caudal medial nTS, with no innervation of other medulla regions. In summary, this study identifies the specific medulla regions innervated by vagal afferent subsets. The distinct terminations provide a neuroanatomic substrate for the diverse range of reflexes initiated by vagal afferent activation.
Collapse
|
45
|
Matamales M, McGovern AE, Mi JD, Mazzone SB, Balleine BW, Bertran-Gonzalez J. Local D2- to D1-neuron transmodulation updates goal-directed learning in the striatum. Science 2020; 367:549-555. [PMID: 32001651 DOI: 10.1126/science.aaz5751] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 12/19/2019] [Indexed: 01/06/2023]
Abstract
Extinction learning allows animals to withhold voluntary actions that are no longer related to reward and so provides a major source of behavioral control. Although such learning is thought to depend on dopamine signals in the striatum, the way the circuits that mediate goal-directed control are reorganized during new learning remains unknown. Here, by mapping a dopamine-dependent transcriptional activation marker in large ensembles of spiny projection neurons (SPNs) expressing dopamine receptor type 1 (D1-SPNs) or 2 (D2-SPNs) in mice, we demonstrate an extensive and dynamic D2- to D1-SPN transmodulation across the striatum that is necessary for updating previous goal-directed learning. Our findings suggest that D2-SPNs suppress the influence of outdated D1-SPN plasticity within functionally relevant striatal territories to reshape volitional action.
Collapse
Affiliation(s)
- Miriam Matamales
- Decision Neuroscience Laboratory, School of Psychology, University of New South Wales, Sydney, NSW, Australia.
| | - Alice E McGovern
- Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, VIC, Australia.,School of Biomedical Sciences, University of Queensland, St Lucia, QLD, Australia
| | - Jia Dai Mi
- Department of Women and Children's Health, Faculty of Life Sciences and Medicine, King's College London, London SE1 7EH, UK
| | - Stuart B Mazzone
- Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, VIC, Australia.,School of Biomedical Sciences, University of Queensland, St Lucia, QLD, Australia
| | - Bernard W Balleine
- Decision Neuroscience Laboratory, School of Psychology, University of New South Wales, Sydney, NSW, Australia
| | - Jesus Bertran-Gonzalez
- Decision Neuroscience Laboratory, School of Psychology, University of New South Wales, Sydney, NSW, Australia.
| |
Collapse
|
46
|
Undem BJ, Sun H. Molecular/Ionic Basis of Vagal Bronchopulmonary C-Fiber Activation by Inflammatory Mediators. Physiology (Bethesda) 2020; 35:57-68. [PMID: 31799905 PMCID: PMC6985783 DOI: 10.1152/physiol.00014.2019] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 05/30/2019] [Accepted: 06/03/2019] [Indexed: 12/11/2022] Open
Abstract
Stimulation of bronchopulmonary vagal afferent C fibers by inflammatory mediators can lead to coughing, chest tightness, and changes in breathing pattern, as well as reflex bronchoconstriction and secretions. These responses serve a defensive function in healthy lungs but likely contribute to many of the signs and symptoms of inflammatory airway diseases. A better understanding of the mechanisms underlying the activation of bronchopulmonary C-fiber terminals may lead to novel therapeutics that would work in an additive or synergic manner with existing anti-inflammatory strategies.
Collapse
Affiliation(s)
| | - Hui Sun
- Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
47
|
Phenotypic distinctions between the nodose and jugular TRPV1-positive vagal sensory neurons in the cynomolgus monkey. Neuroreport 2019; 30:533-537. [PMID: 30896676 DOI: 10.1097/wnr.0000000000001231] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Vagal capsaicin-sensitive afferent C-fibers play an important role in the maintenance of visceral homeostasis and contribute to symptoms in visceral diseases. Based on their developmental origin two functionally distinct types of vagal C-fibers are recognized: those with neurons in the vagal nodose ganglia (derived from epibranchial placodes) and in the vagal jugular ganglia (from neural crest). Studies in nonprimate species demonstrated that the vagal nodose and jugular C-fibers differ in activation profile, neurotrophic regulation, and expression of neurotransmitters. We hypothesized that the expression of selected markers related to key phenotypic properties of vagal C-fibers in the cynomolgus monkey is similar to that reported in nonprimate species. We performed single-cell RT-PCR on nodose and jugular putative C-fiber (TRPV1-positive) neurons isolated from the cynomolgus monkey. We found that the expression of purinergic P2X receptors that underlie selective responsiveness of nodose C-fiber terminals to ATP was conserved in that P2X2 and P2X3 subunits were expressed in nodose, but only P2X3 subunit was expressed in jugular TRPV1-positive neurons. Also conserved was the preferential expression of neurotrophic receptor TrkB in the nodose and preprotachykinin-A in the jugular TRPV1-positive neurons. Several key distinctions in gene expression between nodose and jugular TRPV1-positive (C-fiber) neurons that have been noted in mice, rats, and guinea pigs, are conserved in the cynomolgus monkey. Our results support the translatability of distinct vagal C-fiber phenotypes to primates.
Collapse
|
48
|
Transcriptional Profiling of Individual Airway Projecting Vagal Sensory Neurons. Mol Neurobiol 2019; 57:949-963. [DOI: 10.1007/s12035-019-01782-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 09/12/2019] [Indexed: 12/11/2022]
|
49
|
Kanezaki M, Ebihara S. Effect of facial skin cooling induced by a handheld fan on the cough reflex threshold and urge to cough induced by citric acid. ERJ Open Res 2019; 5:00089-2019. [PMID: 31637251 PMCID: PMC6791964 DOI: 10.1183/23120541.00089-2019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 08/03/2019] [Indexed: 01/04/2023] Open
Abstract
Chronic cough is a condition that impairs the quality of life of affected patients and may persist despite optimal intervention. Thus, a novel therapeutic approach with broad clinical utility based on the nature of the cough reflex is required. Facial skin cooling induced by a handheld fan alleviates the cough reflex and the urge to cough induced by citric acidhttp://bit.ly/2m1kzAP
Collapse
Affiliation(s)
- Masashi Kanezaki
- Dept of Physical Therapy, Faculty of Health Care Sciences, Himeji Dokkyo University, Himeji, Japan
| | - Satoru Ebihara
- Dept of Rehabilitation Medicine, Toho University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
50
|
Wallace E, Guiu Hernandez E, Ang A, Macrae P. Quantifying test-retest variability of natural and suppressed citric acid cough thresholds and urge to cough ratings. Pulm Pharmacol Ther 2019; 58:101838. [DOI: 10.1016/j.pupt.2019.101838] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 09/02/2019] [Accepted: 09/04/2019] [Indexed: 11/25/2022]
|