1
|
Bu Y, Burks J, Yang K, Prince J, Borna A, Coe CL, Simmons A, Tu XM, Baker D, Kimball D, Rao R, Shah V, Huang M, Schwindt P, Coleman TP, Lerman I. Non-invasive ventral cervical magnetoneurography as a proxy of in vivo lipopolysaccharide-induced inflammation. Commun Biol 2024; 7:893. [PMID: 39075164 PMCID: PMC11286963 DOI: 10.1038/s42003-024-06435-8] [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: 09/06/2023] [Accepted: 06/10/2024] [Indexed: 07/31/2024] Open
Abstract
Maintenance of autonomic homeostasis is continuously calibrated by sensory fibers of the vagus nerve and sympathetic chain that convey compound action potentials (CAPs) to the central nervous system. Lipopolysaccharide (LPS) intravenous challenge reliably elicits a robust inflammatory response that can resemble systemic inflammation and acute endotoxemia. Here, we administered LPS intravenously in nine healthy subjects while recording ventral cervical magnetoneurography (vcMNG)-derived CAPs at the rostral Right Nodose Ganglion (RNG) and the caudal Right Carotid Artery (RCA) with optically pumped magnetometers (OPM). We observed vcMNG RNG and RCA neural firing rates that tracked changes in TNF-α levels in the systemic circulation. Further, endotype subgroups based on high and low IL-6 responders segregate RNG CAP frequency (at 30-120 min) and based on high and low IL-10 response discriminate RCA CAP frequency (at 0-30 min). These vcMNG tools may enhance understanding and management of the neuroimmune axis that can guide personalized treatment based on an individual's distinct endophenotype.
Collapse
Affiliation(s)
- Yifeng Bu
- Department of Electrical and Computer Engineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Jamison Burks
- Department of Bioengineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Kun Yang
- Division of Biostatistics and Bioinformatics, University of California San Diego, La Jolla, CA, 92093, USA
| | - Jacob Prince
- Department of Bioengineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Amir Borna
- Quantum Information Sciences, Sandia National Laboratories, Albuquerque, NM, 87123, USA
| | - Christopher L Coe
- Department of Psychology, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Alan Simmons
- Center for Stress and Mental Health (CESAMH) VA San Diego, La Jolla, CA, 92093, USA
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA
| | - Xin M Tu
- Division of Biostatistics and Bioinformatics, University of California San Diego, La Jolla, CA, 92093, USA
| | - Dewleen Baker
- Center for Stress and Mental Health (CESAMH) VA San Diego, La Jolla, CA, 92093, USA
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA
| | - Donald Kimball
- Department of Electrical and Computer Engineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Ramesh Rao
- Department of Electrical and Computer Engineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Vishal Shah
- Quspin Laboratory Head Quarters, Boulder, CO, 80305, USA
| | - Mingxiong Huang
- Department of Electrical and Computer Engineering, University of California San Diego, La Jolla, CA, 92093, USA
- Department of Radiology, University of California San Diego, La Jolla, CA, 92093, USA
| | - Peter Schwindt
- Quantum Information Sciences, Sandia National Laboratories, Albuquerque, NM, 87123, USA
| | - Todd P Coleman
- Department of Electrical and Computer Engineering, University of California San Diego, La Jolla, CA, 92093, USA
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Imanuel Lerman
- Department of Electrical and Computer Engineering, University of California San Diego, La Jolla, CA, 92093, USA.
- Center for Stress and Mental Health (CESAMH) VA San Diego, La Jolla, CA, 92093, USA.
- InflammaSense Incorporated Head Quarters, La Jolla, CA, 92093, USA.
- Department of Anesthesiology, University of California San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
2
|
Merighi A. Brain-Derived Neurotrophic Factor, Nociception, and Pain. Biomolecules 2024; 14:539. [PMID: 38785946 PMCID: PMC11118093 DOI: 10.3390/biom14050539] [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: 02/08/2024] [Revised: 04/26/2024] [Accepted: 04/26/2024] [Indexed: 05/25/2024] Open
Abstract
This article examines the involvement of the brain-derived neurotrophic factor (BDNF) in the control of nociception and pain. BDNF, a neurotrophin known for its essential role in neuronal survival and plasticity, has garnered significant attention for its potential implications as a modulator of synaptic transmission. This comprehensive review aims to provide insights into the multifaceted interactions between BDNF and pain pathways, encompassing both physiological and pathological pain conditions. I delve into the molecular mechanisms underlying BDNF's involvement in pain processing and discuss potential therapeutic applications of BDNF and its mimetics in managing pain. Furthermore, I highlight recent advancements and challenges in translating BDNF-related research into clinical practice.
Collapse
Affiliation(s)
- Adalberto Merighi
- Department of Veterinary Sciences, University of Turin, 10095 Turin, Italy
| |
Collapse
|
3
|
Xu Y, Cao S, Wang SF, Ma W, Gou XJ. Zhisou powder suppresses airway inflammation in LPS and CS-induced post-infectious cough model mice via TRPA1/TRPV1 channels. JOURNAL OF ETHNOPHARMACOLOGY 2024; 324:117741. [PMID: 38224794 DOI: 10.1016/j.jep.2024.117741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 01/05/2024] [Accepted: 01/08/2024] [Indexed: 01/17/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Zhisou Powder (ZSP), a traditional Chinese medicine (TCM) prescription, has been widely used in the clinic for the treatment of post-infectious cough (PIC). However, the exact mechanism is not clear. AIM OF THE STUDY The aim of this study was to investigate the ameliorative effect of ZSP on PIC in mice. The possible mechanisms of action were screened based on network pharmacology, and the potential mechanisms were explored through molecular docking and in vivo experimental validation. MATERIALS AND METHODS Lipopolysaccharide (LPS) (80μg/50 μL) was used to induce PIC in mice, followed by daily exposure to cigarette smoke (CS) for 30 min for 30 d to establish PIC model. The effects of ZSP on PIC mice were observed by detecting the number of coughs and cough latency, peripheral blood and bronchoalveolar lavage fluid (BALF) inflammatory cell counts, enzyme-linked immunosorbent assay (ELISA), and histological analysis. The core targets and key pathways of ZSP on PIC were analyzed using network pharmacology, and TRPA1 and TRPV1 were validated using RT-qPCR and western blotting assays. RESULTS ZSP effectively reduced the number of coughs and prolonged the cough latency in PIC mice. Airway inflammation was alleviated by reducing the expression levels of the inflammatory mediators TNF-α and IL-1β. ZSP modulated the expression of Substance P, Calcitonin gene-related peptide (CGRP), and nerve growth factor (NGF) in BALF. Based on the results of network pharmacology, the mechanism of action of ZSP may exert anti-neurogenic airway-derived inflammation by regulating the expression of TRPA1 and TRPV1 through the natural active ingredients α-spinastero, shionone and didehydrotuberostemonine. CONCLUSION ZSP exerts anti-airway inflammatory effects through inhibition of TRPA1/TRPV1 channels regulating neuropeptides to alleviate cough hypersensitivity and has a favorable therapeutic effect on PIC model mice. It provides theoretical evidence for the clinical application of ZSP.
Collapse
Affiliation(s)
- Yuan Xu
- Respiratory Department and Central Laboratory, Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine of Shanghai, Shanghai 201999, China; School of Pharmacy, Shaanxi Univesity of Chinese Medicine, Shaanxi, Xianyang 712046, China
| | - Shan Cao
- Respiratory Department and Central Laboratory, Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine of Shanghai, Shanghai 201999, China
| | - Shu-Fei Wang
- School of Pharmacy, Shaanxi Univesity of Chinese Medicine, Shaanxi, Xianyang 712046, China
| | - Wei Ma
- Respiratory Department and Central Laboratory, Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine of Shanghai, Shanghai 201999, China.
| | - Xiao-Jun Gou
- Respiratory Department and Central Laboratory, Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine of Shanghai, Shanghai 201999, China.
| |
Collapse
|
4
|
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
|
5
|
Kim JS, Ru F, Meeker S, Undem BJ. Direct activation of airway sensory C-fibers by SARS-CoV-2 S1 spike protein. Physiol Rep 2023; 11:e15900. [PMID: 38123162 PMCID: PMC10733116 DOI: 10.14814/phy2.15900] [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: 10/02/2023] [Revised: 12/02/2023] [Accepted: 12/04/2023] [Indexed: 12/23/2023] Open
Abstract
Respiratory viral infection can lead to activation of sensory afferent nerves as indicated by the consequential sore throat, sneezing, coughing, and reflex secretions. In addition to causing troubling symptoms, sensory nerve activation likely accelerates viral spreading. The mechanism how viruses activate sensory nerve terminals during infection is unknown. In this study, we investigate whether coronavirus spike protein activates sensory nerves terminating in the airways. We used isolated vagally-innervated mouse trachea-lung preparation for two-photon microscopy and extracellular electrophysiological recordings. Using two-photon Ca2+ imaging, we evaluated a total number of 786 vagal bronchopulmonary nerves in six experiments. Approximately 49% of the sensory fibers were activated by S1 protein (4 μg/mL intratracheally). Extracellular nerve recording showed the S1 protein evoked action potential discharge in sensory C-fibers; of 39 airway C-fibers (one fiber per mouse), 17 were activated. Additionally, Fura-2 Ca2+ imaging was performed on neurons dissociated from vagal sensory ganglia (n = 254 from 22 mice). The result showed that 63% of neurons responded to S1 protein. SARS-CoV-2 S1 protein can lead to direct activation of sensory C-fiber nerve terminals in the bronchopulmonary tract. Direct activation of C-fibers may contribute to coronavirus symptoms, and amplify viral spreading in a population.
Collapse
Affiliation(s)
- Joyce S. Kim
- Department of MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Fei Ru
- Department of MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Sonya Meeker
- Department of MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Bradley J. Undem
- Department of MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| |
Collapse
|
6
|
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
|
7
|
Kim JS, Sun H, Meeker S, Undem BJ. Role of Na V 1.9 in inflammatory mediator-induced activation of mouse airway vagal C-fibres. J Physiol 2023; 601:1139-1150. [PMID: 36750759 PMCID: PMC10023385 DOI: 10.1113/jp283751] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 01/23/2023] [Indexed: 02/09/2023] Open
Abstract
The influence of NaV 1.9 on inflammatory mediator-induced activation of airway vagal nodose C-fibres was evaluated by comparing responses in wild-type versus NaV 1.9-/- mice. A single-cell RT-PCR analysis indicated that virtually all nodose C-fibre neurons expressed NaV 1.9 (SCN11A) mRNA. Using extracellular electrophysiological recordings in an isolated vagally innervated mouse trachea-lung preparation, it was noted that mediators acting via G protein-coupled receptors (PAR2), or ionotropic receptors (P2×3) were 70-85% less effective in evoking action potential discharge in the absence of NaV 1.9. However, there was no difference in action potential discharge between wild-type and NaV 1.9-/- when the stimulus was a rapid punctate mechanical stimulus. An analysis of the passive and active properties of isolated nodose neurons revealed no difference between neurons from wild-type and NaV 1.9-/- mice, with the exception of a modest difference in the duration of the afterhyperpolarization. There was also no difference in the amount of current required to evoke action potentials (rheobase) or the action potential voltage threshold. The inward current evoked by the chemical mediator by a P2×3 agonist was the same in wild-type versus NaV 1.9-/- neurons. However, the current was sufficient to evoke action potential only in the wild-type neurons. The data support the speculation that NaV 1.9 could be an attractive therapeutic target for inflammatory airway disease by selectively inhibiting inflammatory mediator-associated vagal C-fibre activation. KEY POINTS: Inflammatory mediators were much less effective in activating the terminals of vagal airway C-fibres in mice lacking NaV 1.9. The active and passive properties of nodose neurons were the same between wild-type neurons and NaV 1.9-/- neurons. Nerves lacking NaV 1.9 responded, normally, with action potential discharge to rapid punctate mechanical stimulation of the terminals or the rapid stimulation of the cell bodies with inward current injections. NaV 1.9 channels could be an attractive target to selectively inhibit vagal nociceptive C-fibre activation evoked by inflammatory mediators without blocking the nerves' responses to the potentially hazardous stimuli associated with aspiration.
Collapse
Affiliation(s)
- Joyce S Kim
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hui Sun
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sonya Meeker
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Bradley J Undem
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
8
|
From Low-Grade Inflammation in Osteoarthritis to Neuropsychiatric Sequelae: A Narrative Review. Int J Mol Sci 2022; 23:ijms232416031. [PMID: 36555670 PMCID: PMC9784931 DOI: 10.3390/ijms232416031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/08/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Nowadays, osteoarthritis (OA), a common, multifactorial musculoskeletal disease, is considered to have a low-grade inflammatory pathogenetic component. Lately, neuropsychiatric sequelae of the disease have gained recognition. However, a link between the peripheral inflammatory process of OA and the development of neuropsychiatric pathology is not completely understood. In this review, we provide a narrative that explores the development of neuropsychiatric disease in the presence of chronic peripheral low-grade inflammation with a focus on its signaling to the brain. We describe the development of a pro-inflammatory environment in the OA-affected joint. We discuss inflammation-signaling pathways that link the affected joint to the central nervous system, mainly using primary sensory afferents and blood circulation via circumventricular organs and cerebral endothelium. The review describes molecular and cellular changes in the brain, recognized in the presence of chronic peripheral inflammation. In addition, changes in the volume of gray matter and alterations of connectivity important for the assessment of the efficacy of treatment in OA are discussed in the given review. Finally, the narrative considers the importance of the use of neuropsychiatric diagnostic tools for a disease with an inflammatory component in the clinical setting.
Collapse
|
9
|
Meerschaert KA, Edwards BS, Epouhe AY, Jefferson B, Friedman R, Babyok OL, Moy JK, Kehinde F, Liu C, Workman CJ, Vignali DAA, Albers KM, Koerber HR, Gold MS, Davis BM, Scheff NN, Saloman JL. Neuronally expressed PDL1, not PD1, suppresses acute nociception. Brain Behav Immun 2022; 106:233-246. [PMID: 36089217 PMCID: PMC10343937 DOI: 10.1016/j.bbi.2022.09.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 08/21/2022] [Accepted: 09/03/2022] [Indexed: 11/20/2022] Open
Abstract
PDL1 is a protein that induces immunosuppression by binding to PD1 expressed on immune cells. In line with historical studies, we found that membrane-bound PD1 expression was largely restricted to immune cells; PD1 was not detectable at either the mRNA or protein level in peripheral neurons using single neuron qPCR, immunolabeling and flow cytometry. However, we observed widespread expression of PDL1 in both sensory and sympathetic neurons that could have important implications for patients receiving immunotherapies targeting this pathway that include unexpected autonomic and sensory related effects. While signaling pathways downstream of PD1 are well established, little to no information is available regarding the intracellular signaling downstream of membrane-bound PDL1 (also known as reverse signaling). Here, we administered soluble PD1 to engage neuronally expressed PDL1 and found that PD1 significantly reduced nocifensive behaviors evoked by algogenic capsaicin. We used calcium imaging to examine the underlying neural mechanism of this reduction and found that exogenous PD1 diminished TRPV1-dependent calcium transients in dissociated sensory neurons. Furthermore, we observed a reduction in membrane expression of TRPV1 following administration of PD1. Exogenous PD1 had no effect on pain-related behaviors in sensory neuron specific PDL1 knockout mice. These data indicate that neuronal PDL1 activation is sufficient to modulate sensitivity to noxious stimuli and as such, may be an important homeostatic mechanism for regulating acute nociception.
Collapse
Affiliation(s)
- Kimberly A Meerschaert
- Pittsburgh Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Center for Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Brian S Edwards
- Pittsburgh Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Ariel Y Epouhe
- Pittsburgh Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Center for Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Bahiyyah Jefferson
- Pittsburgh Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Robert Friedman
- Pittsburgh Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Olivia L Babyok
- Pittsburgh Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Center for Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Jamie K Moy
- Pittsburgh Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Center for Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Faith Kehinde
- Pittsburgh Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Chang Liu
- Department of Immunology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States; Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, United States
| | - Creg J Workman
- Department of Immunology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States; Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, United States
| | - Dario A A Vignali
- Department of Immunology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States; Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, United States; Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, United States
| | - Kathryn M Albers
- Pittsburgh Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Center for Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - H Richard Koerber
- Pittsburgh Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Center for Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Michael S Gold
- Pittsburgh Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Center for Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Brian M Davis
- Pittsburgh Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Center for Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Nicole N Scheff
- Pittsburgh Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Center for Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Biobehavioral Cancer Control Program, UPMC Hillman Cancer Center, Pittsburgh, PA, United States
| | - Jami L Saloman
- Pittsburgh Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Center for Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Biobehavioral Cancer Control Program, UPMC Hillman Cancer Center, Pittsburgh, PA, United States; Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.
| |
Collapse
|
10
|
Sykes DL, Zhang M, Morice AH. Treatment of chronic cough: P2X3 receptor antagonists and beyond. Pharmacol Ther 2022; 237:108166. [DOI: 10.1016/j.pharmthera.2022.108166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/22/2022] [Accepted: 03/02/2022] [Indexed: 10/18/2022]
|
11
|
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: 94] [Impact Index Per Article: 47.0] [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
|
12
|
Sun H, Patil MJ, Ru F, Meeker S, Undem BJ. K
V
1/D‐type potassium channels inhibit the excitability of bronchopulmonary vagal afferent nerves. J Physiol 2022; 600:2953-2971. [PMID: 35430729 PMCID: PMC9203938 DOI: 10.1113/jp282803] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 04/04/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract The KV1/D‐type potassium current (ID) is an important determinant of neuronal excitability. This study explored whether and how ID channels regulate the activation of bronchopulmonary vagal afferent nerves. The single‐neuron RT‐PCR assay revealed that nearly all mouse bronchopulmonary nodose neurons expressed the transcripts of α‐dendrotoxin (α‐DTX)‐sensitive, ID channel‐forming KV1.1, KV1.2 and/or KV1.6 α‐subunits, with the expression of KV1.6 being most prevalent. Patch‐clamp recordings showed that ID, defined as the α‐DTX‐sensitive K+ current, activated at voltages slightly more negative than the resting membrane potential in lung‐specific nodose neurons and displayed little inactivation at subthreshold voltages. Inhibition of ID channels by α‐DTX depolarized the lung‐specific nodose neurons and caused an increase in input resistance, decrease in rheobase, as well as increase in action potential number and firing frequency in response to suprathreshold current steps. Application of α‐DTX to the lungs via trachea in the mouse ex vivo vagally innervated trachea–lungs preparation led to action potential discharges in nearly half of bronchopulmonary nodose afferent nerve fibres, including nodose C‐fibres, as detected by the two‐photon microscopic Ca2+ imaging technique and extracellular electrophysiological recordings. In conclusion, ID channels act as a critical brake on the activation of bronchopulmonary vagal afferent nerves by stabilizing the membrane potential, counterbalancing the subthreshold depolarization and promoting the adaptation of action potential firings. Down‐regulation of ID channels, as occurs in various inflammatory diseases, may contribute to the enhanced C‐fibre activity in airway diseases that are associated with excessive coughing, dyspnoea, and reflex bronchospasm and secretions. Key points The α‐dendrotoxin (α‐DTX)‐sensitive D‐type K+ current (ID) is an important determinant of neuronal excitability. Nearly all bronchopulmonary nodose afferent neurons in the mouse express ID and the transcripts of α‐DTX‐sensitive, ID channel‐forming KV1.1, KV1.2 and/or KV1.6 α‐subunits. Inhibition of ID channels by α‐DTX depolarizes the bronchopulmonary nodose neurons, reduces the minimal depolarizing current needed to evoke an action potential (AP) and increases AP number and AP firing frequency in response to suprathreshold stimulations. Application of α‐DTX to the lungs ex vivo elicits AP discharges in about half of bronchopulmonary nodose C‐fibre terminals.
Our novel finding that ID channels act as a critical brake on the activation of bronchopulmonary vagal afferent nerves suggests that their down‐regulation, as occurs in various inflammatory diseases, may contribute to the enhanced C‐fibre activity in airway inflammation associated with excessive respiratory symptoms.
Collapse
Affiliation(s)
- Hui Sun
- Division of Allergy and Clinical Immunology Department of Medicine Johns Hopkins University School of Medicine 5501 Hopkins Bayview Circle Baltimore 21224
| | - Mayur J. Patil
- Division of Allergy and Clinical Immunology Department of Medicine Johns Hopkins University School of Medicine 5501 Hopkins Bayview Circle Baltimore 21224
| | - Fei Ru
- Division of Allergy and Clinical Immunology Department of Medicine Johns Hopkins University School of Medicine 5501 Hopkins Bayview Circle Baltimore 21224
| | - Sonya Meeker
- Division of Allergy and Clinical Immunology Department of Medicine Johns Hopkins University School of Medicine 5501 Hopkins Bayview Circle Baltimore 21224
| | - Bradley J. Undem
- Division of Allergy and Clinical Immunology Department of Medicine Johns Hopkins University School of Medicine 5501 Hopkins Bayview Circle Baltimore 21224
| |
Collapse
|
13
|
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: 15] [Impact Index Per Article: 7.5] [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
|
14
|
Sun J, Zhan C, Deng Z, Luo W, Chen Q, Jiang M, Zhong N, Lai K. Expression of interferon-γ and its effect on cough hypersensitivity in chronic refractory cough patients. Thorax 2022; 77:621-624. [PMID: 34996851 DOI: 10.1136/thoraxjnl-2021-218403] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 12/07/2021] [Indexed: 11/03/2022]
Abstract
Chronic refractory cough (CRC) is characterised by cough hypersensitivity. Interferon-γ (IFN-γ) has been reported to induce calcium influx, action potentials of vagal neurons in vitro and cough response in guinea pigs. While the effect of IFN-γ in CRC patients remains unknown. Here, via flow-cytometry and inhalation cough challenge, we found CRC patients had significantly increased levels of sputum IFN-γ+CD4+ T cells, IFN-γ+CD8+ T cells as well as supernatant of IFN-γ. The average number of coughs in CRC patients increased as the concentration of inhaled IFN-γ went up in IFN-γ cough challenge. Two or more coughs and five or more coughs elicited by inhaled IFN-γ in CRC patients occurred in 7 of 10 and 2 of 10, respectively. Preinhaled IFN-γ (100 µg/mL) increased the capsaicin cough sensitivity in CRC patients but not healthy volunteers. Targeting IFN-γ may be a potential effective anti-tussive strategy in CRC patients.
Collapse
Affiliation(s)
- Jiayang Sun
- Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.,Department of Respiratory Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Chen Zhan
- Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zheng Deng
- Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Wei Luo
- Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Qiaoli Chen
- Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Mei Jiang
- Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Nanshan Zhong
- Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Kefang Lai
- Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
15
|
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: 33] [Impact Index Per Article: 16.5] [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
|
16
|
Rienzo M, Di Zazzo E, Casamassimi A, Gazzerro P, Perini G, Bifulco M, Abbondanza C. PRDM12 in Health and Diseases. Int J Mol Sci 2021; 22:ijms222112030. [PMID: 34769459 PMCID: PMC8585061 DOI: 10.3390/ijms222112030] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/03/2021] [Accepted: 11/04/2021] [Indexed: 11/18/2022] Open
Abstract
PRDM12 is a member of the PRDI-BF1 (positive regulatory domain I-binding factor 1) homologous domain (PRDM)-containing protein family, a subfamily of Kruppel-like zinc finger proteins, controlling key processes in the development of cancer. PRDM12 is expressed in a spatio-temporal manner in neuronal systems where it exerts multiple functions. PRDM12 is essential for the neurogenesis initiation and activation of a cascade of downstream pro-neuronal transcription factors in the nociceptive lineage. PRDM12 inactivation, indeed, results in a complete absence of the nociceptive lineage, which is essential for pain perception. Additionally, PRDM12 contributes to the early establishment of anorexigenic neuron identity and the maintenance of high expression levels of pro-opiomelanocortin, which impacts on the program bodyweight homeostasis. PRDMs are commonly involved in cancer, where they act as oncogenes/tumor suppressors in a “Yin and Yang” manner. PRDM12 is not usually expressed in adult normal tissues but its expression is re-activated in several cancer types. However, little information is currently available on PRDM12 expression in cancers and its mechanism of action has not been thoroughly described. In this review, we summarize the recent findings regarding PRDM12 by focusing on four main biological processes: neurogenesis, pain perception, oncogenesis and cell metabolism. Moreover, we wish to highlight the importance of future studies focusing on the PRDM12 signaling pathway(s) and its role in cancer onset and progression.
Collapse
Affiliation(s)
- Monica Rienzo
- Department of Environmental, Biological, and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, 81100 Caserta, Italy;
| | - Erika Di Zazzo
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, 86100 Campobasso, Italy;
| | - Amelia Casamassimi
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via L. De Crecchio, 80138 Naples, Italy;
- Correspondence:
| | - Patrizia Gazzerro
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Salerno, Italy;
| | - Giovanni Perini
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy;
| | - Maurizio Bifulco
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples “Federico II”, 80131 Naples, Italy;
| | - Ciro Abbondanza
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via L. De Crecchio, 80138 Naples, Italy;
| |
Collapse
|
17
|
Bob-Manuel J, Gautron L. Detection of G Protein-coupled Receptor Expression in Mouse Vagal Afferent Neurons using Multiplex In Situ Hybridization. J Vis Exp 2021:10.3791/62945. [PMID: 34605820 PMCID: PMC9235148 DOI: 10.3791/62945] [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] [Indexed: 10/31/2022] Open
Abstract
This study describes a protocol for the multiplex in situ hybridization (ISH) of the mouse jugular-nodose ganglia, with a particular emphasis on detecting the expression of G protein-coupled receptors (GPCRs). Formalin-fixed jugular-nodose ganglia were processed with the RNAscope technology to simultaneously detect the expression of two representative GPCRs (cholecystokinin and ghrelin receptors) in combination with one marker gene of either nodose (paired-like homeobox 2b, Phox2b) or jugular afferent neurons (PR domain zinc finger protein 12, Prdm12). Labeled ganglia were imaged using confocal microscopy to determine the distribution and expression patterns of the aforementioned transcripts. Briefly, Phox2b afferent neurons were found to abundantly express the cholecystokinin receptor (Cck1r) but not the ghrelin receptor (Ghsr). A small subset of Prdm12 afferent neurons was also found to express Ghsr and/or Cck1r. Potential technical caveats in the design, processing, and interpretation of multiplex ISH are discussed. The approach described in this article may help scientists in generating accurate maps of the transcriptional profiles of vagal afferent neurons.
Collapse
Affiliation(s)
- Johnson Bob-Manuel
- Center for Hypothalamic Research and Department of Internal Medicine, UTSouthwestern Medical Center at Dallas
| | - Laurent Gautron
- Center for Hypothalamic Research and Department of Internal Medicine, UTSouthwestern Medical Center at Dallas;
| |
Collapse
|
18
|
Proskocil BJ, Wai K, Lebold KM, Norgard MA, Michaelis KA, De La Torre U, Cook M, Marks DL, Fryer AD, Jacoby DB, Drake MG. TLR7 is expressed by support cells, but not sensory neurons, in ganglia. J Neuroinflammation 2021; 18:209. [PMID: 34530852 PMCID: PMC8447680 DOI: 10.1186/s12974-021-02269-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 08/31/2021] [Indexed: 11/10/2022] Open
Abstract
Background Toll-like receptor 7 (TLR7) is an innate immune receptor that detects viral single-stranded RNA and triggers the production of proinflammatory cytokines and type 1 interferons in immune cells. TLR7 agonists also modulate sensory nerve function by increasing neuronal excitability, although studies are conflicting whether sensory neurons specifically express TLR7. This uncertainty has confounded the development of a mechanistic understanding of TLR7 function in nervous tissues. Methods TLR7 expression was tested using in situ hybridization with species-specific RNA probes in vagal and dorsal root sensory ganglia in wild-type and TLR7 knockout (KO) mice and in guinea pigs. Since TLR7 KO mice were generated by inserting an Escherichia coli lacZ gene in exon 3 of the mouse TLR7 gene, wild-type and TLR7 (KO) mouse vagal ganglia were also labeled for lacZ. In situ labeling was compared to immunohistochemistry using TLR7 antibody probes. The effects of influenza A infection on TLR7 expression in sensory ganglia and in the spleen were also assessed. Results In situ probes detected TLR7 in the spleen and in small support cells adjacent to sensory neurons in the dorsal root and vagal ganglia in wild-type mice and guinea pigs, but not in TLR7 KO mice. TLR7 was co-expressed with the macrophage marker Iba1 and the satellite glial cell marker GFAP, but not with the neuronal marker PGP9.5, indicating that TLR7 is not expressed by sensory nerves in either vagal or dorsal root ganglia in mice or guinea pigs. In contrast, TLR7 antibodies labeled small- and medium-sized neurons in wild-type and TLR7 KO mice in a TLR7-independent manner. Influenza A infection caused significant weight loss and upregulation of TLR7 in the spleens, but not in vagal ganglia, in mice. Conclusion TLR7 is expressed by macrophages and satellite glial cells, but not neurons in sensory ganglia suggesting TLR7’s neuromodulatory effects are mediated indirectly via activation of neuronally-associated support cells, not through activation of neurons directly. Our data also suggest TLR7’s primary role in neuronal tissues is not related to antiviral immunity. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02269-x.
Collapse
Affiliation(s)
- Becky J Proskocil
- Division of Pulmonary and Critical Care Medicine, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, UHN67, Portland, OR, 97239, USA
| | - Karol Wai
- Division of Pulmonary and Critical Care Medicine, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, UHN67, Portland, OR, 97239, USA
| | - Katherine M Lebold
- Division of Pulmonary and Critical Care Medicine, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, UHN67, Portland, OR, 97239, USA
| | - Mason A Norgard
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, OR, USA
| | - Katherine A Michaelis
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, OR, USA
| | - Ubaldo De La Torre
- Division of Pulmonary and Critical Care Medicine, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, UHN67, Portland, OR, 97239, USA
| | - Madeline Cook
- Division of Pulmonary and Critical Care Medicine, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, UHN67, Portland, OR, 97239, USA
| | - Daniel L Marks
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, OR, USA
| | - Allison D Fryer
- Division of Pulmonary and Critical Care Medicine, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, UHN67, Portland, OR, 97239, USA
| | - David B Jacoby
- Division of Pulmonary and Critical Care Medicine, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, UHN67, Portland, OR, 97239, USA
| | - Matthew G Drake
- Division of Pulmonary and Critical Care Medicine, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, UHN67, Portland, OR, 97239, USA.
| |
Collapse
|
19
|
Verzele NAJ, Chua BY, Law CW, Zhang A, Ritchie ME, Wightman O, Edwards IN, Hulme KD, Bloxham CJ, Bielefeldt-Ohmann H, Trewella MW, Moe AAK, Chew KY, Mazzone SB, Short KR, McGovern AE. The impact of influenza pulmonary infection and inflammation on vagal bronchopulmonary sensory neurons. FASEB J 2021; 35:e21320. [PMID: 33660333 DOI: 10.1096/fj.202001509r] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 11/20/2020] [Accepted: 12/14/2020] [Indexed: 12/13/2022]
Abstract
Influenza A virus (IAV) is rapidly detected in the airways by the immune system, with resident parenchymal cells and leukocytes orchestrating viral sensing and the induction of antiviral inflammatory responses. The airways are innervated by heterogeneous populations of vagal sensory neurons which also play an important role in pulmonary defense. How these neurons respond to IAV respiratory infection remains unclear. Here, we use a murine model to provide the first evidence that vagal sensory neurons undergo significant transcriptional changes following a respiratory IAV infection. RNA sequencing on vagal sensory ganglia showed that IAV infection induced the expression of many genes associated with an antiviral and pro-inflammatory response and this was accompanied by a significant increase in inflammatory cell recruitment into the vagal ganglia. Assessment of gene expression in single-vagal sensory neurons confirmed that IAV infection induced a neuronal inflammatory phenotype, which was most prominent in bronchopulmonary neurons, and also evident in some neurons innervating other organs. The altered transcriptome could be mimicked by intranasal treatment with cytokines and the lung homogenates of infected mice, in the absence of infectious virus. These data argue that IAV pulmonary infection and subsequent inflammation induces vagal sensory ganglia neuroinflammation and this may have important implications for IAV-induced morbidity.
Collapse
Affiliation(s)
- Nathalie A J Verzele
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia
| | - Brendon Y Chua
- The Peter Doherty Institute for Infection and Immunity, Department of Microbiology and Immunology, University of Melbourne, Melbourne, VIC, Australia
| | - Charity W Law
- Epigenetics and Development Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Albert Zhang
- Epigenetics and Development Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Matthew E Ritchie
- Epigenetics and Development Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Oliver Wightman
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia
| | - Isaac N Edwards
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia
| | - Katina D Hulme
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia
| | - Conor J Bloxham
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia
| | - Helle Bielefeldt-Ohmann
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia.,Australian Infectious Diseases Research Centre, The University of Queensland, St Lucia, QLD, Australia
| | - Matthew W Trewella
- Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, VIC, Australia
| | - Aung Aung Kywe Moe
- Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, VIC, Australia
| | - Keng Yih Chew
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia
| | - Stuart B Mazzone
- Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, VIC, Australia
| | - Kirsty R Short
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia.,Australian Infectious Diseases Research Centre, The University of Queensland, St Lucia, QLD, Australia
| | - Alice E McGovern
- Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
20
|
Yu M, Chang C, Undem BJ, Yu S. Capsaicin-Sensitive Vagal Afferent Nerve-Mediated Interoceptive Signals in the Esophagus. Molecules 2021; 26:3929. [PMID: 34203134 PMCID: PMC8271978 DOI: 10.3390/molecules26133929] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 06/23/2021] [Accepted: 06/24/2021] [Indexed: 01/14/2023] Open
Abstract
Heartburn and non-cardiac chest pain are the predominant symptoms in many esophageal disorders, such as gastroesophageal reflux disease (GERD), non-erosive reflux disease (NERD), functional heartburn and chest pain, and eosinophilic esophagitis (EoE). At present, neuronal mechanisms underlying the process of interoceptive signals in the esophagus are still less clear. Noxious stimuli can activate a subpopulation of primary afferent neurons at their nerve terminals in the esophagus. The evoked action potentials are transmitted through both the spinal and vagal pathways to their central terminals, which synapse with the neurons in the central nervous system to induce esophageal nociception. Over the last few decades, progress has been made in our understanding on the peripheral and central neuronal mechanisms of esophageal nociception. In this review, we focus on the roles of capsaicin-sensitive vagal primary afferent nodose and jugular C-fiber neurons in processing nociceptive signals in the esophagus. We briefly compare their distinctive phenotypic features and functional responses to mechanical and chemical stimulations in the esophagus. Then, we summarize activation and/or sensitization effects of acid, inflammatory cells (eosinophils and mast cells), and mediators (ATP, 5-HT, bradykinin, adenosine, S1P) on these two nociceptive C-fiber subtypes. Lastly, we discuss the potential roles of capsaicin-sensitive esophageal afferent nerves in processing esophageal sensation and nociception. A better knowledge of the mechanism of nociceptive signal processes in primary afferent nerves in the esophagus will help to develop novel treatment approaches to relieve esophageal nociceptive symptoms, especially those that are refractory to proton pump inhibitors.
Collapse
Affiliation(s)
| | | | | | - Shaoyong Yu
- Department of Medicine, Johns Hopkins University School of Medicine, Ross Research Building, 720 Rutland Ave, Baltimore, MD 21205, USA; (M.Y.); (C.C.); (B.J.U.)
| |
Collapse
|
21
|
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: 2.3] [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
|
22
|
Cawthon CR, de La Serre CB. The critical role of CCK in the regulation of food intake and diet-induced obesity. Peptides 2021; 138:170492. [PMID: 33422646 DOI: 10.1016/j.peptides.2020.170492] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 12/16/2020] [Accepted: 12/23/2020] [Indexed: 12/17/2022]
Abstract
In 1973, Gibbs, Young, and Smith showed that exogenous cholecystokinin (CCK) administration reduces food intake in rats. This initial report has led to thousands of studies investigating the physiological role of CCK in regulating feeding behavior. CCK is released from enteroendocrine I cells present along the gastrointestinal (GI) tract. CCK binding to its receptor CCK1R leads to vagal afferent activation providing post-ingestive feedback to the hindbrain. Vagal afferent neurons' (VAN) sensitivity to CCK is modulated by energy status while CCK signaling regulates gene expression of other feeding related signals and receptors expressed by VAN. In addition to its satiation effects, CCK acts all along the GI tract to optimize digestion and nutrient absorption. Diet-induced obesity (DIO) is characterized by reduced sensitivity to CCK and every part of the CCK system is negatively affected by chronic intake of energy-dense foods. EEC have recently been shown to adapt to diet, CCK1R is affected by dietary fats consumption, and the VAN phenotypic flexibility is lost in DIO. Altered endocannabinoid tone, changes in gut microbiota composition, and chronic inflammation are currently being explored as potential mechanisms for diet driven loss in CCK signaling. This review discusses our current understanding of how CCK controls food intake in conditions of leanness and how control is lost in chronic energy excess and obesity, potentially perpetuating excessive intake.
Collapse
Affiliation(s)
- Carolina R Cawthon
- Department of Foods and Nutrition, University of Georgia, Athens, GA, USA
| | | |
Collapse
|
23
|
Ni F, Ogura T, Lin W. Electronic Cigarette Liquid Constituents Induce Nasal and Tracheal Sensory Irritation in Mice in Regionally Dependent Fashion. Nicotine Tob Res 2021; 22:S35-S44. [PMID: 33320249 PMCID: PMC7737480 DOI: 10.1093/ntr/ntaa174] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 10/01/2020] [Indexed: 02/05/2023]
Abstract
INTRODUCTION Electronic cigarettes (e-cigs) are currently used by millions of adults and adolescents worldwide. Major respiratory symptoms, such as coughing reported by e-cig users, including patients with e-cig, or vaping, product use-associated lung injury (EVALI), indicate e-cig constituent-induced sensory irritation. However, e-cig constituent-induced nociceptive activity in nasal and tracheal respiratory epithelia (RE) and neuronal activation in the trigeminal ganglia and brainstem nuclei, which receive airway chemosensory inputs have not been examined and compared. Comparisons of physiological responses between freebase nicotine and nicotine salts are also missing. AIMS AND METHODS Event-related potential (ERP) was recorded electrophysiologically to assess mouse nasal and tracheal RE chemosensory responses to various flavorings, nicotine, including freebase and nicotine salts, e-liquid mixtures, and tussigenic stimuli. Also, mice were subjected to inhalation exposure to aerosol of a vanilla-flavored e-liquid or air (control), and the activated-trigeminal nociceptive neurons and brainstem neurons were examined using immunohistochemistry. RESULTS Individual constituents and mixtures of e-liquids, capsaicin, and citric and acetic acids evoked significantly larger ERP in the nose than in the trachea with the exception of menthol. ERP responses to freebase nicotine were significantly larger than protonated nicotine. Four nicotine salts (benzoate, lactate, levulinate, and salicylate) induced similar responses. Compared with air-exposed mice, e-liquid aerosol-exposed mice showed a significant increase in numbers of activated trigeminal nociceptive neurons and brainstem neurons in the spinal trigeminal nucleus, paratrigeminal nucleus, and nucleus tractus solitarius. CONCLUSIONS E-liquid constituents region-dependently stimulate airway nociceptive chemosensory systems, and freebase nicotine is more potent than protonated nicotine. IMPLICATIONS Neural abnormalities have been implicated in the development of nasal and respiratory illnesses. The higher sensitivity of the nasal nociceptive chemosensory system to nicotine and flavorings may indicate a health risk for e-liquid aerosol-induced upper airway illnesses via neurogenic alteration and warrants further investigation.
Collapse
Affiliation(s)
- Fenge Ni
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, MD
| | - Tatsuya Ogura
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, MD
| | - Weihong Lin
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, MD
| |
Collapse
|
24
|
TLR4 Signaling Selectively and Directly Promotes CGRP Release from Vagal Afferents in the Mouse. eNeuro 2021; 8:ENEURO.0254-20.2020. [PMID: 33318075 PMCID: PMC7877464 DOI: 10.1523/eneuro.0254-20.2020] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 12/03/2020] [Accepted: 12/03/2020] [Indexed: 12/14/2022] Open
Abstract
There has been a long-standing debate regarding the role of peripheral afferents in mediating rapid-onset anorexia among other responses elicited by peripheral inflammatory insults. Thus, the current study assessed the sufficiency of peripheral afferents expressing toll-like receptor 4 (TLR4) to the initiation of the anorexia caused by peripheral bacterial lipopolysaccharide (LPS). We generated a Tlr4 null (Tlr4LoxTB) mouse in which Tlr4 expression is globally disrupted by a loxP-flanked transcription blocking (TB) cassette. This novel mouse model allowed us to restore the endogenous TLR4 expression in specific cell types. Using Zp3-Cre and Nav1.8-Cre mice, we produced mice that express TLR4 in all cells (Tlr4LoxTB X Zp3-Cre) and in peripheral afferents (Tlr4LoxTB X Nav1.8-Cre), respectively. We validated the Tlr4LoxTB mice, which were phenotypically identical to previously reported global TLR4 knock-out mice. Contrary to our expectations, the administration of LPS did not cause rapid-onset anorexia in mice with Nav1.8-restricted TLR4. The later result prompted us to identify Tlr4-expressing vagal afferents using in situ hybridization (ISH). In vivo, we found that Tlr4 mRNA was primarily enriched in vagal Nav1.8 afferents located in the jugular ganglion that co-expressed calcitonin gene-related peptide (CGRP). In vitro, the application of LPS to cultured Nav1.8-restricted TLR4 afferents was sufficient to stimulate the release of CGRP. In summary, we demonstrated using a new mouse model that vagally-expressed TLR4 is selectively involved in stimulating the release of CGRP but not in causing anorexia.
Collapse
|
25
|
Sapio MR, Vazquez FA, Loydpierson AJ, Maric D, Kim JJ, LaPaglia DM, Puhl HL, Lu VB, Ikeda SR, Mannes AJ, Iadarola MJ. Comparative Analysis of Dorsal Root, Nodose and Sympathetic Ganglia for the Development of New Analgesics. Front Neurosci 2021; 14:615362. [PMID: 33424545 PMCID: PMC7793666 DOI: 10.3389/fnins.2020.615362] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 11/25/2020] [Indexed: 12/18/2022] Open
Abstract
Interoceptive and exteroceptive signals, and the corresponding coordinated control of internal organs and sensory functions, including pain, are received and orchestrated by multiple neurons within the peripheral, central and autonomic nervous systems. A central aim of the present report is to obtain a molecularly informed basis for analgesic drug development aimed at peripheral rather than central targets. We compare three key peripheral ganglia: nodose, sympathetic (superior cervical), and dorsal root ganglia in the rat, and focus on their molecular composition using next-gen RNA-Seq, as well as their neuroanatomy using immunocytochemistry and in situ hybridization. We obtained quantitative and anatomical assessments of transmitters, receptors, enzymes and signaling pathways mediating ganglion-specific functions. Distinct ganglionic patterns of expression were observed spanning ion channels, neurotransmitters, neuropeptides, G-protein coupled receptors (GPCRs), transporters, and biosynthetic enzymes. The relationship between ganglionic transcript levels and the corresponding protein was examined using immunohistochemistry for select, highly expressed, ganglion-specific genes. Transcriptomic analyses of spinal dorsal horn and intermediolateral cell column (IML), which form the termination of primary afferent neurons and the origin of preganglionic innervation to the SCG, respectively, disclosed pre- and post-ganglionic molecular-level circuits. These multimodal investigations provide insight into autonomic regulation, nodose transcripts related to pain and satiety, and DRG-spinal cord and IML-SCG communication. Multiple neurobiological and pharmacological contexts can be addressed, such as discriminating drug targets and predicting potential side effects, in analgesic drug development efforts directed at the peripheral nervous system.
Collapse
Affiliation(s)
- Matthew R Sapio
- Anesthesia Section, Department of Perioperative Medicine, National Institutes of Health Clinical Center, Bethesda, MD, United States
| | - Fernando A Vazquez
- Anesthesia Section, Department of Perioperative Medicine, National Institutes of Health Clinical Center, Bethesda, MD, United States
| | - Amelia J Loydpierson
- Anesthesia Section, Department of Perioperative Medicine, National Institutes of Health Clinical Center, Bethesda, MD, United States
| | - Dragan Maric
- Flow and Imaging Cytometry Core Facility, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Jenny J Kim
- Anesthesia Section, Department of Perioperative Medicine, National Institutes of Health Clinical Center, Bethesda, MD, United States
| | - Danielle M LaPaglia
- Anesthesia Section, Department of Perioperative Medicine, National Institutes of Health Clinical Center, Bethesda, MD, United States
| | - Henry L Puhl
- Section on Neurotransmitter Signaling, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, United States
| | - Van B Lu
- Section on Neurotransmitter Signaling, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, United States
| | - Stephen R Ikeda
- Section on Neurotransmitter Signaling, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, United States
| | - Andrew J Mannes
- Anesthesia Section, Department of Perioperative Medicine, National Institutes of Health Clinical Center, Bethesda, MD, United States
| | - Michael J Iadarola
- Anesthesia Section, Department of Perioperative Medicine, National Institutes of Health Clinical Center, Bethesda, MD, United States
| |
Collapse
|
26
|
Falvey A, Duprat F, Simon T, Hugues-Ascery S, Conde SV, Glaichenhaus N, Blancou P. Electrostimulation of the carotid sinus nerve in mice attenuates inflammation via glucocorticoid receptor on myeloid immune cells. J Neuroinflammation 2020; 17:368. [PMID: 33267881 PMCID: PMC7709223 DOI: 10.1186/s12974-020-02016-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 10/29/2020] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND The carotid bodies and baroreceptors are sensors capable of detecting various physiological parameters that signal to the brain via the afferent carotid sinus nerve for physiological adjustment by efferent pathways. Because receptors for inflammatory mediators are expressed by these sensors, we and others have hypothesised they could detect changes in pro-inflammatory cytokine blood levels and eventually trigger an anti-inflammatory reflex. METHODS To test this hypothesis, we surgically isolated the carotid sinus nerve and implanted an electrode, which could deliver an electrical stimulation package prior and following a lipopolysaccharide injection. Subsequently, 90 min later, blood was extracted, and cytokine levels were analysed. RESULTS Here, we found that carotid sinus nerve electrical stimulation inhibited lipopolysaccharide-induced tumour necrosis factor production in both anaesthetised and non-anaesthetised conscious mice. The anti-inflammatory effect of carotid sinus nerve electrical stimulation was so potent that it protected conscious mice from endotoxaemic shock-induced death. In contrast to the mechanisms underlying the well-described vagal anti-inflammatory reflex, this phenomenon does not depend on signalling through the autonomic nervous system. Rather, the inhibition of lipopolysaccharide-induced tumour necrosis factor production by carotid sinus nerve electrical stimulation is abolished by surgical removal of the adrenal glands, by treatment with the glucocorticoid receptor antagonist mifepristone or by genetic inactivation of the glucocorticoid gene in myeloid cells. Further, carotid sinus nerve electrical stimulation increases the spontaneous discharge activity of the hypothalamic paraventricular nucleus leading to enhanced production of corticosterone. CONCLUSION Carotid sinus nerve electrostimulation attenuates inflammation and protects against lipopolysaccharide-induced endotoxaemic shock via increased corticosterone acting on the glucocorticoid receptor of myeloid immune cells. These results provide a rationale for the use of carotid sinus nerve electrostimulation as a therapeutic approach for immune-mediated inflammatory diseases.
Collapse
Affiliation(s)
- Aidan Falvey
- Université Côte d'Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Fabrice Duprat
- Université Côte d'Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Thomas Simon
- Université Côte d'Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | | | - Silvia V Conde
- CEDOC, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Nicolas Glaichenhaus
- Université Côte d'Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Philippe Blancou
- Université Côte d'Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France.
| |
Collapse
|
27
|
Abstract
The gut-brain axis is a coordinated communication system that not only maintains homeostasis, but significantly influences higher cognitive functions and emotions, as well as neurological and behavioral disorders. Among the large populations of sensory and motor neurons that innervate the gut, insights into the function of primary afferent nociceptors, whose cell bodies reside in the dorsal root ganglia and nodose ganglia, have revealed their multiple crosstalk with several cell types within the gut wall, including epithelial, vascular, and immune cells. These bidirectional communications have immunoregulatory functions, control host response to pathogens, and modulate sensations associated with gastrointestinal disorders, through activation of immune cells and glia in the peripheral and central nervous system, respectively. Here, we will review the cellular and neurochemical basis of these interactions at the periphery, in dorsal root ganglia, and in the spinal cord. We will discuss the research gaps that should be addressed to get a better understanding of the multifunctional role of sensory neurons in maintaining gut homeostasis and regulating visceral sensitivity.
Collapse
Affiliation(s)
- Nasser Abdullah
- Department of Physiology and Pharmacology, Inflammation Research Network-Snyder Institute for Chronic Diseases and Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Manon Defaye
- Department of Physiology and Pharmacology, Inflammation Research Network-Snyder Institute for Chronic Diseases and Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Christophe Altier
- Department of Physiology and Pharmacology, Inflammation Research Network-Snyder Institute for Chronic Diseases and Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
28
|
Zalecki M, Juranek J, Pidsudko Z, Mogielnicka-Brzozowska M, Kaleczyc J, Franke-Radowiecka A. Inferior vagal ganglion galaninergic response to gastric ulcers. PLoS One 2020; 15:e0242746. [PMID: 33227035 PMCID: PMC7682887 DOI: 10.1371/journal.pone.0242746] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 11/06/2020] [Indexed: 01/17/2023] Open
Abstract
Galanin is a neuropeptide widely expressed in central and peripheral nerves and is known to be engaged in neuronal responses to pathological changes. Stomach ulcerations are one of the most common gastrointestinal disorders. Impaired stomach function in peptic ulcer disease suggests changes in autonomic nerve reflexes controlled by the inferior vagal ganglion, resulting in stomach dysfunction. In this paper, changes in the galaninergic response of inferior vagal neurons to gastric ulceration in a pig model of the disease were analyzed based on the authors' previous studies. The study was performed on 24 animals (12 control and 12 experimental). Gastric ulcers were induced by submucosal injections of 40% acetic acid solution into stomach submucosa and bilateral inferior vagal ganglia were collected one week afterwards. The number of galanin-immunoreactive perikarya in each ganglion was counted to determine fold-changes between both groups of animals and Q-PCR was applied to verify the changes in relative expression level of mRNA encoding both galanin and its receptor subtypes: GalR1, GalR2, GalR3. The results revealed a 2.72-fold increase in the number of galanin-immunoreactive perikarya compared with the controls. Q-PCR revealed that all studied genes were expressed in examined ganglia in both groups of animals. Statistical analysis revealed a 4.63-fold increase in galanin and a 1.45-fold increase in GalR3 mRNA as compared with the controls. No differences were observed between the groups for GalR1 or GalR2. The current study confirmed changes in the galaninergic inferior vagal ganglion response to stomach ulcerations and demonstrated, for the first time, the expression of mRNA encoding all galanin receptor subtypes in the porcine inferior vagal ganglia.
Collapse
Affiliation(s)
- Michal Zalecki
- Department of Animal Anatomy, Faculty of Veterinary Medicine, University of Warmia and Mazury, Olsztyn, Poland
- * E-mail:
| | - Judyta Juranek
- Department of Human Physiology and Pathophysiology, School of Medicine, University of Warmia and Mazury, Olsztyn, Poland
| | - Zenon Pidsudko
- Department of Animal Anatomy, Faculty of Veterinary Medicine, University of Warmia and Mazury, Olsztyn, Poland
| | - Marzena Mogielnicka-Brzozowska
- Department of Animal Biochemistry and Biotechnology, Faculty of Animal Bioengineering, University of Warmia and Mazury, Olsztyn, Poland
| | - Jerzy Kaleczyc
- Department of Animal Anatomy, Faculty of Veterinary Medicine, University of Warmia and Mazury, Olsztyn, Poland
| | - Amelia Franke-Radowiecka
- Department of Animal Anatomy, Faculty of Veterinary Medicine, University of Warmia and Mazury, Olsztyn, Poland
| |
Collapse
|
29
|
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: 2.3] [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
|
30
|
Cawthon CR, Kirkland RA, Pandya S, Brinson NA, de La Serre CB. Non-neuronal crosstalk promotes an inflammatory response in nodose ganglia cultures after exposure to byproducts from gram positive, high-fat-diet-associated gut bacteria. Physiol Behav 2020; 226:113124. [PMID: 32763334 PMCID: PMC7530053 DOI: 10.1016/j.physbeh.2020.113124] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 07/31/2020] [Accepted: 08/03/2020] [Indexed: 02/06/2023]
Abstract
Vagal afferent neurons (VAN) projecting to the lamina propria of the digestive tract are the primary source of gut-originating signals to the central nervous system (CNS). VAN cell bodies are found in the nodose ganglia (NG). Responsiveness of VAN to gut-originating signals is altered by feeding status with sensitivity to satiety signals such as cholecystokinin (CCK) increasing in the fed state. Chronic high-fat (HF) feeding results in inflammation at the level of the NG associated with a loss of VAN ability to switch phenotype from the fasted to the fed state. HF feeding also leads to compositional changes in the gut microbiota. HF diet consumption notably drives increased Firmicutes to Bacteroidetes phyla ratio and increased members of the Actinobacteria phylum. Firmicutes and Actinobacteria are largely gram positive (GP). In this study, we aimed to determine if byproducts from GP bacteria can induce an inflammatory response in cultured NG and to characterize the mechanism and cell types involved in the response. NG were collected from male Wistar rats and cultured for a total of 72 hours. At 48-68 hours after plating, cultures were treated with neuronal culture media in which Serinicoccus chungangensis had been grown and removed (SUP), lipoteichoic acid (LTA), or meso-diaminopimelic acid (meso-DAP). Some treatments included the glial inhibitors minocycline (MINO) and/or fluorocitrate (FC). The responses were evaluated using immunocytochemistry, qPCR, and electrochemiluminescence. We found that SUP induced an inflammatory response characterized by increased interleukin (IL)-6 staining and increased expression of genes for IL-6, interferon (IFN)γ, and tumor necrosis factor (TNF)α along with genes associated with cell-to-cell communication such as C-C motif chemokine ligand-2 (CCL2). Inclusion of inhibitors attenuated some responses but failed to completely normalize all indications of response, highlighting the role of immunocompetent cellular crosstalk in regulating the inflammatory response. LTA and meso-DAP produced responses that shared characteristics with SUP but were not identical. Our results support a role for HF associated GP bacterial byproducts' ability to contribute to vagal inflammation and to engage signaling from nonneuronal cells.
Collapse
Affiliation(s)
- Carolina R Cawthon
- Department of Foods and Nutrition, The University of Georgia, Athens, Georgia30602, United States
| | - Rebecca A Kirkland
- Department of Foods and Nutrition, The University of Georgia, Athens, Georgia30602, United States
| | - Shreya Pandya
- Department of Foods and Nutrition, The University of Georgia, Athens, Georgia30602, United States
| | - Nigel A Brinson
- Department of Foods and Nutrition, The University of Georgia, Athens, Georgia30602, United States
| | - Claire B de La Serre
- Department of Foods and Nutrition, The University of Georgia, Athens, Georgia30602, United States.
| |
Collapse
|
31
|
Vermeiren S, Bellefroid EJ, Desiderio S. Vertebrate Sensory Ganglia: Common and Divergent Features of the Transcriptional Programs Generating Their Functional Specialization. Front Cell Dev Biol 2020; 8:587699. [PMID: 33195244 PMCID: PMC7649826 DOI: 10.3389/fcell.2020.587699] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 09/08/2020] [Indexed: 12/13/2022] Open
Abstract
Sensory fibers of the peripheral nervous system carry sensation from specific sense structures or use different tissues and organs as receptive fields, and convey this information to the central nervous system. In the head of vertebrates, each cranial sensory ganglia and associated nerves perform specific functions. Sensory ganglia are composed of different types of specialized neurons in which two broad categories can be distinguished, somatosensory neurons relaying all sensations that are felt and visceral sensory neurons sensing the internal milieu and controlling body homeostasis. While in the trunk somatosensory neurons composing the dorsal root ganglia are derived exclusively from neural crest cells, somato- and visceral sensory neurons of cranial sensory ganglia have a dual origin, with contributions from both neural crest and placodes. As most studies on sensory neurogenesis have focused on dorsal root ganglia, our understanding of the molecular mechanisms underlying the embryonic development of the different cranial sensory ganglia remains today rudimentary. However, using single-cell RNA sequencing, recent studies have made significant advances in the characterization of the neuronal diversity of most sensory ganglia. Here we summarize the general anatomy, function and neuronal diversity of cranial sensory ganglia. We then provide an overview of our current knowledge of the transcriptional networks controlling neurogenesis and neuronal diversification in the developing sensory system, focusing on cranial sensory ganglia, highlighting specific aspects of their development and comparing it to that of trunk sensory ganglia.
Collapse
Affiliation(s)
- Simon Vermeiren
- ULB Neuroscience Institute, Université Libre de Bruxelles, Gosselies, Belgium
| | - Eric J Bellefroid
- ULB Neuroscience Institute, Université Libre de Bruxelles, Gosselies, Belgium
| | - Simon Desiderio
- Institute for Neurosciences of Montpellier, INSERM U1051, University of Montpellier, Montpellier, France
| |
Collapse
|
32
|
Patil MJ, Ru F, Sun H, Wang J, Kolbeck RR, Dong X, Kollarik M, Canning BJ, Undem BJ. Acute activation of bronchopulmonary vagal nociceptors by type I interferons. J Physiol 2020; 598:5541-5554. [PMID: 32924209 DOI: 10.1113/jp280276] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 08/17/2020] [Indexed: 12/14/2022] Open
Abstract
KEY POINTS Type I interferon receptors are expressed by the majority of vagal C-fibre neurons innervating the respiratory tract Interferon alpha and beta acutely and directly activate vagal C-fibers in the airways. The interferon-induced activation of C-fibers occurs secondary to stimulation of type 1 interferon receptors Type 1 interferons may contribute to the symptoms as well as the spread of respiratory viral infections by causing coughing and other defensive reflexes associated with vagal C-fibre activation ABSTRACT: We evaluated the ability of type I interferons to acutely activate airway vagal afferent nerve terminals in mouse lungs. Using single cell RT-PCR of lung-specific vagal neurons we found that IFNAR1 and IFNAR2 were expressed in 70% of the TRPV1-positive neurons (a marker for vagal C-fibre neurons) and 44% of TRPV1-negative neurons. We employed an ex vivo vagal innervated mouse trachea-lung preparation to evaluate the effect of interferons in directly activating airway nerves. Utilizing 2-photon microscopy of the nodose ganglion neurons from Pirt-Cre;R26-GCaMP6s mice we found that applying IFNα or IFNβ to the lungs acutely activated the majority of vagal afferent nerve terminals. When the type 1 interferon receptor, IFNAR1, was blocked with a blocking antibody the response to IFNβ was largely inhibited. The type 2 interferon, IFNγ, also activated airway nerves and this was not inhibited by the IFNAR1 blocking antibody. The Janus kinase inhibitor GLPG0634 (1 μm) virtually abolished the nerve activation caused by IFNβ. Consistent with the activation of vagal afferent C-fibers, infusing IFNβ into the mouse trachea led to defensive breathing reflexes including apneas and gasping. These reflexes were prevented by pretreatment with an IFN type-1 receptor blocking antibody. Finally, using whole cell patch-clamp electrophysiology of lung-specific neurons we found that IFNβ (1000 U ml-1 ) directly depolarized the membrane potential of isolated nodose neurons, in some cases beyond to action potential threshold. This acute non-genomic activation of vagal sensory nerve terminals by interferons may contribute to the incessant coughing that is a hallmark of respiratory viral infections.
Collapse
Affiliation(s)
- Mayur J Patil
- The Johns Hopkins School of Medicine Departments of Medicine, Baltimore, MD
| | - Fei Ru
- The Johns Hopkins School of Medicine Departments of Medicine, Baltimore, MD
| | - Hui Sun
- The Johns Hopkins School of Medicine Departments of Medicine, Baltimore, MD
| | - Jingya Wang
- AstraZeneca BioPharmaceuticals R&D Gaithersburg, MD
| | | | - Xinzhong Dong
- The Johns Hopkins School of Medicine Departments of Neuroscience, Baltimore, MD
| | - Marian Kollarik
- The Johns Hopkins School of Medicine Departments of Medicine, Baltimore, MD
| | - Brendan J Canning
- The Johns Hopkins School of Medicine Departments of Medicine, Baltimore, MD
| | - Bradley J Undem
- The Johns Hopkins School of Medicine Departments of Medicine, Baltimore, MD
| |
Collapse
|
33
|
Meerschaert KA, Adelman PC, Friedman RL, Albers KM, Koerber HR, Davis BM. Unique Molecular Characteristics of Visceral Afferents Arising from Different Levels of the Neuraxis: Location of Afferent Somata Predicts Function and Stimulus Detection Modalities. J Neurosci 2020; 40:7216-7228. [PMID: 32817244 PMCID: PMC7534907 DOI: 10.1523/jneurosci.1426-20.2020] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 07/30/2020] [Accepted: 08/07/2020] [Indexed: 02/07/2023] Open
Abstract
Viscera receive innervation from sensory ganglia located adjacent to multiple levels of the brainstem and spinal cord. Here we examined whether molecular profiling could be used to identify functional clusters of colon afferents from thoracolumbar (TL), lumbosacral (LS), and nodose ganglia (NG) in male and female mice. Profiling of TL and LS bladder afferents was also performed. Visceral afferents were back-labeled using retrograde tracers injected into proximal and distal regions of colon or bladder, followed by single-cell qRT-PCR and analysis via an automated hierarchical clustering method. Genes were chosen for assay (32 for bladder; 48 for colon) based on their established role in stimulus detection, regulation of sensitivity/function, or neuroimmune interaction. A total of 132 colon afferents (from NG, TL, and LS ganglia) and 128 bladder afferents (from TL and LS ganglia) were analyzed. Retrograde labeling from the colon showed that NG and TL afferents innervate proximal and distal regions of the colon, whereas 98% of LS afferents only project to distal regions. There were clusters of colon and bladder afferents, defined by mRNA profiling, that localized to either TL or LS ganglia. Mixed TL/LS clustering also was found. In addition, transcriptionally, NG colon afferents were almost completely segregated from colon TL and LS neurons. Furthermore, colon and bladder afferents expressed genes at similar levels, although different gene combinations defined the clusters. These results indicate that genes implicated in both homeostatic regulation and conscious sensations are found at all anatomic levels, suggesting that afferents from different portions of the neuraxis have overlapping functions.SIGNIFICANCE STATEMENT Visceral organs are innervated by sensory neurons whose cell bodies are located in multiple ganglia associated with the brainstem and spinal cord. For the colon, this overlapping innervation is proposed to facilitate visceral sensation and homeostasis, where sensation and pain are mediated by spinal afferents and fear and anxiety (the affective aspects of visceral pain) are the domain of nodose afferents. The transcriptomic analysis performed here reveals that genes implicated in both homeostatic regulation and pain are found in afferents across all ganglia types, suggesting that conscious sensation and homeostatic regulation are the result of convergence, and not segregation, of sensory input.
Collapse
Affiliation(s)
- Kimberly A Meerschaert
- Department of Neurobiology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
- Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| | | | - Robert L Friedman
- Department of Neurobiology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
- Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| | - Kathryn M Albers
- Department of Neurobiology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
- Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| | - H Richard Koerber
- Department of Neurobiology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
- Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| | - Brian M Davis
- Department of Neurobiology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
- Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| |
Collapse
|
34
|
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: 3.3] [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
|
35
|
P2X3-Receptor Antagonists as Potential Antitussives: Summary of Current Clinical Trials in Chronic Cough. Lung 2020; 198:609-616. [PMID: 32661659 DOI: 10.1007/s00408-020-00377-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 07/01/2020] [Indexed: 01/17/2023]
Abstract
Cough is among the most common complaints for which patients worldwide seek medical attention. In a majority of patients with chronic cough (defined as cough of greater than 8 weeks' duration), successful management results from a thorough evaluation and treatment of underlying causes. In a subgroup of patients, however, cough proves refractory to therapeutic trials aimed at known reversible causes of chronic cough. Such patients are appropriately termed as having refractory chronic cough. At present, safe and effective medications are lacking for this challenging patient population. Currently available therapeutic options are usually ineffective or achieve antitussive effect at the expense of intolerable side effects, typically sedation. Fortunately, the past decade has witnessed great progress in elucidating underlying mechanisms of cough. From that knowledge, aided by the development of validated instruments to measure objective and subjective cough-related end points, numerous antitussive drug development programs have emerged. The most active area of inquiry at present involves antagonists of the purinergic P2X receptors. Indeed, four clinical programs (one in Phase 3 and three in Phase 2) are currently underway investigating antagonists of receptors comprised entirely or partially of the P2X3 subunit as potential antitussive medications. Herein we review the foundation on which P2X receptor antagonists were developed as potential antitussive medications and provide an update on current clinical trials.
Collapse
|
36
|
Kupari J, Häring M, Agirre E, Castelo-Branco G, Ernfors P. An Atlas of Vagal Sensory Neurons and Their Molecular Specialization. Cell Rep 2020; 27:2508-2523.e4. [PMID: 31116992 PMCID: PMC6533201 DOI: 10.1016/j.celrep.2019.04.096] [Citation(s) in RCA: 232] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 03/25/2019] [Accepted: 04/22/2019] [Indexed: 12/31/2022] Open
Abstract
Sensory functions of the vagus nerve are critical for conscious perceptions and for monitoring visceral functions in the cardio-pulmonary and gastrointestinal systems. Here, we present a comprehensive identification, classification, and validation of the neuron types in the neural crest (jugular) and placode (nodose) derived vagal ganglia by single-cell RNA sequencing (scRNA-seq) transcriptomic analysis. Our results reveal major differences between neurons derived from different embryonic origins. Jugular neurons exhibit fundamental similarities to the somatosensory spinal neurons, including major types, such as C-low threshold mechanoreceptors (C-LTMRs), A-LTMRs, Aδ-nociceptors, and cold-, and mechano-heat C-nociceptors. In contrast, the nodose ganglion contains 18 distinct types dedicated to surveying the physiological state of the internal body. Our results reveal a vast diversity of vagal neuron types, including many previously unanticipated types, as well as proposed types that are consistent with chemoreceptors, nutrient detectors, baroreceptors, and stretch and volume mechanoreceptors of the respiratory, gastrointestinal, and cardiovascular systems. A comprehensive molecular identification of neuronal types in vagal ganglion complex Prdm12+ jugular ganglion neurons share features with spinal somatosensory neurons Phox2b+ viscerosensory nodose neurons are molecularly versatile and highly specialized Nodose neuron types are consistent with chemo-, baro-, stretch-, tension-, and volume-sensors
Collapse
Affiliation(s)
- Jussi Kupari
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Martin Häring
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Eneritz Agirre
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Gonçalo Castelo-Branco
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden; Ming Wai Lau Centre for Reparative Medicine, Stockholm node, Karolinska Institutet, Stockholm, Sweden
| | - Patrik Ernfors
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden.
| |
Collapse
|
37
|
Mayer F, Gunawan AL, Tso P, Aponte GW. Glucagon-like peptide 1 and glucose-dependent insulinotropic polypeptide stimulate release of substance P from TRPV1- and TRPA1-expressing sensory nerves. Am J Physiol Gastrointest Liver Physiol 2020; 319:G23-G35. [PMID: 32421358 PMCID: PMC7468754 DOI: 10.1152/ajpgi.00189.2019] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) are released from enteroendocrine cells (EECs) in response to nutrient ingestion and lower blood glucose levels by stimulation of insulin secretion and thus are defined as incretins. GLP-1 receptor (GLP-1R) expression has been identified on enteric neurons that include intrinsic afferent neurons, extrinsic spinal, and vagal sensory afferents but has not been shown to have an incretin effect through these nerves. GLP-1 and GIP enter the mesenteric lymphatic fluid (MLF) after a meal via the interstitial fluid (IF) from local tissue secretion and/or blood capillaries. We tested if MLF could induce diet-dependent intransient increases in intracellular calcium ([Ca2+]i) in cultured sensory neurons. Postprandial rat MLF, collected from the superior mesenteric lymphatic duct, induced a significant twofold higher intransient increase in [Ca2+]i in primary-cultured sensory neurons than MLF from fasted rats. Inhibition of transient receptor potential vanilloid 1 (TRPV1) and TRPV1 and ankyrin 1 cation channels (TRPA1) with ruthenium red eliminated the difference. Substance P (SP) (a peptide that stimulates insulin secretion) sensor cells cocultured with sensory neurons showed both the GLP-1R agonist exendin-4 (Ex-4) and GIP induced transient increases in [Ca2+]i directly coupled to SP secretion in the sensory nerves. Ex-4-induced release of SP required expression of either TRPA1 or TRPV1. These data identify unrecognized actions of GLP-1 and GIP as incretins by acting as neurolymphocrines and suggest a mechanism for sensory nerves to respond to the postprandial state through MLF.NEW & NOTEWORTHY Glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) are secreted upon eating to lower blood sugar. GLP-1 and GIP were found to induce the secretion of substance P (SP) from cultured sensory nerves. SP enhances insulin secretion. Mesenteric lymphatic fluid (MLF) also stimulates sensory neurons in a diet-dependent manner. These studies identify new actions of GLP-1 and GIP as incretins and suggest a mechanism for sensory nerves to respond to diet through MLF.
Collapse
Affiliation(s)
- Fahima Mayer
- 1Department of Nutritional Sciences and Toxicology, Graduate Program in Metabolic Biology, University of California, Berkeley, California
| | - Amanda L. Gunawan
- 1Department of Nutritional Sciences and Toxicology, Graduate Program in Metabolic Biology, University of California, Berkeley, California
| | - Patrick Tso
- 2Department of Pathobiology and Molecular Medicine, University of Cincinnati, Reading, Ohio
| | - Gregory W. Aponte
- 1Department of Nutritional Sciences and Toxicology, Graduate Program in Metabolic Biology, University of California, Berkeley, California
| |
Collapse
|
38
|
Bahia PK, Hadley SH, Barannikov I, Sowells I, Kim SH, Taylor-Clark TE. Antimycin A increases bronchopulmonary C-fiber excitability via protein kinase C alpha. Respir Physiol Neurobiol 2020; 278:103446. [PMID: 32360368 DOI: 10.1016/j.resp.2020.103446] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 04/15/2020] [Accepted: 04/18/2020] [Indexed: 12/17/2022]
Abstract
Inflammation can increase the excitability of bronchopulmonary C-fibers leading to excessive sensations and reflexes (e.g. wheeze and cough). We have previously shown modulation of peripheral nerve terminal mitochondria by antimycin A causes hyperexcitability in TRPV1-expressing bronchopulmonary C-fibers through the activation of protein kinase C (PKC). Here, we have investigated the PKC isoform responsible for this signaling. We found PKCβ1, PKCδ and PKCε were expressed by many vagal neurons, with PKCα and PKCβ2 expressed by subsets of vagal neurons. In dissociated vagal neurons, antimycin A caused translocation of PKCα but not the other isoforms, and only in TRPV1-lineage neurons. In bronchopulmonary C-fiber recordings, antimycin A increased the number of action potentials evoked by α,β-methylene ATP. Selective inhibition of PKCα, PKCβ1 and PKCβ2 with 50 nM bisindolylmaleimide I prevented the antimycin-induced bronchopulmonary C-fiber hyperexcitability, whereas selective inhibition of only PKCβ1 and PKCβ2 with 50 nM LY333531 had no effect. We therefore conclude that PKCα is required for antimycin-induced increases in bronchopulmonary C-fiber excitability.
Collapse
Affiliation(s)
- Parmvir K Bahia
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Stephen H Hadley
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Ivan Barannikov
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Isobel Sowells
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Seol-Hee Kim
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Thomas E Taylor-Clark
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA.
| |
Collapse
|
39
|
Koskela HO, Nurmi HM, Purokivi MK. Cough-provocation tests with hypertonic aerosols. ERJ Open Res 2020; 6:00338-2019. [PMID: 32337214 PMCID: PMC7167210 DOI: 10.1183/23120541.00338-2019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 01/23/2020] [Indexed: 12/19/2022] Open
Abstract
Recent advances in cough research suggest a more widespread use of cough-provocation tests to demonstrate the hypersensitivity of the cough reflex arc. Cough-provocation tests with capsaicin or acidic aerosols have been used for decades in scientific studies. Several factors have hindered their use in everyday clinical work: i.e. lack of standardisation, the need for special equipment and the limited clinical importance of the response. Cough-provocation tests with hypertonic aerosols (CPTHAs) involve provocations with hypertonic saline, hypertonic histamine, mannitol and hyperpnoea. They probably act via different mechanisms than capsaicin and acidic aerosols. They are safe and well tolerated and the response is repeatable. CPTHAs can assess not only the sensitivity of the cough reflex arc but also the tendency of the airway smooth muscles to constrict (airway hyper-responsiveness). They can differentiate between subjects with asthma or chronic cough and healthy subjects. The responsiveness to CPTHAs correlates with the cough-related quality of life among asthmatic subjects. Furthermore, the responsiveness to them decreases during treatment of chronic cough. A severe response to CPTHAs may indicate poor long-term prognosis in chronic cough. The mannitol test has been stringently standardised, is easy to administer with simple equipment, and has regulatory approval for the assessment of airway hyper-responsiveness. Manual counting of coughs during a mannitol challenge would allow the measurement of the function of the cough reflex arc as a part of clinical routine. Cough-provocation tests with hypertonic aerosols offer the possibility to measure the function of the cough reflex arc even in everyday clinical workhttp://bit.ly/2RTOfMI
Collapse
Affiliation(s)
- Heikki O Koskela
- Unit for Medicine and Clinical Research, Pulmonary Division, Kuopio University Hospital, Kuopio, Finland.,School of Medicine, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Hanna M Nurmi
- Unit for Medicine and Clinical Research, Pulmonary Division, Kuopio University Hospital, Kuopio, Finland.,School of Medicine, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Minna K Purokivi
- Unit for Medicine and Clinical Research, Pulmonary Division, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
40
|
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: 42] [Impact Index Per Article: 10.5] [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
|
41
|
Kaelberer MM, Caceres AI, Jordt SE. Activation of a nerve injury transcriptional signature in airway-innervating sensory neurons after lipopolysaccharide-induced lung inflammation. Am J Physiol Lung Cell Mol Physiol 2020; 318:L953-L964. [PMID: 32159971 DOI: 10.1152/ajplung.00403.2019] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The lungs and the immune and nervous systems functionally interact to respond to respiratory environmental exposures and infections. The lungs are innervated by vagal sensory neurons of the jugular and nodose ganglia, fused together in smaller mammals as the jugular-nodose complex (JNC). Whereas the JNC shares properties with the other sensory ganglia, the trigeminal (TG) and dorsal root ganglia (DRG), these sensory structures express differential sets of genes that reflect their unique functionalities. Here, we used RNA sequencing (RNA-seq) in mice to identify the differential transcriptomes of the three sensory ganglia types. Using a fluorescent retrograde tracer and fluorescence-activated cell sorting, we isolated a defined population of airway-innervating JNC neurons and determined their differential transcriptional map after pulmonary exposure to lipopolysaccharide (LPS), a major mediator of acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) after infection with gram-negative bacteria or inhalation of organic dust. JNC neurons activated an injury response program, leading to increased expression of gene products such as the G protein-coupled receptor Cckbr, inducing functional changes in neuronal sensitivity to peptides, and Gpr151, also rapidly induced upon neuropathic nerve injury in pain models. Unique JNC-specific transcripts, present at only minimal levels in TG, DRG, and other organs, were identified. These included TMC3, encoding for a putative mechanosensor, and urotensin 2B, a hypertensive peptide. These findings highlight the unique properties of the JNC and reveal that ALI/ARDS rapidly induces a nerve injury-related state, changing vagal excitability.
Collapse
Affiliation(s)
| | - Ana Isabel Caceres
- Department of Anesthesiology, Duke University School of Medicine, Durham, North Carolina
| | - Sven-Eric Jordt
- Department of Anesthesiology, Duke University School of Medicine, Durham, North Carolina.,Department of Pharmacology and Cancer Biology, Duke University School of Medicine. Durham, North Carolina.,Integrated Toxicology and Environmental Health Program (ITEHP), Duke University, Durham, North Carolina
| |
Collapse
|
42
|
Ualiyeva S, Hallen N, Kanaoka Y, Ledderose C, Matsumoto I, Junger W, Barrett N, Bankova L. Airway brush cells generate cysteinyl leukotrienes through the ATP sensor P2Y2. Sci Immunol 2020; 5:5/43/eaax7224. [PMID: 31953256 PMCID: PMC7176051 DOI: 10.1126/sciimmunol.aax7224] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Accepted: 12/19/2019] [Indexed: 01/12/2023]
Abstract
Chemosensory epithelial cells (EpCs) are specialized cells that promote innate type 2 immunity and protective neurally mediated reflexes in the airway. Their effector programs and modes of activation are not fully understood. Here, we define the transcriptional signature of two choline acetyltransferase-expressing nasal EpC populations. They are found in the respiratory and olfactory mucosa and express key chemosensory cell genes including the transcription factor Pou2f3, the cation channel Trpm5, and the cytokine Il25 Moreover, these cells share a core transcriptional signature with chemosensory cells from intestine, trachea and thymus, and cluster with tracheal brush cells (BrCs) independently from other respiratory EpCs, indicating that they are part of the brush/tuft cell family. Both nasal BrC subsets express high levels of transcripts encoding cysteinyl leukotriene (CysLT) biosynthetic enzymes. In response to ionophore, unfractionated nasal BrCs generate CysLTs at levels exceeding that of the adjacent hematopoietic cells isolated from naïve mucosa. Among activating receptors, BrCs express the purinergic receptor P2Y2. Accordingly, the epithelial stress signal ATP and aeroallergens that elicit ATP release trigger BrC CysLT generation, which is mediated by the P2Y2 receptor. ATP- and aeroallergen-elicited CysLT generation in the nasal lavage is reduced in mice lacking Pou2f3, a requisite transcription factor for BrC development. Last, aeroallergen-induced airway eosinophilia is reduced in BrC-deficient mice. These results identify a previously undescribed BrC sensor and effector pathway leading to generation of lipid mediators in response to luminal signals. Further, they suggest that BrC sensing of local damage may provide an important sentinel immune function.
Collapse
Affiliation(s)
- S. Ualiyeva
- Division of Allergy and Clinical Immunology, Jeff and Penny Vinik Center for Allergic Disease Research, Brigham and Women’s Hospital and Department of Medicine, Harvard Medical School, Boston, MA
| | - N. Hallen
- Division of Allergy and Clinical Immunology, Jeff and Penny Vinik Center for Allergic Disease Research, Brigham and Women’s Hospital and Department of Medicine, Harvard Medical School, Boston, MA
| | - Y. Kanaoka
- Division of Allergy and Clinical Immunology, Jeff and Penny Vinik Center for Allergic Disease Research, Brigham and Women’s Hospital and Department of Medicine, Harvard Medical School, Boston, MA
| | - C. Ledderose
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | | | - W. Junger
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - N.A. Barrett
- Division of Allergy and Clinical Immunology, Jeff and Penny Vinik Center for Allergic Disease Research, Brigham and Women’s Hospital and Department of Medicine, Harvard Medical School, Boston, MA
| | - L.G. Bankova
- Division of Allergy and Clinical Immunology, Jeff and Penny Vinik Center for Allergic Disease Research, Brigham and Women’s Hospital and Department of Medicine, Harvard Medical School, Boston, MA
| |
Collapse
|
43
|
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: 18] [Impact Index Per Article: 4.5] [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
|
44
|
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: 3.2] [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
|
45
|
Abstract
The peripheral nervous system (PNS) is highly complicated and heterogenous. Conventional neuromodulatory approaches have revealed numerous essential biological functions of the PNS and provided excellent tools to treat a large variety of human diseases. Yet growing evidence indicated the importance of cell-type-specific neuromodulation in the PNS in not only biological research using animal models but also potential human therapies. Optogenetics is a recently developed neuromodulatory approach combining optics and genetics that can effectively stimulate or silence neuronal activity with high spatial and temporal precision. Here, I review research regarding optogenetic manipulations for cell-type-specific control of the PNS, highlighting the advantages and challenges of current optogenetic tools, and discuss their potential future applications.
Collapse
Affiliation(s)
- Rui B Chang
- Department of Neuroscience, Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut 06520
| |
Collapse
|
46
|
Sun H, Meeker S, Undem BJ. Role of TRP channels in G q-coupled protease-activated receptor 1-mediated activation of mouse nodose pulmonary C-fibers. Am J Physiol Lung Cell Mol Physiol 2019; 318:L192-L199. [PMID: 31664854 DOI: 10.1152/ajplung.00301.2019] [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] [Indexed: 01/06/2023] Open
Abstract
We evaluated the mechanisms underlying protease-activated receptor 1 (PAR1)-mediated activation of nodose C-fibers in mouse lungs. The PAR1-induced action potential discharge at the terminals was strongly inhibited in phospholipase C-β3 (PLCβ3)-deficient animals. At the level of the cell soma, PAR1 activation led to an increase in cytosolic calcium that was largely inhibited by transient receptor potential (TRP) A1 antagonism. Patch-clamp recordings, however, revealed that neither TRPA1 nor TRPV1 or any other ruthenium red-sensitive ion channels are required for the PAR1-mediated inward current or membrane depolarization in isolated nodose neurons. Consistent with these findings, PAR1-mediated action potential discharge in mouse lung nodose C-fiber terminals was unaltered in Trpa1/Trpv1 double-knockout animals and Trpc3/Trpc6 double-knockout animals. The activation of the C-fibers was also not inhibited by ruthenium red at concentrations that blocked TRPV1- and TRPA1-dependent responses. The biophysical data show that PAR1/Gq-mediated activation of nodose C-fibers may involve multiple ion channels downstream from PLCβ3 activation. TRPA1 is an ion channel that participates in PAR1/Gq-mediated elevation in intracellular calcium. There is little evidence, however, that TRPA1, TRPV1, TRPC3, TRPC6, or other ruthenium red-sensitive TRP channels are required for PAR1/Gq-PLCβ3-mediated membrane depolarization and action potential discharge in bronchopulmonary nodose C-fibers in the mouse.
Collapse
Affiliation(s)
- Hui Sun
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Sonya Meeker
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Bradley J Undem
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
47
|
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: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 09/12/2019] [Indexed: 12/11/2022]
|
48
|
Mac Aogáin M, Chotirmall SH. Bronchiectasis and cough: An old relationship in need of renewed attention. Pulm Pharmacol Ther 2019; 57:101812. [PMID: 31176801 PMCID: PMC7110869 DOI: 10.1016/j.pupt.2019.101812] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 05/31/2019] [Accepted: 06/06/2019] [Indexed: 12/18/2022]
Abstract
Bronchiectasis is an increasingly recognised respiratory condition with limited therapeutic options and a complex spectrum of clinical manifestations that invariably includes chronic cough. As the primary presentation of bronchiectasis in most cases, chronic cough and its mechanistic underpinnings are of central importance but remain poorly understood in this setting. Bronchiectasis is also increasingly identified as an underlying cause of chronic cough highlighting the interrelationship between the two conditions that share overlapping clinical features. Several therapeutic approaches have illustrated positive effects on bronchiectasis-associated cough, however, more focused treatment of heterogeneous cough subtypes may yield better outcomes for patients. A current challenge is the identification of bronchiectasis and cough endophenotypes that may allow improved patient stratification and more targeted therapeutic matching of the right treatment to the right patient. Here we discuss the complex disease phenotypes of bronchiectasis and their interrelationship with cough while considering current and emerging treatment options. We discuss some key cough promoters in bronchiectasis including infection, allergy and immune dysfunction.
Collapse
Affiliation(s)
- Micheál Mac Aogáin
- Translational Respiratory Research Laboratory, Lee Kong Chian School of Medicine, Nanyang Technological University, Level 12, Clinical Sciences Building, 11 Mandalay Road, Singapore, 308232, Singapore
| | - Sanjay Haresh Chotirmall
- Translational Respiratory Research Laboratory, Lee Kong Chian School of Medicine, Nanyang Technological University, Level 12, Clinical Sciences Building, 11 Mandalay Road, Singapore, 308232, Singapore.
| |
Collapse
|
49
|
Kollarik M, Sun H, Herbstsomer RA, Ru F, Kocmalova M, Meeker SN, Undem BJ. Different role of TTX-sensitive voltage-gated sodium channel (Na V 1) subtypes in action potential initiation and conduction in vagal airway nociceptors. J Physiol 2019; 596:1419-1432. [PMID: 29435993 DOI: 10.1113/jp275698] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 01/23/2018] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS The action potential initiation in the nerve terminals and its subsequent conduction along the axons of afferent nerves are not necessarily dependent on the same voltage-gated sodium channel (NaV 1) subunits. The action potential initiation in jugular C-fibres within airway tissues is not blocked by TTX; nonetheless, conduction of action potentials along the vagal axons of these nerves is often dependent on TTX-sensitive channels. This is not the case for nodose airway Aδ-fibres and C-fibres, where both action potential initiation and conduction is abolished by TTX or selective NaV 1.7 blockers. The difference between the initiation of action potentials within the airways vs. conduction along the axons should be considered when developing NaV 1 blocking drugs for topical application to the respiratory tract. ABSTRACT The action potential (AP) initiation in the nerve terminals and its subsequent AP conduction along the axons do not necessarily depend on the same subtypes of voltage-gated sodium channels (NaV 1s). We evaluated the role of TTX-sensitive and TTX-resistant NaV 1s in vagal afferent nociceptor nerves derived from jugular and nodose ganglia innervating the respiratory system. Single cell RT-PCR was performed on vagal afferent neurons retrogradely labelled from the guinea pig trachea. Almost all of the jugular neurons expressed the TTX-sensitive channel NaV 1.7 along with TTX-resistant NaV 1.8 and NaV 1.9. Tracheal nodose neurons also expressed NaV 1.7 but, less frequently, NaV 1.8 and NaV 1.9. NaV 1.6 were expressed in ∼40% of the jugular and 25% of nodose tracheal neurons. Other NaV 1 α subunits were only rarely expressed. Single fibre recordings were made from the vagal nodose and jugular nerve fibres innervating the trachea or lung in the isolated perfused vagally-innervated preparations that allowed for selective drug delivery to the nerve terminal compartment (AP initiation) or to the desheathed vagus nerve (AP conduction). AP initiation in jugular C-fibres was unaffected by TTX, although it was inhibited by NaV 1.8 blocker (PF-01247324) and abolished by combination of TTX and PF-01247324. However, AP conduction in the majority of jugular C-fibres was abolished by TTX. By contrast, both AP initiation and conduction in nodose nociceptors was abolished by TTX or selective NaV 1.7 blockers. Distinction between the effect of a drug with respect to inhibiting AP in the nerve terminals within the airways vs. at conduction sites along the vagus nerve is relevant to therapeutic strategies involving inhaled NaV 1 blocking drugs.
Collapse
Affiliation(s)
- M Kollarik
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Pathophysiology, Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovakia
| | - H Sun
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - R A Herbstsomer
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - F Ru
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - M Kocmalova
- Department of Pharmacology, Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovakia
| | - S N Meeker
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - B J Undem
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
50
|
Tao M, Liu Q, Miyazaki Y, Canning BJ. Nicotinic receptor dependent regulation of cough and other airway defensive reflexes. Pulm Pharmacol Ther 2019; 58:101810. [PMID: 31181318 DOI: 10.1016/j.pupt.2019.101810] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 06/05/2019] [Accepted: 06/06/2019] [Indexed: 11/30/2022]
Abstract
Nicotinic receptor activation in the airways evokes airway defensive reflexes including cough. These reflexes are the direct result of bronchopulmonary afferent nerve activation, which may occur directly, through activation of nicotinic receptors expressed on the terminals of airway sensory nerves, or indirectly, secondary to the end organ effects associated with autonomic nerve stimulation. The irritating effects of nicotine delivered topically to the airways are counterbalanced by an inhibitory effect of nicotinic receptor activation in the central nervous system. We present evidence that these nicotinic receptors are components of essential transducing and encoding mechanisms regulating airway defense.
Collapse
Affiliation(s)
- Mayuko Tao
- Tokyo Medical & Dental University, Japan
| | - Qi Liu
- Johns Hopkins Asthma and Allergy Center, Baltimore, MD, USA
| | | | | |
Collapse
|