1
|
Pagiazitis JG, Delestrée N, Sowoidnich L, Sivakumar N, Simon CM, Chatzisotiriou A, Albani M, Mentis GZ. Catecholaminergic dysfunction drives postural and locomotor deficits in a mouse model of spinal muscular atrophy. Cell Rep 2025; 44:115147. [PMID: 39752251 PMCID: PMC11832083 DOI: 10.1016/j.celrep.2024.115147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 10/24/2024] [Accepted: 12/12/2024] [Indexed: 02/01/2025] Open
Abstract
Development and maintenance of posture is essential behavior for overground mammalian locomotion. Dopamine and noradrenaline strongly influence locomotion, and their dysregulation initiates the development of motor impairments linked to neurodegenerative disease. However, the precise cellular and circuit mechanisms are not well defined. Here, we investigated the role of catecholaminergic neuromodulation in a mouse model of spinal muscular atrophy (SMA). SMA is characterized by severe motor dysfunction and postural deficits. We identify progressive loss of catecholaminergic synapses from spinal neurons that occur via non-cell autonomous mechanisms. Importantly, the selective restoration of survival motor neuron (SMN) in either catecholaminergic or serotonergic neurons is sufficient to correct impairments in locomotion. However, only combined SMN restoration in both catecholaminergic and serotonergic neurons or pharmacological treatment with l-dopa improve the severe postural deficits. These findings uncover the synaptic and cellular mechanisms responsible for the postural and motor symptoms in SMA and identify catecholaminergic neuromodulation as a potential therapeutic target.
Collapse
Affiliation(s)
- John G Pagiazitis
- Center for Motor Neuron Biology and Disease, Columbia University, New York, NY 10032, USA; Department of Neurology, Columbia University, New York, NY 10032, USA; Department of Physiology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki 541 24, Greece
| | - Nicolas Delestrée
- Center for Motor Neuron Biology and Disease, Columbia University, New York, NY 10032, USA; Department of Neurology, Columbia University, New York, NY 10032, USA
| | - Leonie Sowoidnich
- Center for Motor Neuron Biology and Disease, Columbia University, New York, NY 10032, USA; Department of Neurology, Columbia University, New York, NY 10032, USA; Carl-Ludwig-Institute for Physiology, Leipzig University, Leipzig, Germany
| | - Nandhini Sivakumar
- Center for Motor Neuron Biology and Disease, Columbia University, New York, NY 10032, USA; Department of Neurology, Columbia University, New York, NY 10032, USA
| | - Christian M Simon
- Carl-Ludwig-Institute for Physiology, Leipzig University, Leipzig, Germany
| | - Athanasios Chatzisotiriou
- Department of Physiology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki 541 24, Greece
| | - Maria Albani
- Department of Physiology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki 541 24, Greece
| | - George Z Mentis
- Center for Motor Neuron Biology and Disease, Columbia University, New York, NY 10032, USA; Department of Neurology, Columbia University, New York, NY 10032, USA; Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
2
|
Heinle JW, Dalessio S, Janicki P, Ouyang A, Vrana KE, Ruiz-Velasco V, Coates MD. Insights into the voltage-gated sodium channel, Na V1.8, and its role in visceral pain perception. Front Pharmacol 2024; 15:1398409. [PMID: 38855747 PMCID: PMC11158627 DOI: 10.3389/fphar.2024.1398409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 04/29/2024] [Indexed: 06/11/2024] Open
Abstract
Pain is a major issue in healthcare throughout the world. It remains one of the major clinical issues of our time because it is a common sequela of numerous conditions, has a tremendous impact on individual quality of life, and is one of the top drivers of cost in medicine, due to its influence on healthcare expenditures and lost productivity in those affected by it. Patients and healthcare providers remain desperate to find new, safer and more effective analgesics. Growing evidence indicates that the voltage-gated sodium channel Nav1.8 plays a critical role in transmission of pain-related signals throughout the body. For that reason, this channel appears to have strong potential to help develop novel, more selective, safer, and efficacious analgesics. However, many questions related to the physiology, function, and clinical utility of Nav1.8 remain to be answered. In this article, we discuss the latest studies evaluating the role of Nav1.8 in pain, with a particular focus on visceral pain, as well as the steps taken thus far to evaluate its potential as an analgesic target. We also review the limitations of currently available studies related to this topic, and describe the next scientific steps that have already been undertaken, or that will need to be pursued, to fully unlock the capabilities of this potential therapeutic target.
Collapse
Affiliation(s)
- J. Westley Heinle
- Division of Gastroenterology and Hepatology, Penn State College of Medicine, Hershey, PA, United States
| | - Shannon Dalessio
- Division of Gastroenterology and Hepatology, Penn State College of Medicine, Hershey, PA, United States
| | - Piotr Janicki
- Department of Anesthesiology and Perioperative Medicine, Penn State College of Medicine, Hershey, PA, United States
| | - Ann Ouyang
- Division of Gastroenterology and Hepatology, Penn State College of Medicine, Hershey, PA, United States
| | - Kent E. Vrana
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, United States
| | - Victor Ruiz-Velasco
- Department of Anesthesiology and Perioperative Medicine, Penn State College of Medicine, Hershey, PA, United States
| | - Matthew D. Coates
- Division of Gastroenterology and Hepatology, Penn State College of Medicine, Hershey, PA, United States
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, United States
| |
Collapse
|
3
|
Gupta S, Viotti A, Eichwald T, Roger A, Kaufmann E, Othman R, Ghasemlou N, Rafei M, Foster SL, Talbot S. Navigating the blurred path of mixed neuroimmune signaling. J Allergy Clin Immunol 2024; 153:924-938. [PMID: 38373475 DOI: 10.1016/j.jaci.2024.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 02/13/2024] [Accepted: 02/14/2024] [Indexed: 02/21/2024]
Abstract
Evolution has created complex mechanisms to sense environmental danger and protect tissues, with the nervous and immune systems playing pivotal roles. These systems work together, coordinating local and systemic reflexes to restore homeostasis in response to tissue injury and infection. By sharing receptors and ligands, they influence the pathogenesis of various diseases. Recently, a less-explored aspect of neuroimmune communication has emerged: the release of neuropeptides from immune cells and cytokines/chemokines from sensory neurons. This article reviews evidence of this unique neuroimmune interplay and its impact on the development of allergy, inflammation, itch, and pain. We highlight the effects of this neuroimmune signaling on vital processes such as host defense, tissue repair, and inflammation resolution, providing avenues for exploration of the underlying mechanisms and therapeutic potential of this signaling.
Collapse
Affiliation(s)
- Surbhi Gupta
- Centre for Neuroscience Studies, Queen's University, Kingston, Ontario, Canada
| | - Alice Viotti
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, Mass
| | - Tuany Eichwald
- Department of Pharmacology and Physiology, Karolinska Institutet, Solna, Sweden; Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Anais Roger
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada; Aix-Marseille University, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Eva Kaufmann
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Rahmeh Othman
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Nader Ghasemlou
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Moutih Rafei
- Department of Pharmacology and Physiology, University of Montréal, Montréal, Québec, Canada
| | - Simmie L Foster
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, Mass
| | - Sebastien Talbot
- Department of Pharmacology and Physiology, Karolinska Institutet, Solna, Sweden; Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada.
| |
Collapse
|
4
|
Wang YB, Page AJ, Gill TK, Melaku YA. Association of dietary and nutrient patterns with systemic inflammation in community dwelling adults. Front Nutr 2022; 9:977029. [PMID: 36082032 PMCID: PMC9445576 DOI: 10.3389/fnut.2022.977029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 08/04/2022] [Indexed: 11/23/2022] Open
Abstract
Purpose Evidence investigating associations between dietary and nutrient patterns and inflammatory biomarkers is inconsistent and scarce. Therefore, we aimed to determine the association of dietary and nutrient patterns with inflammation. Methods Overall, 1,792 participants from the North-West Adelaide Health Study were included in this cross-sectional study. We derived dietary and nutrient patterns from food frequency questionnaire data using principal component analysis. Multivariable ordinal logistic regression determined the association between dietary and nutrient patterns and the grade of inflammation (normal, moderate, and severe) based on C-reactive protein (CRP) values. Subgroup analyses were stratified by gender, obesity and metabolic health status. Results In the fully adjusted model, a plant-sourced nutrient pattern (NP) was strongly associated with a lower grade of inflammation in men (ORQ5vsQ1 = 0.59, 95% CI: 0.38–0.93, p-trend = 0.08), obesity (ORQ5vsQ1 = 0.43; 95% CI: 0.24–0.77, p-trend = 0.03) and metabolically unhealthy obesity (ORQ5vsQ1 = 0.24; 95% CI: 0.11–0.52, p-trend = 0.01). A mixed NP was positively associated with higher grade of inflammation (ORQ5vsQ1 = 1.35; 95% CI: 0.99–1.84, p-trend = 0.03) in all participants. A prudent dietary pattern was inversely associated with a lower grade of inflammation (ORQ5vsQ1 = 0.72, 95% CI: 0.52–1.01, p-trend = 0.14). In contrast, a western dietary pattern and animal-sourced NP were associated with a higher grade of inflammation in the all participants although BMI attenuated the magnitude of association (ORQ5vsQ1 = 0.83, 95% CI: 0.55–1.25; and ORQ5vsQ1 = 0.94, 95% CI: 0.63–1.39, respectively) in the fully adjusted model. Conclusion A plant-sourced NP was independently associated with lower inflammation. The association was stronger in men, and those classified as obese and metabolically unhealthy obese. Increasing consumption of plant-based foods may mitigate obesity-induced inflammation and its consequences.
Collapse
Affiliation(s)
- Yoko Brigitte Wang
- Vagal Afferent Research Group, School of Biomedicine, University of Adelaide, Adelaide, SA, Australia
- Nutrition, Diabetes and Gut Health, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Amanda J. Page
- Vagal Afferent Research Group, School of Biomedicine, University of Adelaide, Adelaide, SA, Australia
- Nutrition, Diabetes and Gut Health, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Tiffany K. Gill
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
| | - Yohannes Adama Melaku
- Nutrition, Diabetes and Gut Health, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
- Adelaide Institute for Sleep Health, College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia
- Cancer Council Victoria, Cancer Epidemiology Division, Melbourne, VIC, Australia
- *Correspondence: Yohannes Adama Melaku
| |
Collapse
|
5
|
Rahimi RA, Cho JL, Jakubzick CV, Khader SA, Lambrecht BN, Lloyd CM, Molofsky AB, Talbot S, Bonham CA, Drake WP, Sperling AI, Singer BD. Advancing Lung Immunology Research: An Official American Thoracic Society Workshop Report. Am J Respir Cell Mol Biol 2022; 67:e1-18. [PMID: 35776495 PMCID: PMC9273224 DOI: 10.1165/rcmb.2022-0167st] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The mammalian airways and lungs are exposed to a myriad of inhaled particulate matter, allergens, and pathogens. The immune system plays an essential role in protecting the host from respiratory pathogens, but a dysregulated immune response during respiratory infection can impair pathogen clearance and lead to immunopathology. Furthermore, inappropriate immunity to inhaled antigens can lead to pulmonary diseases. A complex network of epithelial, neural, stromal, and immune cells has evolved to sense and respond to inhaled antigens, including the decision to promote tolerance versus a rapid, robust, and targeted immune response. Although there has been great progress in understanding the mechanisms governing immunity to respiratory pathogens and aeroantigens, we are only beginning to develop an integrated understanding of the cellular networks governing tissue immunity within the lungs and how it changes after inflammation and over the human life course. An integrated model of airway and lung immunity will be necessary to improve mucosal vaccine design as well as prevent and treat acute and chronic inflammatory pulmonary diseases. Given the importance of immunology in pulmonary research, the American Thoracic Society convened a working group to highlight central areas of investigation to advance the science of lung immunology and improve human health.
Collapse
|
6
|
Tolman Z, Chaverra M, George L, Lefcort F. Elp1 is required for development of visceral sensory peripheral and central circuitry. Dis Model Mech 2022; 15:275184. [PMID: 35481599 PMCID: PMC9187870 DOI: 10.1242/dmm.049274] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 04/20/2022] [Indexed: 11/23/2022] Open
Abstract
Cardiovascular instability and a blunted respiratory drive in hypoxic conditions are hallmark features of the genetic sensory and autonomic neuropathy, familial dysautonomia (FD). FD results from a mutation in the gene ELP1, the encoded protein of which is a scaffolding subunit of the six-subunit Elongator complex. In mice, we and others have shown that Elp1 is essential for the normal development of neural crest-derived dorsal root ganglia sensory neurons. Whether Elp1 is also required for development of ectodermal placode-derived visceral sensory receptors, which are required for normal baroreception and chemosensory responses, has not been investigated. Using mouse models for FD, we here show that the entire circuitry underlying baroreception and chemoreception is impaired due to a requirement for Elp1 in the visceral sensory neuron ganglia, as well as for normal peripheral target innervation, and in their central nervous system synaptic partners in the medulla. Thus, Elp1 is required in both placode- and neural crest-derived sensory neurons, and its reduction aborts the normal development of neuronal circuitry essential for autonomic homeostasis and interoception. This article has an associated First Person interview with the first author of the paper. Summary: Our data indicate that Elp1 is required in both placode- and neural crest-derived sensory neurons, and that it exerts comparable effects, including survival, axonal morphology and target innervation in both lineages.
Collapse
Affiliation(s)
- Zariah Tolman
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT 59717, USA
| | - Marta Chaverra
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT 59717, USA
| | - Lynn George
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT 59717, USA.,Department of Biological and Physical Sciences, Montana State University Billings, Billings, MT 59101, USA
| | - Frances Lefcort
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT 59717, USA
| |
Collapse
|
7
|
Uhlig F, Grundy L, Garcia-Caraballo S, Brierley SM, Foster SJ, Grundy D. Identification of a Quorum Sensing-Dependent Communication Pathway Mediating Bacteria-Gut-Brain Cross Talk. iScience 2020; 23:101695. [PMID: 33163947 PMCID: PMC7607502 DOI: 10.1016/j.isci.2020.101695] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 07/03/2020] [Accepted: 10/14/2020] [Indexed: 12/12/2022] Open
Abstract
Despite recently established contributions of the intestinal microbiome to human health and disease, our understanding of bacteria-host communication pathways with regard to the gut-brain axis remains limited. Here we provide evidence that intestinal neurons are able to "sense" bacteria independently of the host immune system. Using supernatants from cultures of the opportunistic pathogen Staphylococcus aureus (S. aureus) we demonstrate the release of mediators with neuromodulatory properties at high population density. These mediators induced a biphasic response in extrinsic sensory afferent nerves, increased membrane permeability in cultured sensory neurons, and altered intestinal motility and secretion. Genetic manipulation of S. aureus revealed two key quorum sensing-regulated classes of pore forming toxins that mediate excitation and inhibition of extrinsic sensory nerves, respectively. As such, bacterial mediators have the potential to directly modulate gut-brain communication to influence intestinal symptoms and reflex function in vivo, contributing to homeostatic, behavioral, and sensory consequences of infection.
Collapse
Affiliation(s)
- Friederike Uhlig
- Department of Biomedical Science, University of Sheffield, Sheffield, UK
| | - Luke Grundy
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute (FHMRI), Flinders University, Bedford Park, SA, Australia
- Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, SA, Australia
- Discipline of Medicine, University of Adelaide, Adelaide, SA, Australia
| | - Sonia Garcia-Caraballo
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute (FHMRI), Flinders University, Bedford Park, SA, Australia
- Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, SA, Australia
- Discipline of Medicine, University of Adelaide, Adelaide, SA, Australia
| | - Stuart M. Brierley
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute (FHMRI), Flinders University, Bedford Park, SA, Australia
- Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, SA, Australia
- Discipline of Medicine, University of Adelaide, Adelaide, SA, Australia
| | - Simon J. Foster
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, UK
- Florey Institute, University of Sheffield, Sheffield, UK
| | - David Grundy
- Department of Biomedical Science, University of Sheffield, Sheffield, UK
| |
Collapse
|
8
|
Mapping of Extrinsic Innervation of the Gastrointestinal Tract in the Mouse Embryo. J Neurosci 2020; 40:6691-6708. [PMID: 32690615 DOI: 10.1523/jneurosci.0309-20.2020] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 06/05/2020] [Accepted: 07/12/2020] [Indexed: 12/12/2022] Open
Abstract
Precise extrinsic afferent (visceral sensory) and efferent (sympathetic and parasympathetic) innervation of the gut is fundamental for gut-brain cross talk. Owing to the limitation of intrinsic markers to distinctively visualize the three classes of extrinsic axons, which intimately associate within the gut mesentery, detailed information on the development of extrinsic gut-innervating axons remains relatively sparse. Here, we mapped extrinsic innervation of the gut and explored the relationships among various types of extrinsic axons during embryonic development in mice. Visualization with characterized intrinsic markers revealed that visceral sensory, sympathetic, and parasympathetic axons arise from different anatomic locations, project in close association via the gut mesentery, and form distinctive innervation patterns within the gut from embryonic day (E)10.5 to E16.5. Genetic ablation of visceral sensory trajectories results in the erratic extension of both sympathetic and parasympathetic axons, implicating that afferent axons provide an axonal scaffold to route efferent axons. Coculture assay further confirmed the attractive effect of sensory axons on sympathetic axons. Taken together, our study provides key information regarding the development of extrinsic gut-innervating axons occurring through heterotypic axonal interactions and provides an anatomic basis to uncover neural circuit assembly in the gut-brain axis (GBA).SIGNIFICANCE STATEMENT Understanding the development of extrinsic innervation of the gut is essential to unravel the bidirectional neural communication between the brain and the gut. Here, with characterized intrinsic markers targeting vagal sensory, spinal sensory, sympathetic, and parasympathetic axons, respectively, we comprehensively traced the spatiotemporal development of extrinsic axons to the gut during embryonic development in mice. Moreover, in line with the somatic nervous system, pretarget sorting via heterotypic axonal interactions is revealed to play critical roles in patterning extrinsic efferent trajectories to the gut. These findings provide basic anatomic information to explore the mechanisms underlying the process of assembling neural circuitry in the gut-brain axis (GBA).
Collapse
|
9
|
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: 254] [Impact Index Per Article: 50.8] [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
|
10
|
Yu Y, Villalobos-Hernandez EC, Pradhananga S, Baker CC, Keating C, Grundy D, Lomax AE, Reed DE. Deoxycholic acid activates colonic afferent nerves via 5-HT 3 receptor-dependent and -independent mechanisms. Am J Physiol Gastrointest Liver Physiol 2019; 317:G275-G284. [PMID: 31216174 DOI: 10.1152/ajpgi.00016.2019] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Increased bile acids in the colon can evoke increased epithelial secretion resulting in diarrhea, but little is known about whether colonic bile acids contribute to abdominal pain. This study aimed to investigate the mechanisms underlying activation of colonic extrinsic afferent nerves and their neuronal cell bodies by a major secondary bile acid, deoxycholic acid (DCA). All experiments were performed on male C57BL/6 mice. Afferent sensitivity was evaluated using in vitro extracellular recordings from mesenteric nerves in the proximal colon (innervated by vagal and spinal afferents) and distal colon (spinal afferents only). Neuronal excitability of cultured dorsal root ganglion (DRG) and nodose ganglion (NG) neurons was examined with perforated patch clamp. Colonic 5-HT release was assessed using ELISA, and 5-HT immunoreactive enterochromaffin (EC) cells were quantified. Intraluminal DCA increased afferent nerve firing rate concentration dependently in both proximal and distal colon. This DCA-elicited increase was significantly inhibited by a 5-HT3 antagonist in the proximal colon but not in the distal colon, which may be in part due to lower 5-HT immunoreactive EC cell density and lower 5-HT levels in the distal colon following DCA stimulation. DCA increased the excitability of DRG neurons, whereas it decreased the excitability of NG neurons. DCA potentiated mechanosensitivity of high-threshold spinal afferents independent of 5-HT release. Together, this study suggests that DCA can excite colonic afferents via direct and indirect mechanisms but the predominant mechanism may differ between vagal and spinal afferents. Furthermore, DCA increased mechanosensitivity of high-threshold spinal afferents and may be a mechanism of visceral hypersensitivity.NEW & NOTEWORTHY Deoxycholic acid (DCA) directly excites spinal afferents and, to a lesser extent, indirectly via mucosal 5-HT release. DCA potentiates mechanosensitivity of high-threshold spinal afferents independent of 5-HT release. DCA increases vagal afferent firing in proximal colon via 5-HT release but directly inhibits the excitability of their cell bodies.
Collapse
Affiliation(s)
- Yang Yu
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | | | - Sabindra Pradhananga
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | - Corey C Baker
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | - Christopher Keating
- Department of Biomedical Science, University of Sheffield, Sheffield, United Kingdom
| | - David Grundy
- Department of Biomedical Science, University of Sheffield, Sheffield, United Kingdom
| | - Alan E Lomax
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | - David E Reed
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
11
|
Crosson T, Roversi K, Balood M, Othman R, Ahmadi M, Wang JC, Seadi Pereira PJ, Tabatabaei M, Couture R, Eichwald T, Latini A, Prediger RD, Rangachari M, Seehus CR, Foster SL, Talbot S. Profiling of how nociceptor neurons detect danger - new and old foes. J Intern Med 2019; 286:268-289. [PMID: 31282104 DOI: 10.1111/joim.12957] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The host evolves redundant mechanisms to preserve physiological processing and homeostasis. These functions range from sensing internal and external threats, creating a memory of the insult and generating reflexes, which aim to resolve inflammation. Impairment in such functioning leads to chronic inflammatory diseases. By interacting through a common language of ligands and receptors, the immune and sensory nervous systems work in concert to accomplish such protective functions. Whilst this bidirectional communication helps to protect from danger, it can contribute to disease pathophysiology. Thus, the somatosensory nervous system is anatomically positioned within primary and secondary lymphoid tissues and mucosa to modulate immunity directly. Upstream of this interplay, neurons detect danger, which prompts the release of neuropeptides initiating (i) defensive reflexes (ranging from withdrawal response to coughing) and (ii) chemotaxis, adhesion and local infiltration of immune cells. The resulting outcome of such neuro-immune interplay is still ill-defined, but consensual findings start to emerge and support neuropeptides not only as blockers of TH 1-mediated immunity but also as drivers of TH 2 immune responses. However, the modalities detected by nociceptors revealed broader than mechanical pressure and temperature sensing and include signals as various as cytokines and pathogens to immunoglobulins and even microRNAs. Along these lines, we aggregated various dorsal root ganglion sensory neuron expression profiling datasets supporting such wide-ranging sensing capabilities to help identifying new danger detection modalities of these cells. Thus, revealing unexpected aspects of nociceptor neuron biology might prompt the identification of novel drivers of immunity, means to resolve inflammation and strategies to safeguard homeostasis.
Collapse
Affiliation(s)
- T Crosson
- From the, Département de Pharmacologie et Physiologie, Faculté de Médecine, Université de Montréal, Montréal, QC, Canada
| | - K Roversi
- From the, Département de Pharmacologie et Physiologie, Faculté de Médecine, Université de Montréal, Montréal, QC, Canada.,Departamento de Farmacologia Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - M Balood
- From the, Département de Pharmacologie et Physiologie, Faculté de Médecine, Université de Montréal, Montréal, QC, Canada.,Axe Neurosciences, Centre de recherche du CHU, Université Laval, Québec, QC, Canada.,Département de Médecine Moléculaire, Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - R Othman
- From the, Département de Pharmacologie et Physiologie, Faculté de Médecine, Université de Montréal, Montréal, QC, Canada
| | - M Ahmadi
- From the, Département de Pharmacologie et Physiologie, Faculté de Médecine, Université de Montréal, Montréal, QC, Canada
| | - J-C Wang
- From the, Département de Pharmacologie et Physiologie, Faculté de Médecine, Université de Montréal, Montréal, QC, Canada.,Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | | | - M Tabatabaei
- From the, Département de Pharmacologie et Physiologie, Faculté de Médecine, Université de Montréal, Montréal, QC, Canada
| | - R Couture
- From the, Département de Pharmacologie et Physiologie, Faculté de Médecine, Université de Montréal, Montréal, QC, Canada
| | - T Eichwald
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - A Latini
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - R D Prediger
- Departamento de Farmacologia Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - M Rangachari
- Axe Neurosciences, Centre de recherche du CHU, Université Laval, Québec, QC, Canada.,Département de Médecine Moléculaire, Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - C R Seehus
- FM Kirby Neurobiology Center, Children's Hospital, Boston, MA, USA
| | - S L Foster
- Depression Clinical Research Program, Massachusetts General Hospital, Boston, MA, USA
| | - S Talbot
- From the, Département de Pharmacologie et Physiologie, Faculté de Médecine, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
12
|
Gunasekaran M, Chatterjee PK, Shih A, Imperato GH, Addorisio M, Kumar G, Lee A, Graf JF, Meyer D, Marino M, Puleo C, Ashe J, Cox MA, Mak TW, Bouton C, Sherry B, Diamond B, Andersson U, Coleman TR, Metz CN, Tracey KJ, Chavan SS. Immunization Elicits Antigen-Specific Antibody Sequestration in Dorsal Root Ganglia Sensory Neurons. Front Immunol 2018; 9:638. [PMID: 29755449 PMCID: PMC5932385 DOI: 10.3389/fimmu.2018.00638] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Accepted: 03/14/2018] [Indexed: 12/11/2022] Open
Abstract
The immune and nervous systems are two major organ systems responsible for host defense and memory. Both systems achieve memory and learning that can be retained, retrieved, and utilized for decades. Here, we report the surprising discovery that peripheral sensory neurons of the dorsal root ganglia (DRGs) of immunized mice contain antigen-specific antibodies. Using a combination of rigorous molecular genetic analyses, transgenic mice, and adoptive transfer experiments, we demonstrate that DRGs do not synthesize these antigen-specific antibodies, but rather sequester primarily IgG1 subtype antibodies. As revealed by RNA-seq and targeted quantitative PCR (qPCR), dorsal root ganglion (DRG) sensory neurons harvested from either naïve or immunized mice lack enzymes (i.e., RAG1, RAG2, AID, or UNG) required for generating antibody diversity and, therefore, cannot make antibodies. Additionally, transgenic mice that express a reporter fluorescent protein under the control of Igγ1 constant region fail to express Ighg1 transcripts in DRG sensory neurons. Furthermore, neural sequestration of antibodies occurs in mice rendered deficient in neuronal Rag2, but antibody sequestration is not observed in DRG sensory neurons isolated from mice that lack mature B cells [e.g., Rag1 knock out (KO) or μMT mice]. Finally, adoptive transfer of Rag1-deficient bone marrow (BM) into wild-type (WT) mice or WT BM into Rag1 KO mice revealed that antibody sequestration was observed in DRG sensory neurons of chimeric mice with WT BM but not with Rag1-deficient BM. Together, these results indicate that DRG sensory neurons sequester and retain antigen-specific antibodies released by antibody-secreting plasma cells. Coupling this work with previous studies implicating DRG sensory neurons in regulating antigen trafficking during immunization raises the interesting possibility that the nervous system collaborates with the immune system to regulate antigen-mediated responses.
Collapse
Affiliation(s)
- Manojkumar Gunasekaran
- Center for Biomedical Science, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Prodyot K. Chatterjee
- Center for Biomedical Science, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Andrew Shih
- Center for Genomics and Human Genetics, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Gavin H. Imperato
- Center for Biomedical Science, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
- Elmezzi Graduate School, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Meghan Addorisio
- Center for Biomedical Science, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Gopal Kumar
- Center for Biomedical Science, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
- Elmezzi Graduate School, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Annette Lee
- Center for Genomics and Human Genetics, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
- Elmezzi Graduate School, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
| | - John F. Graf
- GE Global Research Center, Niskayuna, NY, United States
| | - Dan Meyer
- GE Global Research Center, Niskayuna, NY, United States
| | | | | | - Jeffrey Ashe
- GE Global Research Center, Niskayuna, NY, United States
| | - Maureen A. Cox
- The Campbell Family Institute for Breast Cancer Research, University Health Network, Toronto, ON, Canada
| | - Tak W. Mak
- The Campbell Family Institute for Breast Cancer Research, University Health Network, Toronto, ON, Canada
| | - Chad Bouton
- Center for Bioelectronic Medicine, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Barbara Sherry
- Elmezzi Graduate School, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
- Center for Immunology and Inflammation, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Betty Diamond
- Elmezzi Graduate School, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
- Center for Autoimmune, Musculoskeletal and Hematopoietic Diseases, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Ulf Andersson
- Department of Women’s and Children’s Health, Karolinska Institutet, Solna, Sweden
| | - Thomas R. Coleman
- Center for Molecular Innovation, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Christine N. Metz
- Center for Biomedical Science, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
- Elmezzi Graduate School, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
| | - Kevin J. Tracey
- Center for Biomedical Science, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
- Elmezzi Graduate School, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
- Center for Bioelectronic Medicine, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Sangeeta S. Chavan
- Center for Biomedical Science, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
- Elmezzi Graduate School, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
- Center for Bioelectronic Medicine, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
| |
Collapse
|
13
|
Foster SL, Seehus CR, Woolf CJ, Talbot S. Sense and Immunity: Context-Dependent Neuro-Immune Interplay. Front Immunol 2017; 8:1463. [PMID: 29163530 PMCID: PMC5675863 DOI: 10.3389/fimmu.2017.01463] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 10/19/2017] [Indexed: 12/21/2022] Open
Abstract
The sensory nervous and immune systems, historically considered autonomous, actually work in concert to promote host defense and tissue homeostasis. These systems interact with each other through a common language of cell surface G protein-coupled receptors and receptor tyrosine kinases as well as cytokines, growth factors, and neuropeptides. While this bidirectional communication is adaptive in many settings, helping protect from danger, it can also become maladaptive and contribute to disease pathophysiology. The fundamental logic of how, where, and when sensory neurons and immune cells contribute to either health or disease remains, however, unclear. Our lab and others’ have begun to explore how this neuro-immune reciprocal dialog contributes to physiological and pathological immune responses and sensory disorders. The cumulative results collected so far indicate that there is an important role for nociceptors (noxious stimulus detecting sensory neurons) in driving immune responses, but that this is highly context dependent. To illustrate this concept, we present our findings in a model of airway inflammation, in which nociceptors seem to have major involvement in type 2 but not type 1 adaptive immunity.
Collapse
Affiliation(s)
- Simmie L Foster
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, United States.,Department of Neurobiology, Harvard Medical School, Boston, MA, United States.,Department of Psychiatry, Harvard Medical School, Boston, MA, United States.,Depression Clinical Research Program, Massachusetts General Hospital, Boston, MA, United States
| | - Corey R Seehus
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, United States.,Department of Neurobiology, Harvard Medical School, Boston, MA, United States
| | - Clifford J Woolf
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, United States.,Department of Neurobiology, Harvard Medical School, Boston, MA, United States
| | - Sébastien Talbot
- Département de Pharmacologie et Physiologie, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
14
|
Distinct and common expression of receptors for inflammatory mediators in vagal nodose versus jugular capsaicin-sensitive/TRPV1-positive neurons detected by low input RNA sequencing. PLoS One 2017; 12:e0185985. [PMID: 28982197 PMCID: PMC5628920 DOI: 10.1371/journal.pone.0185985] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 09/23/2017] [Indexed: 11/19/2022] Open
Abstract
Capsaicin-sensitive sensory C-fibers derived from vagal ganglia innervate the visceral organs, and respond to inflammatory mediators and noxious stimuli. These neurons play an important role in maintenance of visceral homeostasis, and contribute to the symptoms of visceral inflammatory diseases. Vagal sensory neurons are located in two ganglia, the jugular ganglia (derived from the neural crest), and the nodose ganglia (from the epibranchial placodes). The functional difference, especially in response to immune mediators, between jugular and nodose neurons is not fully understood. In this study, we microscopically isolated murine nodose and jugular capsaicin-sensitive / Trpv1-expressing C-fiber neurons and performed transcriptome profiling using ultra-low input RNA sequencing. RNAseq detected genes with significantly differential expression in jugular and nodose neurons, which were mostly involved in neural functions. Transcriptional regulators, including Cited1, Hoxb5 and Prdm12 showed distinct expression patterns in the two C-fiber neuronal populations. Common and specific expression of immune receptor proteins was characterized in each neuronal type. The expression of immune receptors that have received little or no attention from vagal sensory biologists is highlighted including receptors for certain chemokines (CXCLs), interleukins (IL-4) and interferons (IFNα, IFNγ). Stimulation of immune receptors with their cognate ligands led to activation of the C-fibers in isolated functional assays.
Collapse
|
15
|
Dorsal root ganglion neurons and tyrosine hydroxylase--an intriguing association with implications for sensation and pain. Pain 2016; 157:314-320. [PMID: 26447702 DOI: 10.1097/j.pain.0000000000000381] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
16
|
Szklarczyk R, Megchelenbrink W, Cizek P, Ledent M, Velemans G, Szklarczyk D, Huynen MA. WeGET: predicting new genes for molecular systems by weighted co-expression. Nucleic Acids Res 2015; 44:D567-73. [PMID: 26582928 PMCID: PMC4702868 DOI: 10.1093/nar/gkv1228] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 10/30/2015] [Indexed: 01/04/2023] Open
Abstract
We have developed the Weighted Gene Expression Tool and database (WeGET, http://weget.cmbi.umcn.nl) for the prediction of new genes of a molecular system by correlated gene expression. WeGET utilizes a compendium of 465 human and 560 murine gene expression datasets that have been collected from multiple tissues under a wide range of experimental conditions. It exploits this abundance of expression data by assigning a high weight to datasets in which the known genes of a molecular system are harmoniously up- and down-regulated. WeGET ranks new candidate genes by calculating their weighted co-expression with that system. A weighted rank is calculated for human genes and their mouse orthologs. Then, an integrated gene rank and p-value is computed using a rank-order statistic. We applied our method to predict novel genes that have a high degree of co-expression with Gene Ontology terms and pathways from KEGG and Reactome. For each query set we provide a list of predicted novel genes, computed weights for transcription datasets used and cell and tissue types that contributed to the final predictions. The performance for each query set is assessed by 10-fold cross-validation. Finally, users can use the WeGET to predict novel genes that co-express with a custom query set.
Collapse
Affiliation(s)
- Radek Szklarczyk
- Maastricht University Medical Centre, P. Debyelaan 25, 6226XM Maastricht, The Netherlands Centre for Molecular and Biomolecular Informatics (CMBI), Radboud University Medical Centre, Geert Grooteplein Zuid 26-28, 6525 GA Nijmegen, Nijmegen, The Netherlands
| | - Wout Megchelenbrink
- Centre for Molecular and Biomolecular Informatics (CMBI), Radboud University Medical Centre, Geert Grooteplein Zuid 26-28, 6525 GA Nijmegen, Nijmegen, The Netherlands Institute for Computing and Information Sciences (ICIS), Radboud University, Toernooiveld 212, 6525 EC Nijmegen, The Netherlands Centre for Systems Biology and Bioenergetics (CSBB), Radboud University Medical Centre, Geert Grooteplein Zuid 10, 6500 HB Nijmegen, Nijmegen, The Netherlands
| | - Pavel Cizek
- Centre for Molecular and Biomolecular Informatics (CMBI), Radboud University Medical Centre, Geert Grooteplein Zuid 26-28, 6525 GA Nijmegen, Nijmegen, The Netherlands
| | - Marie Ledent
- Maastricht University Medical Centre, P. Debyelaan 25, 6226XM Maastricht, The Netherlands
| | - Gonny Velemans
- Centre for Molecular and Biomolecular Informatics (CMBI), Radboud University Medical Centre, Geert Grooteplein Zuid 26-28, 6525 GA Nijmegen, Nijmegen, The Netherlands
| | - Damian Szklarczyk
- Institute of Molecular Life Sciences and Swiss Institute of Bioinformatics, University of Zurich, 8057 Zurich, Switzerland
| | - Martijn A Huynen
- Centre for Molecular and Biomolecular Informatics (CMBI), Radboud University Medical Centre, Geert Grooteplein Zuid 26-28, 6525 GA Nijmegen, Nijmegen, The Netherlands Centre for Systems Biology and Bioenergetics (CSBB), Radboud University Medical Centre, Geert Grooteplein Zuid 10, 6500 HB Nijmegen, Nijmegen, The Netherlands
| |
Collapse
|
17
|
Isensee J, Wenzel C, Buschow R, Weissmann R, Kuss AW, Hucho T. Subgroup-elimination transcriptomics identifies signaling proteins that define subclasses of TRPV1-positive neurons and a novel paracrine circuit. PLoS One 2014; 9:e115731. [PMID: 25551770 PMCID: PMC4281118 DOI: 10.1371/journal.pone.0115731] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 11/29/2014] [Indexed: 12/24/2022] Open
Abstract
Normal and painful stimuli are detected by specialized subgroups of peripheral sensory neurons. The understanding of the functional differences of each neuronal subgroup would be strongly enhanced by knowledge of the respective subgroup transcriptome. The separation of the subgroup of interest, however, has proven challenging as they can hardly be enriched. Instead of enriching, we now rapidly eliminated the subgroup of neurons expressing the heat-gated cation channel TRPV1 from dissociated rat sensory ganglia. Elimination was accomplished by brief treatment with TRPV1 agonists followed by the removal of compromised TRPV1(+) neurons using density centrifugation. By differential microarray and sequencing (RNA-Seq) based expression profiling we compared the transcriptome of all cells within sensory ganglia versus the same cells lacking TRPV1 expressing neurons, which revealed 240 differentially expressed genes (adj. p<0.05, fold-change>1.5). Corroborating the specificity of the approach, many of these genes have been reported to be involved in noxious heat or pain sensitization. Beyond the expected enrichment of ion channels, we found the TRPV1 transcriptome to be enriched for GPCRs and other signaling proteins involved in adenosine, calcium, and phosphatidylinositol signaling. Quantitative population analysis using a recent High Content Screening (HCS) microscopy approach identified substantial heterogeneity of expressed target proteins even within TRPV1-positive neurons. Signaling components defined distinct further subgroups within the population of TRPV1-positive neurons. Analysis of one such signaling system showed that the pain sensitizing prostaglandin PGD2 activates DP1 receptors expressed predominantly on TRPV1(+) neurons. In contrast, we found the PGD2 producing prostaglandin D synthase to be expressed exclusively in myelinated large-diameter neurons lacking TRPV1, which suggests a novel paracrine neuron-neuron communication. Thus, subgroup analysis based on the elimination rather than enrichment of the subgroup of interest revealed proteins that define subclasses of TRPV1-positive neurons and suggests a novel paracrine circuit.
Collapse
Affiliation(s)
- Jörg Isensee
- Department of Anesthesiology and Intensive Care Medicine, Experimental Anesthesiology and Pain Research, University Hospital of Cologne, Cologne, Germany
- Department for Human Molecular Genetics, Max Planck Institute for Molecular Genetics, Berlin, Germany
- * E-mail:
| | - Carsten Wenzel
- Department for Human Molecular Genetics, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Rene Buschow
- Department for Human Molecular Genetics, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Robert Weissmann
- Department of Human Genetics, University Medicine Greifswald and Interfaculty Institute of Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| | - Andreas W. Kuss
- Department of Human Genetics, University Medicine Greifswald and Interfaculty Institute of Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| | - Tim Hucho
- Department of Anesthesiology and Intensive Care Medicine, Experimental Anesthesiology and Pain Research, University Hospital of Cologne, Cologne, Germany
- Department for Human Molecular Genetics, Max Planck Institute for Molecular Genetics, Berlin, Germany
| |
Collapse
|
18
|
Abnormal cardiac autonomic regulation in mice lacking ASIC3. BIOMED RESEARCH INTERNATIONAL 2014; 2014:709159. [PMID: 24804235 PMCID: PMC3996306 DOI: 10.1155/2014/709159] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2013] [Accepted: 02/25/2014] [Indexed: 12/11/2022]
Abstract
Integration of sympathetic and parasympathetic outflow is essential in maintaining normal cardiac autonomic function. Recent studies demonstrate that acid-sensing ion channel 3 (ASIC3) is a sensitive acid sensor for cardiac ischemia and prolonged mild acidification can open ASIC3 and evoke a sustained inward current that fires action potentials in cardiac sensory neurons. However, the physiological role of ASIC3 in cardiac autonomic regulation is not known. In this study, we elucidate the role of ASIC3 in cardiac autonomic function using Asic3−/− mice. Asic3−/− mice showed normal baseline heart rate and lower blood pressure as compared with their wild-type littermates. Heart rate variability analyses revealed imbalanced autonomic regulation, with decreased sympathetic function. Furthermore, Asic3−/− mice demonstrated a blunted response to isoproterenol-induced cardiac tachycardia and prolonged duration to recover to baseline heart rate. Moreover, quantitative RT-PCR analysis of gene expression in sensory ganglia and heart revealed that no gene compensation for muscarinic acetylcholines receptors and beta-adrenalin receptors were found in Asic3−/− mice. In summary, we unraveled an important role of ASIC3 in regulating cardiac autonomic function, whereby loss of ASIC3 alters the normal physiological response to ischemic stimuli, which reveals new implications for therapy in autonomic nervous system-related cardiovascular diseases.
Collapse
|
19
|
Tarr AJ, Chen Q, Wang Y, Sheridan JF, Quan N. Neural and behavioral responses to low-grade inflammation. Behav Brain Res 2012; 235:334-41. [PMID: 22898545 DOI: 10.1016/j.bbr.2012.07.038] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2012] [Revised: 06/26/2012] [Accepted: 07/26/2012] [Indexed: 12/23/2022]
Abstract
Neural and behavioral responses after peripheral immune challenge have been observed in numerous studies. The majority of these studies have utilized relatively high doses of lipopolysaccharide (LPS) as the immune stimulant. Little attention has been given to the effects of LPS dose ranges that simulate low grade-inflammation. The current studies were designed to characterize neural and behavioral responses following low-dose LPS stimulation. Results show burrowing and open field activity was significantly impaired following a single i.p. injection of 10, but not 1, μg/kg of LPS. In addition, following repeated 1 μg/kg LPS administration for 10 days, animals showed the progressive development of motor deficits over time. To correlate behavior with CNS activity, cFos activation was determined in the paraventricular nucleus, nucleus of the solitary tract, central amygdaloid nucleus, and ventrolateral medulla. Data revealed there was a dose-dependent activation in all brain areas examined, but only the PVN showed significant activation by low-dose LPS. Additionally, animals that received 1 μg/kg of LPS for 8 days had PVN cFos activation similar to animals that received a single 10 μg/kg LPS injection. These data demonstrate neural and behavior responses can be induced by low-grade inflammation and chronic exposure to sub-threshold levels of LPS can precipitate significantly heightened neural and behavioral responses.
Collapse
Affiliation(s)
- Andrew J Tarr
- Division of Oral Biology, The Ohio State University, 305 W. 12th Ave, Columbus, OH 43210, USA.
| | | | | | | | | |
Collapse
|
20
|
Buchser WJ, Smith RP, Pardinas JR, Haddox CL, Hutson T, Moon L, Hoffman SR, Bixby JL, Lemmon VP. Peripheral nervous system genes expressed in central neurons induce growth on inhibitory substrates. PLoS One 2012; 7:e38101. [PMID: 22701605 PMCID: PMC3368946 DOI: 10.1371/journal.pone.0038101] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Accepted: 05/03/2012] [Indexed: 11/28/2022] Open
Abstract
Trauma to the spinal cord and brain can result in irreparable loss of function. This failure of recovery is in part due to inhibition of axon regeneration by myelin and chondroitin sulfate proteoglycans (CSPGs). Peripheral nervous system (PNS) neurons exhibit increased regenerative ability compared to central nervous system neurons, even in the presence of inhibitory environments. Previously, we identified over a thousand genes differentially expressed in PNS neurons relative to CNS neurons. These genes represent intrinsic differences that may account for the PNS's enhanced regenerative ability. Cerebellar neurons were transfected with cDNAs for each of these PNS genes to assess their ability to enhance neurite growth on inhibitory (CSPG) or permissive (laminin) substrates. Using high content analysis, we evaluated the phenotypic profile of each neuron to extract meaningful data for over 1100 genes. Several known growth associated proteins potentiated neurite growth on laminin. Most interestingly, novel genes were identified that promoted neurite growth on CSPGs (GPX3, EIF2B5, RBMX). Bioinformatic approaches also uncovered a number of novel gene families that altered neurite growth of CNS neurons.
Collapse
Affiliation(s)
- William J. Buchser
- Miami Project to Cure Paralysis, Departments of Pharmacology and Neurological Surgery, and Neuroscience Program, University of Miami, Miller School of Medicine, Miami, Florida, United States of America
| | - Robin P. Smith
- Miami Project to Cure Paralysis, Departments of Pharmacology and Neurological Surgery, and Neuroscience Program, University of Miami, Miller School of Medicine, Miami, Florida, United States of America
| | - Jose R. Pardinas
- Egea Biosciences, La Jolla, California, United States of America
| | - Candace L. Haddox
- Miami Project to Cure Paralysis, Departments of Pharmacology and Neurological Surgery, and Neuroscience Program, University of Miami, Miller School of Medicine, Miami, Florida, United States of America
| | - Thomas Hutson
- Wolfson Centre for Age-Related Diseases, King’s College London, London, United Kingdom
| | - Lawrence Moon
- Wolfson Centre for Age-Related Diseases, King’s College London, London, United Kingdom
| | - Stanley R. Hoffman
- Division of Rheumatology and Immunology, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - John L. Bixby
- Miami Project to Cure Paralysis, Departments of Pharmacology and Neurological Surgery, and Neuroscience Program, University of Miami, Miller School of Medicine, Miami, Florida, United States of America
| | - Vance P. Lemmon
- Miami Project to Cure Paralysis, Departments of Pharmacology and Neurological Surgery, and Neuroscience Program, University of Miami, Miller School of Medicine, Miami, Florida, United States of America
| |
Collapse
|
21
|
Brozmanova M, Ru F, Surdenikova L, Mazurova L, Taylor-Clark T, Kollarik M. Preferential activation of the vagal nodose nociceptive subtype by TRPA1 agonists in the guinea pig esophagus. Neurogastroenterol Motil 2011; 23:e437-45. [PMID: 21883700 PMCID: PMC3175634 DOI: 10.1111/j.1365-2982.2011.01768.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND The TRPA1 receptor is directly activated by a wide range of chemicals including many endogenous molecules relevant for esophageal pathophysiology. We addressed the hypothesis that the TRPA1 agonists differentially activate esophageal nociceptive subtypes depending on their embryological source (neural crest or epibranchial placodes). METHODS Single cell RT-PCR and whole cell patch clamp recordings were performed on the vagal neurons retrogradely labeled from the guinea pig esophagus. Extracellular recordings were made in the isolated innervated esophagus preparation ex vivo. KEY RESULTS Single cell RT-PCR revealed that the majority of the nodose (placodes-derived) and jugular (neural crest-derived) TRPV1-positive esophageal nociceptors express TRPA1. Single fiber recording showed that the TRPA1 agonists allyl-isothiocyanate (AITC) and cinnamaldehyde were effective in inducing robust action potential discharge in the nerve terminals of nodose nociceptors, but had far less effect in jugular nociceptors (approximately fivefold less). Higher efficacy of the TRPA1 agonists to activate nodose nociceptors was confirmed in the isolated esophagus-labeled vagal neurons in the whole cell patch clamp studies. Similarly to neural crest-derived vagal jugular nociceptors, the spinal DRG nociceptors that are also neural crest-derived were only modestly activated by allyl-isothiocyanate. CONCLUSIONS & INFERENCES We conclude that the TRPA1 agonists are substantially more effective activators of the placodes-derived than the neural crest-derived esophageal nociceptors. Our data predict that in esophageal diseases the presence of endogenous TRPA1 activators will be preferentially signaled by the vagal nodose nociceptors.
Collapse
Affiliation(s)
- M Brozmanova
- Pathophysiology, Jessenius Medical School, Comenius University, Martin, Slovakia
| | - F Ru
- Medicine, The Johns Hopkins School of Medicine, Baltimore, MD
| | - L Surdenikova
- Pathophysiology, Jessenius Medical School, Comenius University, Martin, Slovakia,Medicine, The Johns Hopkins School of Medicine, Baltimore, MD
| | - L Mazurova
- Pathophysiology, Jessenius Medical School, Comenius University, Martin, Slovakia
| | - T Taylor-Clark
- Medicine, The Johns Hopkins School of Medicine, Baltimore, MD
| | - M. Kollarik
- Medicine, The Johns Hopkins School of Medicine, Baltimore, MD
| |
Collapse
|
22
|
Ratcliffe EM. Molecular development of the extrinsic sensory innervation of the gastrointestinal tract. Auton Neurosci 2010; 161:1-5. [PMID: 21147045 DOI: 10.1016/j.autneu.2010.11.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2009] [Revised: 10/27/2010] [Accepted: 11/02/2010] [Indexed: 12/19/2022]
Abstract
The extrinsic sensory innervation of the gastrointestinal tract is the conduit through which the gut and the central nervous system communicate. The hindbrain receives information directly from the bowel via the vagus nerve, while information from spinal afferents arrives in the central nervous system through the dorsal root ganglia. This review focuses on the molecular development of these vagal and spinal innervations, with an emphasis on mechanisms that involve axon guidance. During development, axons from both the nodose ganglia and dorsal root ganglia grow into the gut, innervate their appropriate enteric targets and avoid inappropriate cells in the gut wall. These developmental outcomes suggest that both attractive and repellent molecules are important in establishing the normal pattern of the extrinsic sensory innervation. In the fetal mouse gut, the guidance of vagal sensory axons is mediated by axon guidance molecules, such as netrin and the netrin receptor, deleted in colorectal cancer (DCC), as well as extracellular matrix molecules, such as laminin-111. Dorsal root ganglion neurons are known to express, and their axons to respond to, axon guidance molecules. The question of whether or not these molecules are involved in guiding spinal afferents to the bowel, however, has not yet been examined. It is anticipated that a better understanding of how vagal and spinal afferents innervate the fetal gut and reach specific enteric locations will provide deeper insights into the underlying mechanisms of normal and abnormal sensation from the gastrointestinal tract.
Collapse
Affiliation(s)
- Elyanne M Ratcliffe
- Division of Gastroenterology and Nutrition, Department of Pediatrics, McMaster University, Canada.
| |
Collapse
|
23
|
Heterosynaptic crosstalk: GABA-glutamate metabotropic receptors interactively control glutamate release in solitary tract nucleus. Neuroscience 2010; 174:1-9. [PMID: 21129447 DOI: 10.1016/j.neuroscience.2010.11.053] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2010] [Revised: 11/12/2010] [Accepted: 11/24/2010] [Indexed: 11/22/2022]
Abstract
Synaptic terminals often contain metabotropic receptors that act as autoreceptors to control neurotransmitter release. Less appreciated is the heterosynaptic crossover of glutamate receptors to control GABA release and vice versa GABA receptors which control glutamate release. In the brainstem, activation of solitary tract (ST) afferents releases glutamate onto second-order neurons within the solitary tract nucleus (NTS). Multiple metabotropic receptors are expressed in NTS for glutamate (mGluRs) and for GABA (GABA(B)). The present report identifies mGluR regulation of glutamate release at second and higher order sensory neurons in NTS slices. We found strong inhibition of glutamate release to group II and III mGluR activation on mechanically isolated NTS neurons. However, the same mGluR-selective antagonists paradoxically decreased glutamate release (miniature, mEPSCs) at identified second-order NTS neurons. Unaltered amplitudes were consistent with selective presynaptic mGluR actions. GABA(B) blockade in slices resolved the paradoxical differences and revealed a group II/III mGluR negative feedback of mEPSC frequency similar to isolated neurons. Thus, the balance of glutamate control is tipped by mGluR receptors on GABA terminals resulting in predominating heterosynaptic GABA(B) inhibition of glutamate release. Regulation by mGluR or GABA(B) was not consistently evident in excitatory postsynaptic currents (EPSCs) in higher-order NTS neurons demonstrating metabotropic receptor distinctions in processing at different NTS pathway stages. These cellular localizations may figure importantly in understanding interventions such as brain-penetrant compounds or microinjections. We conclude that afferent glutamate release in NTS produces a coordinate presynaptic activation of co-localized mGluR and GABA(B) feedback on cranial afferent terminals to regulate glutamate release.
Collapse
|
24
|
Fischer MJM, Leffler A, Niedermirtl F, Kistner K, Eberhardt M, Reeh PW, Nau C. The general anesthetic propofol excites nociceptors by activating TRPV1 and TRPA1 rather than GABAA receptors. J Biol Chem 2010; 285:34781-92. [PMID: 20826794 PMCID: PMC2966094 DOI: 10.1074/jbc.m110.143958] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2010] [Revised: 08/19/2010] [Indexed: 11/06/2022] Open
Abstract
Anesthetic agents can induce a paradox activation and sensitization of nociceptive sensory neurons and, thus, potentially facilitate pain processing. Here we identify distinct molecular mechanisms that mediate an activation of sensory neurons by 2,6-diisopropylphenol (propofol), a commonly used intravenous anesthetic known to elicit intense pain upon injection. Clinically relevant concentrations of propofol activated the recombinant transient receptor potential (TRP) receptors TRPA1 and TRPV1 heterologously expressed in HEK293t cells. In dorsal root ganglion (DRG) neurons, propofol-induced activation correlated better to expression of TRPA1 than of TRPV1. However, pretreatment with the protein kinase C activator 4β-phorbol 12-myristate 13-acetate (PMA) resulted in a significantly sensitized propofol-induced activation of TRPV1 in DRG neurons as well as in HEK293t cells. Pharmacological and genetic silencing of both TRPA1 and TRPV1 only partially abrogated propofol-induced responses in DRG neurons. The remaining propofol-induced activation was abolished by the selective γ-aminobutyric acid, type A (GABA(A)) receptor antagonist picrotoxin. Propofol but not GABA evokes a release of calcitonin gene-related peptide, a key component of neurogenic inflammation, from isolated peripheral nerves of wild-type but not TRPV1 and TRPA1-deficient mice. Moreover, propofol but not GABA induced an intense pain upon intracutaneous injection. As both the release of calcitonin gene-related peptide and injection pain by propofol seem to be independent of GABA(A) receptors, our data identify TRPV1 and TRPA1 as key molecules for propofol-induced excitation of sensory neurons. This study warrants further investigations into the role of anesthetics to induce nociceptor sensitization and to foster postoperative pain.
Collapse
Affiliation(s)
- Michael J M Fischer
- Institute of Physiology and Pathophysiology, University of Erlangen-Nuremberg, Erlangen 91054, Germany
| | | | | | | | | | | | | |
Collapse
|
25
|
Smith RP, Lerch-Haner JK, Pardinas JR, Buchser WJ, Bixby JL, Lemmon VP. Transcriptional profiling of intrinsic PNS factors in the postnatal mouse. Mol Cell Neurosci 2010; 46:32-44. [PMID: 20696251 DOI: 10.1016/j.mcn.2010.07.015] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2010] [Revised: 07/22/2010] [Accepted: 07/27/2010] [Indexed: 11/25/2022] Open
Abstract
Neurons in the peripheral nervous system (PNS) display a higher capacity to regenerate after injury than those in the central nervous system, suggesting cell specific transcriptional modules underlying axon growth and inhibition. We report a systems biology based search for PNS specific transcription factors (TFs). Messenger RNAs enriched in dorsal root ganglion (DRG) neurons compared to cerebellar granule neurons (CGNs) were identified using subtractive hybridization and DNA microarray approaches. Network and transcription factor binding site enrichment analyses were used to further identify TFs that may be differentially active. Combining these techniques, we identified 32 TFs likely to be enriched and/or active in the PNS. Twenty-five of these TFs were then tested for an ability to promote CNS neurite outgrowth in an overexpression screen. Real-time PCR and immunohistochemical studies confirmed that one representative TF, STAT3, is intrinsic to PNS neurons, and that constitutively active STAT3 is sufficient to promote CGN neurite outgrowth.
Collapse
Affiliation(s)
- Robin P Smith
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, Miller School of Medicine, University of Miami, 1400 NW 12th Ave, Miami, FL 33136, USA
| | | | | | | | | | | |
Collapse
|
26
|
Mazzoni M, Clavenzani P, Minieri L, Russo D, Chiocchetti R, Lalatta-Costerbosa G. Extrinsic afferents supplying the ovine duodenum and ileum. Res Vet Sci 2010; 88:361-8. [DOI: 10.1016/j.rvsc.2009.11.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2009] [Revised: 10/14/2009] [Accepted: 11/19/2009] [Indexed: 10/20/2022]
|
27
|
Pozsgai G, Bodkin JV, Graepel R, Bevan S, Andersson DA, Brain SD. Evidence for the pathophysiological relevance of TRPA1 receptors in the cardiovascular system in vivo. Cardiovasc Res 2010; 87:760-8. [PMID: 20442136 DOI: 10.1093/cvr/cvq118] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
AIMS The aim of the study is to investigate transient receptor potential ankyrin 1 (TRPA1)-induced responses in the vasculature and on blood pressure and heart rate (HR), in response to TRPA1 agonists using wild-type (WT) and TRPA1 knockout (KO) mice. METHODS AND RESULTS TRPA1 agonists allyl isothiocyanate and cinnamaldehyde (CA) significantly increased blood flow in the skin of anaesthetized WT, but not in TRPA1 KO mice. CA also induced TRPA1-dependent relaxation of mesenteric arteries. Intravenously injected CA induced a transient hypotensive response accompanied by decreased HR that was, depending on genotype and dose, followed by a more sustained dose-dependent pressor response (10-320 micromol/kg). CA (80 micromol/kg) induced a depressor response that was significantly less in TRPA1 KO mice, with minimal pressor effects. The pressor response of a higher CA dose (320 micromol/kg) was observed in WT but not in TRPA1 KO mice, indicating involvement of TRPA1. Experiments using TRP vanilloid 1 (TRPV1) KO and calcitonin gene-related peptide (CGRP) KO mice provided little evidence for the involvement of TRPV1 or CGRP, nor did blocking substance P receptors affect responses. However, the cholinergic antagonist atropine sulphate (5 mg/kg) significantly inhibited the depressor response and slowed HR with CA (80 micromol/kg), but had no effect on pressor responses. The pressor response remained unaffected, even in the presence of the ganglion blocker hexamethonium bromide (1 mg/kg). The alpha-adrenergic blocker prazosin hydrochloride (1 mg/kg) significantly inhibited both components, but not slowed HR. CONCLUSION TRPA1 is involved in mediating vasodilation. TRPA1 can also influence changes in blood pressure of possible relevance to autonomic system reflexes and potentially to vasovagal/neurocardiogenic syncope disorders.
Collapse
Affiliation(s)
- Gabor Pozsgai
- King's College London British Heart Foundation Centre, Cardiovascular Division and Centre of Integrative Biomedicine, King's College London, Franklin-Wilkins Building, London SE1 9NH, UK
| | | | | | | | | | | |
Collapse
|
28
|
Villarreal CF, Funez MI, Figueiredo F, Cunha FQ, Parada CA, Ferreira SH. Acute and persistent nociceptive paw sensitisation in mice: the involvement of distinct signalling pathways. Life Sci 2009; 85:822-9. [PMID: 19896488 DOI: 10.1016/j.lfs.2009.10.018] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2009] [Revised: 10/20/2009] [Accepted: 10/26/2009] [Indexed: 11/26/2022]
Abstract
AIMS Many fundamental pharmacological studies in pain and inflammation have been performed on rats. However, the pharmacological findings were generally not extended to other species in order to increase their predictive therapeutic value. We studied acute and chronic inflammatory nociceptive sensitisation of mouse hind paws by prostaglandin E(2) (PGE(2)) or dopamine (DA), as previously described in rats. We also investigated the participation of the signalling pathways in acute and persistent sensitisation. MAIN METHODS Mechanical sensitisation (hypernociception) induced by intraplantar administrations of PGE(2) or DA was evaluated with an electronic pressure meter. The signalling pathways were pharmacologically investigated with the pre-administration of adenylyl cyclase (AC), cAMP-dependent protein kinase (PKA), protein kinase Cepsilon (PKCepsilon), and the extracellular signal-related kinase (ERK) inhibitors. KEY FINDINGS Single or 14days of successive intraplantar injections of PGE(2) or DA-induced acute and persistent hypernociception (lasting for more than 30days), respectively. The involvement of AC, PKA or PKCepsilon was observed in the acute hypernociception induced by PGE(2), while PKA or PKCepsilon were continuously activated during the period of persistent hypernociception. The acute hypernociception induced by DA involves activation of ERK, PKCepsilon, AC or PKA, while persistent hypernociception implicated ERK activation, but not PKA, PKCepsilon or AC. SIGNIFICANCE In mice, acute and persistent paw sensitisation involves the different activation of kinases, as previously described for rats. This study opens the possibility of comparing pharmacological approaches in both species to further understand acute and chronic inflammatory sensitisation, and possibly associated genetic manipulations.
Collapse
|
29
|
van den Wijngaard RM, Klooker TK, Welting O, Stanisor OI, Wouters MM, van der Coelen D, Bulmer DC, Peeters PJ, Aerssens J, de Hoogt R, Lee K, de Jonge WJ, Boeckxstaens GE. Essential role for TRPV1 in stress-induced (mast cell-dependent) colonic hypersensitivity in maternally separated rats. Neurogastroenterol Motil 2009; 21:1107-e94. [PMID: 19523146 DOI: 10.1111/j.1365-2982.2009.01339.x] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Irritable bowel syndrome is in part characterized by an increased sensitivity to colonic distension. Stress is an important trigger factor for symptom generation. We hypothesized that stress induces visceral hypersensitivity via mast cell degranulation and transient receptor ion channel 1 (TRPV1) modulation. We used the rat model of neonatal maternal separation (MS) to investigate this hypothesis. The visceromotor response to colonic distention was assessed in adult MS and non-handled (NH) rats before and after acute water avoidance (WA) stress. We evaluated the effect of the mast cell stabilizer doxantrazole, neutralizing antiserum against the mast cell mediator nerve growth factor (NGF) and two different TRPV1 antagonists; capsazepine (non-specific) and SB-705498 (TRPV1-specific). Immunohistochemistry was used to assess post-WA TRPV1 expression in dorsal root ganglia and the presence of immunocytes in proximal and distal colon. Retrograde labelled and microdissected dorsal root ganglia sensory neurons were used to evaluate TRPV1 gene transcription. Results showed that acute stress induces colonic hypersensitivity in MS but not in NH rats. Hypersensitivity was prevented by prestress administration of doxantrazole and anti-NGF. Capsazepine inhibited and SB-705498 reversed poststress hypersensitivity. In MS rats, acute stress induced a slight increase in colonic mast cell numbers without further signs of inflammation. Post-WA TRPV1 transcription and expression was not higher in MS than NH rats. In conclusion, the present data on stress-induced visceral hypersensitivity confirm earlier reports on the essential role of mast cells and NGF. Moreover, the results also suggest that TRPV1 modulation (in the absence of overt inflammation) is involved in this response. Thus, mast cells and TRPV1 are potential targets to treat stress-induced visceral hypersensitivity.
Collapse
Affiliation(s)
- R M van den Wijngaard
- Division of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Tan LL, Bornstein JC, Anderson CR. Neurochemical and morphological phenotypes of vagal afferent neurons innervating the adult mouse jejunum. Neurogastroenterol Motil 2009; 21:994-1001. [PMID: 19413682 DOI: 10.1111/j.1365-2982.2009.01322.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Whilst much is known about the function and influence of vagal afferents on the mammalian upper gastrointestinal tract, the phenotypes of the different types of vagal afferent neurons innervating the jejunum is unknown. We have previously shown that spinal afferents supplying the jejunum are predominantly medium-sized sensory neurons that express specific combinations of transient receptor potential vanilloid type 1 (TRPV1), neuronal nitric oxide synthase (NOS) and calcitonin-gene related peptide (CGRP) and that they lack binding for isolectin B4 (IB4). This study aimed to identify the chemical phenotypes and somal sizes of jejunal afferent neurons in the mouse vagal ganglion. Jejunal vagal afferents were identified by retrograde labelling with sub-serosal injections of cholera toxin B (CTB) into the jejunal wall and assessed for IB4-binding, TRPV1-, NOS- and CGRP-immunoreactivities using fluorescent microscopy. Almost all (99%) of CTB-labelled vagal afferent neurons were small- and medium-sized sensory cells. Most (81%) jejunal vagal afferents bound IB4 but fewer (32%) expressed TRPV1. A quarter (25%) of those that bound IB4 co-expressed TRPV1-immunoreactivity whilst 77% of TRPV1-expressing jejunal vagal afferent neurons bound IB4. NOS (0%) and CGRP (0%) expression was absent from all CTB-labelled cells examined. In conclusion, vagal afferents innervating the jejunum differ in their expression of IB4, TRPV1, CGRP and NOS from their spinal counterparts, suggesting that the peripheral endings for extrinsic sensory neurons terminating within the enteric nervous system can be identified selectively.
Collapse
Affiliation(s)
- L L Tan
- Department of Physiology, University of Melbourne, Parkville, VIC, Australia.
| | | | | |
Collapse
|
31
|
Ma X, Mao YK, Wang B, Huizinga JD, Bienenstock J, Kunze W. Lactobacillus reuteri ingestion prevents hyperexcitability of colonic DRG neurons induced by noxious stimuli. Am J Physiol Gastrointest Liver Physiol 2009; 296:G868-75. [PMID: 19179624 DOI: 10.1152/ajpgi.90511.2008] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Lactobacillus species ingestion can decrease autonomic responses and spinal fiber discharge to nociceptive colorectal distension (CRD), even in the absence of inflammation. The present study aimed to determine whether dorsal root ganglion (DRG) somas could be a locus where the antinociceptive probiotic may have an effect. Healthy rats were fed with Lactobacillus reuteri or vehicle control for 9 days whereupon they were anesthetized, and intermittent distal colonic CRD at 80 mmHg distension was either performed for 1 h or not. The animals were immediately euthanized and patch-clamp recordings taken after isolation and overnight culture from those DRG that projected to the distal colon. CRD decreased the threshold for action potential generation and increased the number of spikes discharged during a standard depolarizing test stimulus, and this effect was blocked by prior probiotic ingestion. The increase in excitability was paralleled by an increase in DRG capacitance, which was not altered by Lactobacillus reuteri ingestion. CRD did not increase tissue weight or myeloperoxidase activity. We suggest that the effects of CRD may have been caused by activity-dependent neurotransmission between DRG somas. CRD evoked increases in action potential upstroke speed, which suggests that it may also have led to augmentation of sodium channel conductances. Probiotic ingestion may have interfered with this hypothetical mechanism since it blocked the effect of CRD on the action potential.
Collapse
Affiliation(s)
- Xuelian Ma
- Institute of Physiology, School of Medicine, Shandong University, Shandong, China
| | | | | | | | | | | |
Collapse
|
32
|
Fasanella KE, Christianson JA, Chanthaphavong RS, Davis BM. Distribution and neurochemical identification of pancreatic afferents in the mouse. J Comp Neurol 2008; 509:42-52. [PMID: 18418900 DOI: 10.1002/cne.21736] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Dysfunction of primary afferents innervating the pancreas has been shown to contribute to the development of painful symptoms during acute and chronic pancreatitis. To investigate the distribution and neurochemical phenotype of pancreatic afferents, Alexa Fluor-conjugated cholera toxin B (CTB) was injected into the pancreatic head (CTB-488) and tail (CTB-555) of adult male mice to label neurons retrogradely in both the dorsal root ganglia (DRG) and nodose ganglia (NG). The NG and DRG (T5-T13) were processed for fluorescent immunohistochemistry and visualized by using confocal microscopy. Spinal pancreatic afferents were observed from T5 to T13, with the greatest contribution coming from T9-T12. The pancreatic afferents were equally distributed between right and left spinal ganglia; however, the innervation from the left NG was significantly greater than from the right. For both spinal and vagal afferents there was significantly greater innervation of the pancreatic head relative to the tail. The total number of retrogradely labeled afferents in the nodose was very similar to the total number of DRG afferents. The neurochemical phenotype of DRG neurons was dominated by transient receptor potential vanilloid 1 (TRPV1)-positive neurons (75%), GDNF family receptor alpha-3 (GFRalpha3)-positive neurons (67%), and calcitonin gene-related peptide (CGRP)-positive neurons(65%) neurons. In the NG, TRPV1-, GFRalpha3-, and CGRP-positive neurons constituted only 35%, 1%, and 15% of labeled afferents, respectively. The disparity in peptide and receptor expression between pancreatic afferents in the NG and DRG suggests that even though they contribute a similar number of primary afferents to the pancreas, these two populations may differ in regard to their nociceptive properties and growth factor dependency.
Collapse
Affiliation(s)
- Kenneth E Fasanella
- Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania 15261, USA
| | | | | | | |
Collapse
|
33
|
Grundy D. Ainsley Iggo's early recordings from C fibre vagal afferents supplying the gut. Exp Physiol 2008; 93:1170-3. [PMID: 18945759 DOI: 10.1113/expphysiol.2007.038984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- David Grundy
- Department of Biomedical Science, The University of Sheffield, Western Bank, Sheffield S10 3RA, UK.
| |
Collapse
|
34
|
TRPA1 channels mediate cold temperature sensing in mammalian vagal sensory neurons: pharmacological and genetic evidence. J Neurosci 2008; 28:7863-75. [PMID: 18667618 DOI: 10.1523/jneurosci.1696-08.2008] [Citation(s) in RCA: 124] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Cold thermoreceptors have been described in different territories of the vagus nerve. Application of cold temperature to these visceral afferents can evoke major protective reflexes and thermoregulatory responses. However, virtually nothing is known about the transduction mechanisms underlying cold sensitivity in vagal afferents. Here, we investigated the effects of cold stimulation on intracellular calcium responses and excitability of cultured vagal sensory neurons in the rat nodose ganglion. A large fraction of vagal neurons were activated by cold, with a mean threshold of approximately 24 degrees C. Cooling was accompanied by development of a small inward current and the firing of action potentials. Most cold-sensitive neurons were also activated by heat and capsaicin, suggesting a nociceptive function. The pharmacological response to TRPM8 and TRPA1 agonists and antagonists suggested that, unlike results observed in somatic tissues, TRPA1 is the major mediator of cold-evoked responses in vagal visceral neurons. Thus, most cold-evoked responses were potentiated by cinnamaldehyde, menthol, icilin, and BCTC [4-(3-chloro-pyridin-2-yl)-piperazine-1-carboxylic acid (4-tert-butyl-phenyl)-amide], agonists of TRPA1, and were inhibited by ruthenium red, camphor, and HC03001 [2-(1,3-dimethyl-2,6-dioxo-1,2,3,6-tetrahydro-7H-purin-7-yl)-N-(4-isopropylphenyl)acetamide]. Results in mouse nodose neurons revealed a similar pharmacological profile of cold-evoked responses. Furthermore, experiments in TRPA1 knock-out mice showed a large reduction in the percentage of cold-sensitive neurons compared with wild-type animals. Together, these results support an important role of TRPA1 channels in visceral thermosensation and indicate major differences in the transduction of temperature signals between somatic and visceral sensory neurons.
Collapse
|
35
|
Nassenstein C, Kwong K, Taylor-Clark T, Kollarik M, Macglashan DM, Braun A, Undem BJ. Expression and function of the ion channel TRPA1 in vagal afferent nerves innervating mouse lungs. J Physiol 2008; 586:1595-604. [PMID: 18218683 DOI: 10.1113/jphysiol.2007.148379] [Citation(s) in RCA: 236] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Transient receptor potential (TRP) A1 and TRPM8 are ion channels that have been localized to afferent nociceptive nerves. These TRP channels may be of particular relevance to respiratory nociceptors in that they can be activated by various inhaled irritants and/or cold air. We addressed the hypothesis that mouse vagal sensory nerves projecting to the airways express TRPA1 and TRPM8 and that they can be activated via these receptors. Single cell RT-PCR analysis revealed that TRPA1 mRNA, but not TRPM8, is uniformly expressed in lung-labelled TRPV1-expressing vagal sensory neurons. Neither TRPA1 nor TRPM8 mRNA was expressed in TRPV1-negative neurons. Capsaicin-sensitive, but not capsaicin-insensitive, lung-specific neurons responded to cinnamaldehyde, a TRPA1 agonist, with increases in intracellular calcium. Menthol, a TRPM8 agonist, was ineffective at increasing cellular calcium in lung-specific vagal sensory neurons. Cinnamaldehyde also induced TRPA1-like inward currents (as measured by means of whole cell patch clamp recordings) in capsaicin-sensitive neurons. In an ex vivo vagal innervated mouse lung preparation, cinnamaldehyde evoked action potential discharge in mouse vagal C-fibres with a peak frequency similar to that observed with capsaicin. Cinnamaldehyde inhalation in vivo mimicked capsaicin in eliciting strong central-reflex changes in breathing pattern. Taken together, our results support the hypothesis that TRPA1, but not TRPM8, is expressed in vagal sensory nerves innervating the airways. TRPA1 activation provides a mechanism by which certain environmental stimuli may elicit action potential discharge in airway afferent C-fibres and the consequent nocifensor reflexes.
Collapse
Affiliation(s)
- Christina Nassenstein
- Johns Hopkins Allergy and Asthma Center 5501 Hopkins Bayview Circle Baltimore, MD 21224, USA
| | | | | | | | | | | | | |
Collapse
|
36
|
Oort PJ, Warden CH, Baumann TK, Knotts TA, Adams SH. Characterization of Tusc5, an adipocyte gene co-expressed in peripheral neurons. Mol Cell Endocrinol 2007; 276:24-35. [PMID: 17689857 DOI: 10.1016/j.mce.2007.06.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2007] [Accepted: 06/22/2007] [Indexed: 12/25/2022]
Abstract
Tumor suppressor candidate 5 (Tusc5, also termed brain endothelial cell derived gene-1 or BEC-1), a CD225 domain-containing, cold-repressed gene identified during brown adipose tissue (BAT) transcriptome analyses was found to be robustly-expressed in mouse white adipose tissue (WAT) and BAT, with similarly high expression in human adipocytes. Tusc5 mRNA was markedly increased from trace levels in pre-adipocytes to significant levels in developing 3T3-L1 adipocytes, coincident with several mature adipocyte markers (phosphoenolpyruvate carboxykinase 1, GLUT4, adipsin, leptin). The Tusc5 transcript levels were increased by the peroxisome proliferator activated receptor-gamma (PPARgamma) agonist GW1929 (1microg/mL, 18h) by >10-fold (pre-adipocytes) to approximately 1.5-fold (mature adipocytes) versus controls (p<0.0001). Taken together, these results suggest an important role for Tusc5 in maturing adipocytes. Intriguingly, we discovered robust co-expression of the gene in peripheral nerves (primary somatosensory neurons). In light of the marked repression of the gene observed after cold exposure, these findings may point to participation of Tusc5 in shared adipose-nervous system functions linking environmental cues, CNS signals, and WAT-BAT physiology. Characterization of such links is important for clarifying the molecular basis for adipocyte proliferation and could have implications for understanding the biology of metabolic disease-related neuropathies.
Collapse
Affiliation(s)
- Pieter J Oort
- USDA/Agricultural Research Service Western Human Nutrition Research Center, University of California, Davis, CA 95616, USA
| | | | | | | | | |
Collapse
|
37
|
Aerssens J, Hillsley K, Peeters PJ, de Hoogt R, Stanisz A, Lin JH, Van den Wyngaert I, Göhlmann HW, Grundy D, Stead RH, Coulie B. Alterations in the brain-gut axis underlying visceral chemosensitivity in Nippostrongylus brasiliensis-infected mice. Gastroenterology 2007; 132:1375-87. [PMID: 17408648 DOI: 10.1053/j.gastro.2007.02.019] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2006] [Accepted: 01/04/2007] [Indexed: 12/18/2022]
Abstract
BACKGROUND & AIMS Visceral hypersensitivity, a hallmark of irritable bowel syndrome, is generally considered to be mechanosensitive in nature and mediated via spinal afferents. Both stress and inflammation are implicated in visceral hypersensitivity, but the underlying molecular mechanisms of visceral hypersensitivity are unknown. METHODS Mice were infected with Nippostrongylus brasiliensis (Nb) larvae, exposed to environmental stress and the following separate studies performed 3-4 weeks later. Mesenteric afferent nerve activity was recorded in response to either ramp balloon distention (60 mm Hg), or to an intraluminal perfusion of hydrochloric acid (50 mmol/L), or to octreotide administration (2 micromol/L). Intraperitoneal injection of cholera toxin B-488 identified neurons projecting to the abdominal viscera. Fluorescent neurons in dorsal root and nodose ganglia were isolated using laser-capture microdissection. RNA was hybridized to Affymetrix Mouse whole genome arrays for analysis to evaluate the effects of stress and infection. RESULTS In mice previously infected with Nb, there was no change in intestinal afferent mechanosensitivity, but there was an increase in chemosensitive responses to intraluminal hydrochloric acid when compared with control animals. Gene expression profiles in vagal but not spinal visceral sensory neurons were significantly altered in stressed Nb-infected mice. Decreased afferent responses to somatostatin receptor 2 stimulation correlated with lower expression of vagal somatostatin receptor 2 in stressed Nb-infected mice, confirming a link between molecular data and functional sequelae. CONCLUSIONS Alterations in the intestinal brain-gut axis, in chemosensitivity but not mechanosensitivity, and through vagal rather than spinal pathways, are implicated in stress-induced postinflammatory visceral hypersensitivity.
Collapse
Affiliation(s)
- Jeroen Aerssens
- Johnson & Johnson Pharmaceutical Research and Development, Beerse, Belgium
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
PURPOSE OF REVIEW Enteric neurobiology is a rapidly advancing field of investigation providing insight into the way in which diverse gastrointestinal functions are controlled, coordinated and integrated with central mechanisms important for food intake regulation, illness behaviour and sensory mechanisms. Our aim was to highlight recent advances. RECENT FINDINGS With such a large number of studies to choose from and given our emphasis in previous years on developmental aspects, sensory transmission, and neuro-immune interactions, we have focused on two themes. One reflecting the current interest in the way the enteric nervous system is altered in disease and the second covering the enormous interest in the contribution of enteric mechanisms to the control of energy balance. SUMMARY The new basic science information gathered during the past year provides insight into pathophysiological processes and will pave the way for improved understanding of both organic and 'functional' gastrointestinal disorders.
Collapse
Affiliation(s)
- David Grundy
- Department of Biomedical Science, University of Sheffield, Western Bank, Sheffield, UK.
| | | |
Collapse
|
39
|
Ploski JE, Newton SS, Duman RS. Electroconvulsive seizure-induced gene expression profile of the hippocampus dentate gyrus granule cell layer. J Neurochem 2007; 99:1122-32. [PMID: 17081142 DOI: 10.1111/j.1471-4159.2006.04156.x] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Electroconvulsive shock (ECS) is the most effective treatment for depression, but the mechanism underlying the therapeutic action of this treatment is still unknown. To better understand the molecular changes that may be necessary for the clinical effectiveness of ECS we have combined the technologies of gene expression profiling using cDNA microarrays with T7-based RNA amplification and laser microdissection to identify regulated genes in the dentate gyrus granule cell layer of the hippocampus. We have identified genes previously reported to be up-regulated following ECS, including brain-derived neurotrophic factor, neuropeptide Y, and thyrotrophin releasing hormone, as well as several novel genes. Notably, we have identified additional genes that are known to be involved in neuroprotection, such as growth arrest DNA damage inducible beta (Gadd45beta), and the excitatory amino acid transporter-1 (EAAC1/Slc1A1). In addition, via in situ hybridization we show that EAAC1 is specifically up-regulated in the dentate gyrus, but not in other hippocampal subfields. This study demonstrates the utility of microarray analysis of microdissected subregions of limbic brain regions and identifies novel ECS-regulated genes.
Collapse
Affiliation(s)
- Jonathan E Ploski
- Laboratory of Molecular Psychiatry, Departments of Psychiatry and Pharmacology, Yale University School of Medicine, New Haven, Connecticut 06508, USA
| | | | | |
Collapse
|
40
|
Abstract
The gastrointestinal (GI) tract must balance ostensibly opposite functions. On the one hand, it must undertake the process of digestion and absorption of nutrients. At the same time, the GI tract must protect itself from potential harmful antigenic and pathogenic material. Central to these processes is the ability to 'sense' the mechanical and chemical environment in the gut wall and lumen in order to orchestrate the appropriate response that facilitates nutrient assimilation or the rapid expulsion through diarrhoea and/or vomiting. In this respect, the GI tract is richly endowed with sensory elements that monitor the gut environment. Enteric neurones provide one source of such sensory innervation and are responsible for the ability of the decentralized gut to perform complex reflex functions. Extrinsic afferents not only contribute to this reflex control, but also contribute to homeostatic mechanisms and can give rise to sensations, under certain circumstances. The enteric and extrinsic sensory mechanisms share a number of common features but also some remarkably different properties. The purpose of this review is to summarize current views on sensory processing within both the enteric and extrinsic innervation and to specifically address the pharmacology of nociceptive extrinsic sensory pathways.
Collapse
Affiliation(s)
- L A Blackshaw
- Nerve Gut Research Laboratory, Hanson Institute, Royal Adelaide Hospital, University of Adelaide, Adelaide, SA, Australia
| | | | | | | |
Collapse
|
41
|
Hillsley K, Lin JH, Stanisz A, Grundy D, Aerssens J, Peeters PJ, Moechars D, Coulie B, Stead RH. Dissecting the role of sodium currents in visceral sensory neurons in a model of chronic hyperexcitability using Nav1.8 and Nav1.9 null mice. J Physiol 2006; 576:257-67. [PMID: 16857712 PMCID: PMC1995629 DOI: 10.1113/jphysiol.2006.113597] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Tetrodotoxin-resistant (TTX-R) sodium currents have been proposed to underlie sensory neuronal hyperexcitability in acute inflammatory models, but their role in chronic models is unknown. Since no pharmacological tools to separate TTX-R currents are available, this study employs Na(v)1.8 and Na(v)1.9 null mice to evaluate these currents roles in a chronic hyperexcitability model after the resolution of an inflammatory insult. Transient jejunitis was induced by infection with Nippostrongylus brasiliensis (Nb) in Na(v)1.9 and Na(v)1.8 null, wild-type and naïve mice. Retrogradely labelled dorsal root ganglia (DRG) neurons were harvested on day 20-24 post-infection for patch clamp recording. Rheobase and action potential (AP) parameters were recorded as measures of excitability, and Na(v)1.9 and Na(v)1.8 currents were recorded. DRG neuronal excitability was significantly increased in post-infected mice compared to sham animals, despite the absence of ongoing inflammation (sham = 1.9 +/- 0.3, infected = 3.6 +/- 0.7 APs at 2x rheobase, P = 0.02). Hyperexcitability was associated with a significantly increased amplitude of TTX-R currents. Hyperexcitability was maintained in Na(v)1.9(-/-) mice, but hyperexcitability was absent and APs were blunted in Na(v)1.8(-/-) mice. This study identifies a critical role for Na(v)1.8 in chronic post-infectious visceral hyperexcitability, with no contribution from Na(v)1.9. Nb infection-induced hyperexcitability is not observed in Na(v)1.8(-/-) mice, but is still present in Na(v)1.9(-/-) mice. It is not clear whether hyperexcitability is due to a change in the function of Na(v)1.8 channels or a change in the number of Na(v)1.8 channels.
Collapse
MESH Headings
- Action Potentials/drug effects
- Action Potentials/physiology
- Amino Acid Sequence
- Anesthetics, Local/pharmacology
- Animals
- Cells, Cultured
- Electrophysiology
- Ganglia, Spinal/metabolism
- Ganglia, Spinal/pathology
- Ganglia, Spinal/physiology
- Ganglia, Spinal/physiopathology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Molecular Sequence Data
- NAV1.8 Voltage-Gated Sodium Channel
- NAV1.9 Voltage-Gated Sodium Channel
- Neurons, Afferent/metabolism
- Neurons, Afferent/pathology
- Neurons, Afferent/physiology
- Neuropeptides/analysis
- Neuropeptides/drug effects
- Neuropeptides/genetics
- Neuropeptides/physiology
- Nippostrongylus
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Sodium Channels/analysis
- Sodium Channels/drug effects
- Sodium Channels/genetics
- Sodium Channels/physiology
- Strongylida Infections/pathology
- Strongylida Infections/physiopathology
- Tetrodotoxin/pharmacology
- Viscera/innervation
Collapse
Affiliation(s)
- Kirk Hillsley
- Holburn Group, 1100 Bennett Road, Bowmanville, Canada ON L1C 3K5
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Brumovsky P, Villar MJ, Hökfelt T. Tyrosine hydroxylase is expressed in a subpopulation of small dorsal root ganglion neurons in the adult mouse. Exp Neurol 2006; 200:153-65. [PMID: 16516890 DOI: 10.1016/j.expneurol.2006.01.023] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2005] [Revised: 01/05/2006] [Accepted: 01/24/2006] [Indexed: 11/17/2022]
Abstract
The expression of tyrosine hydroxylase (TH) was studied in adult mouse dorsal root ganglia (DRGs) and spinal cord by means of immunohistochemistry and in situ hybridization. TH immunoreactivity and TH mRNA were present in 10-15% of lumbar DRG neurons, in most cases being small/medium-sized. Only very few of these neurons coexpressed calcitonin gene-related peptide (CGRP), and only around 6% bound isolectin B4 (IB4). Dopamine beta-hydroxylase-positive(+) or aromatic amino acid decarboxylase (AADC)+ DRG neurons were rare and did not colocalize TH. No evidence for dopamine transporter expression was obtained. Axotomy of the sciatic nerve only showed a tendency towards reduction in the number of TH+ neurons. In the dorsal horn of the spinal cord, moderately dense and widespread TH+ nerve terminals were observed, mainly in the gray matter and they did not show a typical primary afferent pattern. Also, dorsal rhizotomy or peripheral axotomy had no apparent effect on TH-LI in the dorsal horn. In the skin, along with an abundant TH+ innervation of blood vessels and sweat gland acini, a number of fibers was observed in close relation to the skin surface, some even penetrating into the epithelium. These results demonstrate presence, in normal adult mouse DRGs, of a subpopulation of TH+, essentially CGRP- and IB4-negative small/medium-sized neurons. No evidence for transport of TH into central afferents was obtained, but the enzyme may be present in some sensory fibers in the skin. The fact that neither AADC nor the dopamine transporter could be visualized suggests of non-dopaminergic transmitter phenotype, but the levels of these two dopaminergic markers may be too low to be detected with the present methodology. A further alternative is that L-DOPA after release is extracellularly converted to dopamine.
Collapse
Affiliation(s)
- Pablo Brumovsky
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | | | | |
Collapse
|