101
|
Borgmann D, Ciglieri E, Biglari N, Brandt C, Cremer AL, Backes H, Tittgemeyer M, Wunderlich FT, Brüning JC, Fenselau H. Gut-brain communication by distinct sensory neurons differently controls feeding and glucose metabolism. Cell Metab 2021; 33:1466-1482.e7. [PMID: 34043943 PMCID: PMC8280952 DOI: 10.1016/j.cmet.2021.05.002] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 02/23/2021] [Accepted: 05/03/2021] [Indexed: 01/26/2023]
Abstract
Sensory neurons relay gut-derived signals to the brain, yet the molecular and functional organization of distinct populations remains unclear. Here, we employed intersectional genetic manipulations to probe the feeding and glucoregulatory function of distinct sensory neurons. We reconstruct the gut innervation patterns of numerous molecularly defined vagal and spinal afferents and identify their downstream brain targets. Bidirectional chemogenetic manipulations, coupled with behavioral and circuit mapping analysis, demonstrated that gut-innervating, glucagon-like peptide 1 receptor (GLP1R)-expressing vagal afferents relay anorexigenic signals to parabrachial nucleus neurons that control meal termination. Moreover, GLP1R vagal afferent activation improves glucose tolerance, and their inhibition elevates blood glucose levels independent of food intake. In contrast, gut-innervating, GPR65-expressing vagal afferent stimulation increases hepatic glucose production and activates parabrachial neurons that control normoglycemia, but they are dispensable for feeding regulation. Thus, distinct gut-innervating sensory neurons differentially control feeding and glucoregulatory neurocircuits and may provide specific targets for metabolic control.
Collapse
Affiliation(s)
- Diba Borgmann
- Synaptic Transmission in Energy Homeostasis Group, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany; Translational Neurocircuitry Group, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany; Center for Anatomy II, Neuroanatomy, University Hospital Cologne, Joseph-Stelzmann Str. 9, 50937 Cologne, Germany
| | - Elisa Ciglieri
- Synaptic Transmission in Energy Homeostasis Group, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Strasse 26, 50937 Cologne, Germany
| | - Nasim Biglari
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Strasse 50, 50931 Cologne, Germany
| | - Claus Brandt
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Strasse 50, 50931 Cologne, Germany
| | - Anna Lena Cremer
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Strasse 50, 50931 Cologne, Germany
| | - Heiko Backes
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Strasse 50, 50931 Cologne, Germany
| | - Marc Tittgemeyer
- Translational Neurocircuitry Group, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Straße 26, Cologne 50931, Germany
| | - F Thomas Wunderlich
- Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Strasse 26, 50937 Cologne, Germany; Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Strasse 50, 50931 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Straße 26, Cologne 50931, Germany; Center of Molecular Medicine Cologne (CMMC), University of Cologne, Robert-Koch-Straße 21, 50931 Cologne, Germany
| | - Jens C Brüning
- Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Strasse 26, 50937 Cologne, Germany; Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Strasse 50, 50931 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Straße 26, Cologne 50931, Germany; Center of Molecular Medicine Cologne (CMMC), University of Cologne, Robert-Koch-Straße 21, 50931 Cologne, Germany
| | - Henning Fenselau
- Synaptic Transmission in Energy Homeostasis Group, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Strasse 26, 50937 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Straße 26, Cologne 50931, Germany.
| |
Collapse
|
102
|
Barrantes FJ. The unfolding palette of COVID-19 multisystemic syndrome and its neurological manifestations. Brain Behav Immun Health 2021; 14:100251. [PMID: 33842898 PMCID: PMC8019247 DOI: 10.1016/j.bbih.2021.100251] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/23/2021] [Accepted: 03/28/2021] [Indexed: 02/07/2023] Open
Abstract
Although our current knowledge of the pathophysiology of COVID-19 is still fragmentary, the information so far accrued on the tropism and life cycle of its etiological agent SARS-CoV-2, together with the emerging clinical data, suffice to indicate that the severe acute pulmonary syndrome is the main, but not the only manifestation of COVID-19. Necropsy studies are increasingly revealing underlying endothelial vasculopathies in the form of micro-haemorrhages and micro-thrombi. Intertwined with defective antiviral responses, dysregulated coagulation mechanisms, abnormal hyper-inflammatory reactions and responses, COVID-19 is disclosing a wide pathophysiological palette. An additional property in categorising the disease is the combination of tissue (e.g. neuro- and vasculo-tropism) with organ tropism, whereby the virus preferentially attacks certain organs with highly developed capillary beds, such as the lungs, gastrointestinal tract, kidney and brain. These multiple clinical presentations confirm that the acute respiratory syndrome as described initially is increasingly unfolding as a more complex nosological entity, a multiorgan syndrome of systemic breadth. The neurological manifestations of COVID-19, the focus of this review, reflect this manifold nature of the disease.
Collapse
Affiliation(s)
- Francisco J. Barrantes
- Institute of Biomedical Research (BIOMED), UCA-CONICET, Av. Alicia Moreau de Justo 1600, C1107AFF, Buenos Aires, Argentina
| |
Collapse
|
103
|
Abstract
Mechanosensation is the ability to detect dynamic mechanical stimuli (e.g., pressure, stretch, and shear stress) and is essential for a wide variety of processes, including our sense of touch on the skin. How touch is detected and transduced at the molecular level has proved to be one of the great mysteries of sensory biology. A major breakthrough occurred in 2010 with the discovery of a family of mechanically gated ion channels that were coined PIEZOs. The last 10 years of investigation have provided a wealth of information about the functional roles and mechanisms of these molecules. Here we focus on PIEZO2, one of the two PIEZO proteins found in humans and other mammals. We review how work at the molecular, cellular, and systems levels over the past decade has transformed our understanding of touch and led to unexpected insights into other types of mechanosensation beyond the skin.
Collapse
Affiliation(s)
- Marcin Szczot
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, Maryland 20892, USA; .,Center for Social and Affective Neuroscience, Department of Clinical and Experimental Medicine, Linköping University, 583 30 Linköping, Sweden
| | - Alec R Nickolls
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, Maryland 20892, USA;
| | - Ruby M Lam
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, Maryland 20892, USA; .,NIH-Brown University Graduate Program in Neuroscience, Providence, Rhode Island 02912, USA
| | - Alexander T Chesler
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, Maryland 20892, USA; .,National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
104
|
Hurtado-Lorenzo A, Honig G, Weaver SA, Larkin PB, Heller C. Chronic Abdominal Pain in IBD Research Initiative: Unraveling Biological Mechanisms and Patient Heterogeneity to Personalize Treatment and Improve Clinical Outcomes. CROHN'S & COLITIS 360 2021; 3:otab034. [PMID: 36776666 PMCID: PMC9802354 DOI: 10.1093/crocol/otab034] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Andrés Hurtado-Lorenzo
- Research Department, Crohn’s & Colitis Foundation, New York, New York, USA,Address correspondence to: Andrés Hurtado-Lorenzo, PhD, Crohn’s & Colitis Foundation, 733 3rd Ave Suite 510, New York, NY 10017, USA ()
| | - Gerard Honig
- Research Department, Crohn’s & Colitis Foundation, New York, New York, USA
| | | | - Paul B Larkin
- Research Department, Crohn’s & Colitis Foundation, New York, New York, USA
| | - Caren Heller
- Research Department, Crohn’s & Colitis Foundation, New York, New York, USA
| |
Collapse
|
105
|
Mwirigi J, Kume M, Hassler SN, Ahmad A, Ray PR, Jiang C, Chamessian A, Mseeh N, Ludwig BP, Rivera BD, Nieman MT, Van de Ven T, Ji RR, Dussor G, Boitano S, Vagner J, Price TJ. A Role for Protease Activated Receptor Type 3 (PAR3) in Nociception Demonstrated Through Development of a Novel Peptide Agonist. THE JOURNAL OF PAIN 2021; 22:692-706. [PMID: 33429107 PMCID: PMC8197731 DOI: 10.1016/j.jpain.2020.12.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 12/02/2020] [Accepted: 12/20/2020] [Indexed: 02/07/2023]
Abstract
The protease activated receptor (PAR) family is a group of G-protein coupled receptors (GPCRs) activated by proteolytic cleavage of the extracellular domain. PARs are expressed in a variety of cell types with crucial roles in homeostasis, immune responses, inflammation, and pain. PAR3 is the least researched of the four PARs, with little known about its expression and function. We sought to better understand its potential function in the peripheral sensory nervous system. Mouse single-cell RNA sequencing data demonstrates that PAR3 is widely expressed in dorsal root ganglion (DRG) neurons. Co-expression of PAR3 mRNA with other PARs was identified in various DRG neuron subpopulations, consistent with its proposed role as a coreceptor of other PARs. We developed a lipid tethered PAR3 agonist, C660, that selectively activates PAR3 by eliciting a Ca2+ response in DRG and trigeminal neurons. In vivo, C660 induces mechanical hypersensitivity and facial grimacing in WT but not PAR3-/- mice. We characterized other nociceptive phenotypes in PAR3-/- mice and found a loss of hyperalgesic priming in response to IL-6, carrageenan, and a PAR2 agonist, suggesting that PAR3 contributes to long-lasting nociceptor plasticity in some contexts. To examine the potential role of PAR3 in regulating the activity of other PARs in sensory neurons, we administered PAR1, PAR2, and PAR4 agonists and assessed mechanical and affective pain behaviors in WT and PAR3-/- mice. We observed that the nociceptive effects of PAR1 agonists were potentiated in the absence of PAR3. Our findings suggest a complex role of PAR3 in the physiology and plasticity of nociceptors. PERSPECTIVE: We evaluated the role of PAR3, a G-protein coupled receptor, in nociception by developing a selective peptide agonist. Our findings suggest that PAR3 contributes to nociception in various contexts and plays a role in modulating the activity of other PARs.
Collapse
Affiliation(s)
- Juliet Mwirigi
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Richardson, Texas
| | - Moeno Kume
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Richardson, Texas
| | - Shayne N Hassler
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Richardson, Texas
| | - Ayesha Ahmad
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Richardson, Texas
| | - Pradipta R Ray
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Richardson, Texas
| | - Changyu Jiang
- Duke University School of Medicine, Department of Anesthesiology, Pharmacology, and Cancer Biology, Durham, North Carolina
| | - Alexander Chamessian
- Duke University School of Medicine, Department of Anesthesiology, Pharmacology, and Cancer Biology, Durham, North Carolina
| | - Nakleh Mseeh
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Richardson, Texas
| | - Breya P Ludwig
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Richardson, Texas
| | - Benjamin D Rivera
- Department of Physiology, University of Arizona, Asthma and Airway Disease Research Center, Tucson, Arizona
| | - Marvin T Nieman
- Case Western Reserve University School of Medicine, Department of Pharmacology, Cleveland, Ohio
| | - Thomas Van de Ven
- Duke University School of Medicine, Department of Anesthesiology, Pharmacology, and Cancer Biology, Durham, North Carolina
| | - Ru-Rong Ji
- Duke University School of Medicine, Department of Anesthesiology, Pharmacology, and Cancer Biology, Durham, North Carolina
| | - Gregory Dussor
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Richardson, Texas
| | - Scott Boitano
- Department of Physiology, University of Arizona, Asthma and Airway Disease Research Center, Tucson, Arizona
| | - Josef Vagner
- University of Arizona, Bio5 Research Institute, Tucson, Arizona
| | - Theodore J Price
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Richardson, Texas.
| |
Collapse
|
106
|
|
107
|
Lee MC, Nahorski MS, Hockley JRF, Lu VB, Ison G, Pattison LA, Callejo G, Stouffer K, Fletcher E, Brown C, Drissi I, Wheeler D, Ernfors P, Menon D, Reimann F, Smith ESJ, Woods CG. Human Labor Pain Is Influenced by the Voltage-Gated Potassium Channel K V6.4 Subunit. Cell Rep 2021; 32:107941. [PMID: 32697988 PMCID: PMC7383234 DOI: 10.1016/j.celrep.2020.107941] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 03/19/2020] [Accepted: 06/30/2020] [Indexed: 12/22/2022] Open
Abstract
By studying healthy women who do not request analgesia during their first delivery, we investigate genetic effects on labor pain. Such women have normal sensory and psychometric test results, except for significantly higher cuff pressure pain. We find an excess of heterozygotes carrying the rare allele of SNP rs140124801 in KCNG4. The rare variant KV6.4-Met419 has a dominant-negative effect and cannot modulate the voltage dependence of KV2.1 inactivation because it fails to traffic to the plasma membrane. In vivo, Kcng4 (KV6.4) expression occurs in 40% of retrograde-labeled mouse uterine sensory neurons, all of which express KV2.1, and over 90% express the nociceptor genes Trpv1 and Scn10a. In neurons overexpressing KV6.4-Met419, the voltage dependence of inactivation for KV2.1 is more depolarized compared with neurons overexpressing KV6.4. Finally, KV6.4-Met419-overexpressing neurons have a higher action potential threshold. We conclude that KV6.4 can influence human labor pain by modulating the excitability of uterine nociceptors. KCNG4 variant highly prevalent in women requiring no analgesia in childbirth KCNG4 variant encodes KV6.4Met-419; KV6.4 is a silent subunit modifying KV activity KV6.4Met-419 is retained in the cytoplasm and acts in a dominant-negative manner KV6.4Met-419 overexpression results in hypoexcitable sensory neurons
Collapse
Affiliation(s)
- Michael C Lee
- University Division of Anaesthesia, University of Cambridge, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK.
| | - Michael S Nahorski
- Cambridge Institute for Medical Research, Wellcome Trust MRC Building, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK
| | - James R F Hockley
- Department of Pharmacology, Tennis Court Road, Cambridge CB2 1PD, UK
| | - Van B Lu
- Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK
| | - Gillian Ison
- University Division of Anaesthesia, University of Cambridge, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK
| | - Luke A Pattison
- Department of Pharmacology, Tennis Court Road, Cambridge CB2 1PD, UK
| | - Gerard Callejo
- Department of Pharmacology, Tennis Court Road, Cambridge CB2 1PD, UK
| | - Kaitlin Stouffer
- Cambridge Institute for Medical Research, Wellcome Trust MRC Building, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK
| | - Emily Fletcher
- Cambridge Institute for Medical Research, Wellcome Trust MRC Building, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK
| | - Christopher Brown
- Department of Psychological Sciences, Institute of Psychology, Health and Society, University of Liverpool, Liverpool L69 7ZA, UK
| | - Ichrak Drissi
- University Division of Anaesthesia, University of Cambridge, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK
| | - Daniel Wheeler
- University Division of Anaesthesia, University of Cambridge, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK
| | - Patrik Ernfors
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - David Menon
- University Division of Anaesthesia, University of Cambridge, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK
| | - Frank Reimann
- Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK.
| | | | - C Geoffrey Woods
- Cambridge Institute for Medical Research, Wellcome Trust MRC Building, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK.
| |
Collapse
|
108
|
|
109
|
Wangzhou A, Paige C, Neerukonda SV, Naik DK, Kume M, David ET, Dussor G, Ray PR, Price TJ. A ligand-receptor interactome platform for discovery of pain mechanisms and therapeutic targets. Sci Signal 2021; 14:14/674/eabe1648. [PMID: 33727337 DOI: 10.1126/scisignal.abe1648] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In the peripheral nervous system, ligand-receptor interactions between cells and neurons shape sensory experience, including pain. We set out to identify the potential interactions between sensory neurons and peripheral cell types implicated in disease-associated pain. Using mouse and human RNA sequencing datasets and computational analysis, we created interactome maps between dorsal root ganglion (DRG) sensory neurons and an array of normal cell types, as well as colitis-associated glial cells, rheumatoid arthritis-associated synovial macrophages, and pancreatic tumor tissue. These maps revealed a common correlation between the abundance of heparin-binding EGF-like growth factor (HBEGF) in peripheral cells with that of its receptor EGFR (a member of the ErbB family of receptors) in DRG neurons. Subsequently, we confirmed that increased abundance of HBEGF enhanced nociception in mice, likely acting on DRG neurons through ErbB family receptors. Collectively, these interactomes highlight ligand-receptor interactions that may lead to treatments for disease-associated pain and, furthermore, reflect the complexity of cell-to-neuron signaling in chronic pain states.
Collapse
Affiliation(s)
- Andi Wangzhou
- School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, University of Texas at Dallas, 800 W Campbell Rd., Richardson, TX 75080, USA
| | - Candler Paige
- School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, University of Texas at Dallas, 800 W Campbell Rd., Richardson, TX 75080, USA
| | - Sanjay V Neerukonda
- School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, University of Texas at Dallas, 800 W Campbell Rd., Richardson, TX 75080, USA
| | - Dhananjay K Naik
- School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, University of Texas at Dallas, 800 W Campbell Rd., Richardson, TX 75080, USA
| | - Moeno Kume
- School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, University of Texas at Dallas, 800 W Campbell Rd., Richardson, TX 75080, USA
| | - Eric T David
- School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, University of Texas at Dallas, 800 W Campbell Rd., Richardson, TX 75080, USA
| | - Gregory Dussor
- School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, University of Texas at Dallas, 800 W Campbell Rd., Richardson, TX 75080, USA
| | - Pradipta R Ray
- School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, University of Texas at Dallas, 800 W Campbell Rd., Richardson, TX 75080, USA.
| | - Theodore J Price
- School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, University of Texas at Dallas, 800 W Campbell Rd., Richardson, TX 75080, USA.
| |
Collapse
|
110
|
Smith-Edwards KM, Edwards BS, Wright CM, Schneider S, Meerschaert KA, Ejoh LL, Najjar SA, Howard MJ, Albers KM, Heuckeroth RO, Davis BM. Sympathetic Input to Multiple Cell Types in Mouse and Human Colon Produces Region-Specific Responses. Gastroenterology 2021; 160:1208-1223.e4. [PMID: 32980343 PMCID: PMC7956113 DOI: 10.1053/j.gastro.2020.09.030] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 07/18/2020] [Accepted: 09/15/2020] [Indexed: 01/16/2023]
Abstract
BACKGROUND & AIMS The colon is innervated by intrinsic and extrinsic neurons that coordinate functions necessary for digestive health. Sympathetic input suppresses colon motility by acting on intrinsic myenteric neurons, but the extent of sympathetic-induced changes on large-scale network activity in myenteric circuits has not been determined. Compounding the complexity of sympathetic function, there is evidence that sympathetic transmitters can regulate activity in non-neuronal cells (such as enteric glia and innate immune cells). METHODS We performed anatomical tracing, immunohistochemistry, optogenetic (GCaMP calcium imaging, channelrhodopsin), and colon motility studies in mice and single-cell RNA sequencing in human colon to investigate how sympathetic postganglionic neurons modulate colon function. RESULTS Individual neurons in each sympathetic prevertebral ganglion innervated the proximal or distal colon, with processes closely opposed to multiple cell types. Calcium imaging in semi-intact mouse colon preparations revealed changes in spontaneous and evoked neural activity, as well as activation of non-neuronal cells, induced by sympathetic nerve stimulation. The overall pattern of response to sympathetic stimulation was unique to the proximal or distal colon. Region-specific changes in cellular activity correlated with motility patterns produced by electrical and optogenetic stimulation of sympathetic pathways. Pharmacology experiments (mouse) and RNA sequencing (human) indicated that appropriate receptors were expressed on different cell types to account for the responses to sympathetic stimulation. Regional differences in expression of α-1 adrenoceptors in human colon emphasize the translational relevance of our mouse findings. CONCLUSIONS Sympathetic neurons differentially regulate activity of neurons and non-neuronal cells in proximal and distal colon to promote distinct changes in motility patterns, likely reflecting the distinct roles played by these 2 regions.
Collapse
Affiliation(s)
- Kristen M. Smith-Edwards
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania,Pittsburgh Center for Pain Research, University of Pittsburgh, Pennsylvania,Center for Neuroscience at the University of Pittsburgh, Pittsburgh, Pennsylvania,For correspondence: Kristen M. Smith-Edwards, University of Pittsburgh, Department of Neurobiology, 200 Lothrop Street, Pittsburgh, PA 15216, , Ph: 412-648-9745
| | - Brian S. Edwards
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania,Pittsburgh Center for Pain Research, University of Pittsburgh, Pennsylvania,Center for Neuroscience at the University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Christina M. Wright
- The Children’s Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania
| | - Sabine Schneider
- The Children’s Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania
| | - Kimberly A. Meerschaert
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania,Pittsburgh Center for Pain Research, University of Pittsburgh, Pennsylvania,Center for Neuroscience at the University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Lindsay L. Ejoh
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Sarah A. Najjar
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania,Pittsburgh Center for Pain Research, University of Pittsburgh, Pennsylvania,Center for Neuroscience at the University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | - Kathryn M. Albers
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania,Pittsburgh Center for Pain Research, University of Pittsburgh, Pennsylvania,Center for Neuroscience at the University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Robert O. Heuckeroth
- The Children’s Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania
| | - Brian M. Davis
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania,Pittsburgh Center for Pain Research, University of Pittsburgh, Pennsylvania,Center for Neuroscience at the University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
111
|
Vanneste M, Segal A, Voets T, Everaerts W. Transient receptor potential channels in sensory mechanisms of the lower urinary tract. Nat Rev Urol 2021; 18:139-159. [PMID: 33536636 DOI: 10.1038/s41585-021-00428-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/08/2021] [Indexed: 01/30/2023]
Abstract
Disruptions to sensory pathways in the lower urinary tract commonly occur and can give rise to lower urinary tract symptoms (LUTS). The unmet clinical need for treatment of LUTS has stimulated research into the molecular mechanisms that underlie neuronal control of the bladder and transient receptor potential (TRP) channels have emerged as key regulators of the sensory processes that regulate bladder function. TRP channels function as molecular sensors in urothelial cells and afferent nerve fibres and can be considered the origin of bladder sensations. TRP channels in the lower urinary tract contribute to the generation of normal and abnormal bladder sensations through a variety of mechanisms, and have demonstrated potential as targets for the treatment of LUTS in functional disorders of the lower urinary tract.
Collapse
Affiliation(s)
- Matthias Vanneste
- Laboratory of Ion Channel Research, VIB Center for Brain & Disease Research, Leuven, and Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Andrei Segal
- Laboratory of Ion Channel Research, VIB Center for Brain & Disease Research, Leuven, and Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Thomas Voets
- Laboratory of Ion Channel Research, VIB Center for Brain & Disease Research, Leuven, and Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Wouter Everaerts
- Laboratory of Experimental Urology, Department of Development and Regeneration, KU Leuven, Leuven, Belgium.
| |
Collapse
|
112
|
Cibert-Goton V, Lam C, Lingaya M, Falcone Y, Wood JN, Bulmer DC, Spiller R. Pain Severity Correlates With Biopsy-Mediated Colonic Afferent Activation But Not Psychological Scores in Patients With IBS-D. Clin Transl Gastroenterol 2021; 12:e00313. [PMID: 33617189 PMCID: PMC7901800 DOI: 10.14309/ctg.0000000000000313] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 01/07/2021] [Indexed: 12/26/2022] Open
Abstract
INTRODUCTION Despite heterogeneity, an increased prevalence of psychological comorbidity and an altered pronociceptive gut microenvironment have repeatedly emerged as causative pathophysiology in patients with irritable bowel syndrome (IBS). Our aim was to study these phenomena by comparing gut-related symptoms, psychological scores, and biopsy samples generated from a detailed diarrhea-predominant IBS patient (IBS-D) cohort before their entry into a previously reported clinical trial. METHODS Data were generated from 42 patients with IBS-D who completed a daily 2-week bowel symptom diary, the Hospital Anxiety and Depression score, and the Patient Health Questionnaire-12 Somatic Symptom score and underwent unprepared flexible sigmoidoscopy. Sigmoid mucosal biopsies were separately evaluated using immunohistochemistry and culture supernatants to determine cellularity, mediator levels, and ability to stimulate colonic afferent activity. RESULTS Pain severity scores significantly correlated with the daily duration of pain (r = 0.67, P < 0.00001), urgency (r = 0.57, P < 0.0005), and bloating (r = 0.39, P < 0.05), but not with psychological symptom scores for anxiety, depression, or somatization. Furthermore, pain severity scores from individual patients with IBS-D were significantly correlated (r = 0.40, P < 0.008) with stimulation of colonic afferent activation mediated by their biopsy supernatant, but not with biopsy cell counts nor measured mediator levels. DISCUSSION Peripheral pronociceptive changes in the bowel seem more important than psychological factors in determining pain severity within a tightly phenotyped cohort of patients with IBS-D. No individual mediator was identified as the cause of this pronociceptive change, suggesting that nerve targeting therapeutic approaches may be more successful than mediator-driven approaches for the treatment of pain in IBS-D.
Collapse
Affiliation(s)
- Vincent Cibert-Goton
- National Centre for Bowel Research, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK;
| | - Ching Lam
- NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | - Melanie Lingaya
- NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | - Yirga Falcone
- NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | - John N. Wood
- Molecular Nociception Group, Wolfson Institute of Biomedical Sciences, University College London, London, UK;
| | - David C. Bulmer
- National Centre for Bowel Research, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK;
- Department of Pharmacology, University of Cambridge, Cambridge, UK.
| | - Robin Spiller
- NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK
| |
Collapse
|
113
|
Serhan N, Cenac N, Basso L, Gaudenzio N. Mas-related G protein-coupled receptors (Mrgprs) - Key regulators of neuroimmune interactions. Neurosci Lett 2021; 749:135724. [PMID: 33600909 DOI: 10.1016/j.neulet.2021.135724] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 02/03/2021] [Accepted: 02/05/2021] [Indexed: 02/06/2023]
Abstract
Interplay between physiological systems in the body plays a prominent role in health and disease. At the cellular level, such interplay is orchestrated through the binding of specific ligands to their receptors expressed on cell surface. G protein-coupled receptors (GPCR) are seven-transmembrane domain receptors that initiate various cellular responses and regulate homeostasis. In this review, we focus on particular GPCRs named Mas-related G protein-coupled receptors (Mrgprs) mainly expressed by sensory neurons and specialized immune cells. We describe the different subfamilies of Mrgprs and their specific ligands, as well as recent advances in the field that illustrate the role played by these receptors in neuro-immune biological processes, including itch, pain and inflammation in diverse organs.
Collapse
Affiliation(s)
- Nadine Serhan
- Toulouse Institute for Infectious and Inflammatory Diseases, INSERM UMR1291, CNRS UMR5051, University of Toulouse III, Toulouse, France
| | - Nicolas Cenac
- IRSD, Université de Toulouse, INSERM, INRA, INP-ENVT, Université de Toulouse 3 Paul Sabatier, Toulouse, France
| | - Lilian Basso
- Toulouse Institute for Infectious and Inflammatory Diseases, INSERM UMR1291, CNRS UMR5051, University of Toulouse III, Toulouse, France.
| | - Nicolas Gaudenzio
- Toulouse Institute for Infectious and Inflammatory Diseases, INSERM UMR1291, CNRS UMR5051, University of Toulouse III, Toulouse, France.
| |
Collapse
|
114
|
Cardoso FC, Castro J, Grundy L, Schober G, Garcia-Caraballo S, Zhao T, Herzig V, King GF, Brierley SM, Lewis RJ. A spider-venom peptide with multitarget activity on sodium and calcium channels alleviates chronic visceral pain in a model of irritable bowel syndrome. Pain 2021; 162:569-581. [PMID: 32826759 DOI: 10.1097/j.pain.0000000000002041] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 08/04/2020] [Indexed: 12/19/2022]
Abstract
ABSTRACT Chronic pain is a serious debilitating condition that affects ∼20% of the world's population. Currently available drugs fail to produce effective pain relief in many patients and have dose-limiting side effects. Several voltage-gated sodium (NaV) and calcium (CaV) channels are implicated in the etiology of chronic pain, particularly NaV1.1, NaV1.3, NaV1.7-NaV1.9, CaV2.2, and CaV3.2. Numerous NaV and CaV modulators have been described, but with few exceptions, they display poor potency and/or selectivity for pain-related channel subtypes. Here, we report the discovery and characterization of 2 novel tarantula-venom peptides (Tap1a and Tap2a) isolated from Theraphosa apophysis venom that modulate the activity of both NaV and CaV3 channels. Tap1a and Tap2a inhibited on-target NaV and CaV3 channels at nanomolar to micromolar concentrations and displayed moderate off-target selectivity for NaV1.6 and weak affinity for NaV1.4 and NaV1.5. The most potent inhibitor, Tap1a, nearly ablated neuronal mechanosensitivity in afferent fibers innervating the colon and the bladder, with in vivo intracolonic administration reversing colonic mechanical hypersensitivity in a mouse model of irritable bowel syndrome. These findings suggest that targeting a specific combination of NaV and CaV3 subtypes provides a novel route for treatment of chronic visceral pain.
Collapse
Affiliation(s)
- Fernanda C Cardoso
- Centre for Pain Research, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland, Australia
| | - Joel Castro
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, South Australia, Australia
- Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, South Australia, Australia
| | - Luke Grundy
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, South Australia, Australia
- Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, South Australia, Australia
| | - Gudrun Schober
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, South Australia, Australia
- Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, South Australia, Australia
| | - Sonia Garcia-Caraballo
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, South Australia, Australia
- Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, South Australia, Australia
| | - Tianjiao Zhao
- Centre for Pain Research, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland, Australia
| | - Volker Herzig
- Centre for Pain Research, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland, Australia
- School of Science and Engineering, University of the Sunshine Coast, Sippy Downs, Queensland, Australia
| | - Glenn F King
- Centre for Pain Research, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland, Australia
| | - Stuart M Brierley
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, South Australia, Australia
- Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, South Australia, Australia
| | - Richard J Lewis
- Centre for Pain Research, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland, Australia
| |
Collapse
|
115
|
Kameneva P, Kastriti ME, Adameyko I. Neuronal lineages derived from the nerve-associated Schwann cell precursors. Cell Mol Life Sci 2021; 78:513-529. [PMID: 32748156 PMCID: PMC7873084 DOI: 10.1007/s00018-020-03609-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 05/18/2020] [Accepted: 07/22/2020] [Indexed: 12/26/2022]
Abstract
For a long time, neurogenic placodes and migratory neural crest cells were considered the immediate sources building neurons of peripheral nervous system. Recently, a number of discoveries revealed the existence of another progenitor type-a nerve-associated multipotent Schwann cell precursors (SCPs) building enteric and parasympathetic neurons as well as neuroendocrine chromaffin cells. SCPs are neural crest-derived and are similar to the crest cells by their markers and differentiation potential. Such similarities, but also considerable differences, raise many questions pertaining to the medical side, fundamental developmental biology and evolution. Here, we discuss the genesis of Schwann cell precursors, their role in building peripheral neural structures and ponder on their role in the origin in congenial diseases associated with peripheral nervous systems.
Collapse
Affiliation(s)
- Polina Kameneva
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, 171 77, Sweden
| | - Maria Eleni Kastriti
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, 171 77, Sweden
- Department of Molecular Neurosciences, Center for Brain Research, Medical University Vienna, Vienna, 1090, Austria
| | - Igor Adameyko
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, 171 77, Sweden.
- Department of Molecular Neurosciences, Center for Brain Research, Medical University Vienna, Vienna, 1090, Austria.
| |
Collapse
|
116
|
The Somatosensory World of the African Naked Mole-Rat. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1319:197-220. [PMID: 34424517 DOI: 10.1007/978-3-030-65943-1_7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The naked mole-rat (Heterocephalus glaber) is famous for its longevity and unusual physiology. This eusocial species that lives in highly ordered and hierarchical colonies with a single breeding queen, also discovered secrets enabling somewhat pain-free living around 20 million years ago. Unlike most mammals, naked mole-rats do not feel the burn of chili pepper's active ingredient, capsaicin, nor the sting of acid. Indeed, by accumulating mutations in genes encoding proteins that are only now being exploited as targets for new pain therapies (the nerve growth factor receptor TrkA and voltage-gated sodium channel, NaV1.7), this species mastered the art of analgesia before humans evolved. Recently, we have identified pain-insensitivity as a trait shared by several closely related African mole-rat species. In this chapter we will show how African mole-rats have evolved pain insensitivity as well as discussing what the proximate factors may have been that led to the evolution of pain-free traits.
Collapse
|
117
|
McFarland AJ, Yousuf MS, Shiers S, Price TJ. Neurobiology of SARS-CoV-2 interactions with the peripheral nervous system: implications for COVID-19 and pain. Pain Rep 2021; 6:e885. [PMID: 33458558 PMCID: PMC7803673 DOI: 10.1097/pr9.0000000000000885] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/26/2020] [Accepted: 11/14/2020] [Indexed: 02/07/2023] Open
Abstract
SARS-CoV-2 is a novel coronavirus that infects cells through the angiotensin-converting enzyme 2 receptor, aided by proteases that prime the spike protein of the virus to enhance cellular entry. Neuropilin 1 and 2 (NRP1 and NRP2) act as additional viral entry factors. SARS-CoV-2 infection causes COVID-19 disease. There is now strong evidence for neurological impacts of COVID-19, with pain as an important symptom, both in the acute phase of the disease and at later stages that are colloquially referred to as "long COVID." In this narrative review, we discuss how COVID-19 may interact with the peripheral nervous system to cause pain in the early and late stages of the disease. We begin with a review of the state of the science on how viruses cause pain through direct and indirect interactions with nociceptors. We then cover what we currently know about how the unique cytokine profiles of moderate and severe COVID-19 may drive plasticity in nociceptors to promote pain and worsen existing pain states. Finally, we review evidence for direct infection of nociceptors by SARS-CoV-2 and the implications of this potential neurotropism. The state of the science points to multiple potential mechanisms through which COVID-19 could induce changes in nociceptor excitability that would be expected to promote pain, induce neuropathies, and worsen existing pain states.
Collapse
Affiliation(s)
- Amelia J. McFarland
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, USA
| | - Muhammad S. Yousuf
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, USA
| | - Stephanie Shiers
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, USA
| | - Theodore J. Price
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, USA
| |
Collapse
|
118
|
Single-Cell RNA Sequencing Reveals that the Switching of the Transcriptional Profiles of Cysteine-Related Genes Alters the Virulence of Entamoeba histolytica. mSystems 2020; 5:5/6/e01095-20. [PMID: 33361325 PMCID: PMC7762796 DOI: 10.1128/msystems.01095-20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Studies on the trophozoite of Entamoeba histolytica suggested this organism could accumulate polyploid cells in its proliferative phase and differentiate its cell cycle from that of other eukaryotes. Therefore, a single-cell sequencing technique was used to study the switching of the RNA transcription profiles of single amoebic trophozoites. Entamoeba histolytica is an intestinal protozoan that causes human amoebic colitis and extraintestinal abscesses. Virulence variation is observed in the pathogenicity of E. histolytica trophozoites, but the detailed mechanism remains unclear. Here, a single trophozoite was cultured alone, and the progeny of the trophozoites of each generation were subjected to single-cell RNA sequencing (scRNA-seq) to study the transcriptional profiles of trophozoites. The scRNA-seq analysis indicated the importance of sulfur metabolism and the proteasome pathway in pathogenicity, whereas the isobaric tags for relative and absolute quantitation (iTRAQ) proteomic analysis did not identify the bulk trophozoites. The trophozoite improved the synthesis of cysteine under cysteine-deficient conditions but downregulated the expression of the intermediate subunit of the lectin of E. histolytica trophozoites and retained the expression of the heavy subunit of lectin, resulting in decreased amoebic phagocytosis and cytotoxicity. The variation in the transmembrane kinase gene family might be critical in regulating the proteasome pathway. Thus, the scRNA-seq technique provided an improved understanding of the biological characteristics and the mechanism of virulence variation of amoebic trophozoites. IMPORTANCE Studies on the trophozoite of Entamoeba histolytica suggested this organism could accumulate polyploid cells in its proliferative phase and differentiate its cell cycle from that of other eukaryotes. Therefore, a single-cell sequencing technique was used to study the switching of the RNA transcription profiles of single amoebic trophozoites. We separated individual trophozoites from axenic cultured trophozoites, CHO cell-incubated trophozoites, and in vivo trophozoites. We found important changes in the sulfur and cysteine metabolism in pathogenicity. The trophozoites strategically regulated the expression of the cysteine-rich protein-encoding genes under cysteine-deficient conditions, thereby decreasing amoebic phagocytosis and cytotoxicity. The single-cell sequencing technique shows evident advantages in comparison with the isobaric tags for relative and absolute quantitation (iTRAQ) proteomic technology (bulk trophozoite level) and reveals the regulation strategy of trophozoites in the absence of exogenous cysteine. This regulation strategy may be the mechanism of virulence variation of amoebic trophozoites.
Collapse
|
119
|
Abstract
The gut-brain axis is a coordinated communication system that not only maintains homeostasis, but significantly influences higher cognitive functions and emotions, as well as neurological and behavioral disorders. Among the large populations of sensory and motor neurons that innervate the gut, insights into the function of primary afferent nociceptors, whose cell bodies reside in the dorsal root ganglia and nodose ganglia, have revealed their multiple crosstalk with several cell types within the gut wall, including epithelial, vascular, and immune cells. These bidirectional communications have immunoregulatory functions, control host response to pathogens, and modulate sensations associated with gastrointestinal disorders, through activation of immune cells and glia in the peripheral and central nervous system, respectively. Here, we will review the cellular and neurochemical basis of these interactions at the periphery, in dorsal root ganglia, and in the spinal cord. We will discuss the research gaps that should be addressed to get a better understanding of the multifunctional role of sensory neurons in maintaining gut homeostasis and regulating visceral sensitivity.
Collapse
Affiliation(s)
- Nasser Abdullah
- Department of Physiology and Pharmacology, Inflammation Research Network-Snyder Institute for Chronic Diseases and Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Manon Defaye
- Department of Physiology and Pharmacology, Inflammation Research Network-Snyder Institute for Chronic Diseases and Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Christophe Altier
- Department of Physiology and Pharmacology, Inflammation Research Network-Snyder Institute for Chronic Diseases and Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
120
|
Qiao LY, Tiwari N. Spinal neuron-glia-immune interaction in cross-organ sensitization. Am J Physiol Gastrointest Liver Physiol 2020; 319:G748-G760. [PMID: 33084399 PMCID: PMC7792669 DOI: 10.1152/ajpgi.00323.2020] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Inflammatory bowel disease (IBD) and irritable bowel syndrome (IBS), historically considered as regional gastrointestinal disorders with heightened colonic sensitivity, are increasingly recognized to have concurrent dysfunction of other visceral and somatic organs, such as urinary bladder hyperactivity, leg pain, and skin hypersensitivity. The interorgan sensory cross talk is, at large, termed "cross-organ sensitization." These organs, anatomically distant from one another, physiologically interlock through projecting their sensory information into dorsal root ganglia (DRG) and then the spinal cord for integrative processing. The fundamental question of how sensitization of colonic afferent neurons conveys nociceptive information to activate primary afferents that innervate distant organs remains ambiguous. In DRG, primary afferent neurons are surrounded by satellite glial cells (SGCs) and macrophage accumulation in response to signals of injury to form a neuron-glia-macrophage triad. Astrocytes and microglia are major resident nonneuronal cells in the spinal cord to interact, physically and chemically, with sensory synapses. Cumulative evidence gathered so far indicate the indispensable roles of paracrine/autocrine interactions among neurons, glial cells, and immune cells in sensory cross-activation. Dichotomizing afferents, sensory convergency in the spinal cord, spinal nerve comingling, and extensive sprouting of central axons of primary afferents each has significant roles in the process of cross-organ sensitization; however, more results are required to explain their functional contributions. DRG that are located outside the blood-brain barrier and reside upstream in the cascade of sensory flow from one organ to the other in cross-organ sensitization could be safer therapeutic targets to produce less central adverse effects.
Collapse
Affiliation(s)
- Liya Y. Qiao
- 1Department of Physiology and Biophysics, Commonwealth University School of Medicine, Richmond, Virginia,2Department of Internal Medicine, Commonwealth University School of Medicine, Richmond, Virginia
| | - Namrata Tiwari
- 1Department of Physiology and Biophysics, Commonwealth University School of Medicine, Richmond, Virginia
| |
Collapse
|
121
|
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
|
122
|
Affiliation(s)
- Stuart M Brierley
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders University, Bedford Park, SA, 5042, Australia. .,Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, SA, 5000, Australia. .,Discipline of Medicine, University of Adelaide, Adelaide, SA, 5000, Australia.
| |
Collapse
|
123
|
Abstract
Primary nociceptors are a heterogeneous class of peripheral somatosensory neurons, responsible for detecting noxious, pruriceptive, and thermal stimuli. These neurons are further divided into several molecularly defined subtypes that correlate with their functional sensory modalities and morphological features. During development, all nociceptors arise from a common pool of embryonic precursors, and then segregate progressively into their mature specialized phenotypes. In this review, we summarize the intrinsic transcriptional programs and extrinsic trophic factor signaling mechanisms that interact to control nociceptor diversification. We also discuss how recent transcriptome profiling studies have significantly advanced the field of sensory neuron development.
Collapse
Affiliation(s)
- Suna L Cranfill
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Wenqin Luo
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.
| |
Collapse
|
124
|
Hockley JR, Barker KH, Taylor TS, Callejo G, Husson ZM, Bulmer DC, Smith ESJ. Acid and inflammatory sensitisation of naked mole-rat colonic afferent nerves. Mol Pain 2020; 16:1744806920903150. [PMID: 31992138 PMCID: PMC6990608 DOI: 10.1177/1744806920903150] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Acid sensing in the gastrointestinal tract is required for gut homeostasis and the detection of tissue acidosis caused by ischaemia, inflammation and infection. In the colorectum, activation of colonic afferents by low pH contributes to visceral hypersensitivity and abdominal pain in human disease including during inflammatory bowel disease. The naked mole-rat (Heterocephalus glaber) shows no pain-related behaviour to subcutaneous acid injection and cutaneous afferents are insensitive to acid, an adaptation thought to be a consequence of the subterranean, likely hypercapnic, environment in which it lives. As such we sought to investigate naked mole-rat interoception within the gastrointestinal tract and how this differed from the mouse (Mus Musculus). Here, we show the presence of calcitonin gene-related peptide expressing extrinsic nerve fibres innervating both mesenteric blood vessels and the myenteric plexi of the smooth muscle layers of the naked mole-rat colorectum. Using ex vivo colonic-nerve electrophysiological recordings, we show differential sensitivity of naked mole-rat, compared to mouse, colonic afferents to acid and the prototypic inflammatory mediator bradykinin, but not direct mechanical stimuli. In naked mole-rat, but not mouse, we observed mechanical hypersensitivity to acid, whilst both species sensitised to bradykinin. Collectively, these findings suggest that naked mole-rat colonic afferents are capable of detecting acidic stimuli; however, their intracellular coupling to downstream molecular effectors of neuronal excitability and mechanotransduction likely differs between species.
Collapse
Affiliation(s)
- James Rf Hockley
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Katie H Barker
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Toni S Taylor
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Gerard Callejo
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Zoe M Husson
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - David C Bulmer
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Ewan St J Smith
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| |
Collapse
|
125
|
Shiers S, Ray PR, Wangzhou A, Sankaranarayanan I, Tatsui CE, Rhines LD, Li Y, Uhelski ML, Dougherty PM, Price TJ. ACE2 and SCARF expression in human dorsal root ganglion nociceptors: implications for SARS-CoV-2 virus neurological effects. Pain 2020; 161:2494-2501. [PMID: 32826754 PMCID: PMC7572821 DOI: 10.1097/j.pain.0000000000002051] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
SARS-CoV-2 has created a global crisis. COVID-19, the disease caused by the virus, is characterized by pneumonia, respiratory distress, and hypercoagulation and can be fatal. An early sign of infection is loss of smell, taste, and chemesthesis-loss of chemical sensation. Other neurological effects of the disease have been described, but not explained. It is now apparent that many of these neurological effects (for instance joint pain and headache) can persist for at least months after infection, suggesting a sensory neuronal involvement in persistent disease. We show that human dorsal root ganglion (DRG) neurons express the SARS-CoV-2 receptor, angiotensin-converting enzyme 2 at the RNA and protein level. We also demonstrate that SARS-CoV-2 and coronavirus-associated factors and receptors are broadly expressed in human DRG at the lumbar and thoracic level as assessed by bulk RNA sequencing. ACE2 mRNA is expressed by a subset of nociceptors that express MRGPRD mRNA, suggesting that SARS-CoV-2 may gain access to the nervous system through entry into neurons that form free nerve endings at the outermost layers of skin and luminal organs. Therefore, DRG sensory neurons are a potential target for SARS-CoV-2 invasion of the peripheral nervous system, and viral infection of human nociceptors may cause some of the persistent neurological effects seen in COVID-19.
Collapse
Affiliation(s)
- Stephanie Shiers
- University of Texas at Dallas, Department of Neuroscience and Center for Advanced Pain Studies, Richardson, TX
| | - Pradipta R. Ray
- University of Texas at Dallas, Department of Neuroscience and Center for Advanced Pain Studies, Richardson, TX
| | - Andi Wangzhou
- University of Texas at Dallas, Department of Neuroscience and Center for Advanced Pain Studies, Richardson, TX
| | - Ishwarya Sankaranarayanan
- University of Texas at Dallas, Department of Neuroscience and Center for Advanced Pain Studies, Richardson, TX
| | | | - Laurence D. Rhines
- University of Texas MD Anderson Cancer Center, Department of Neurosurgery, Houston, TX
| | - Yan Li
- University of Texas MD Anderson Cancer Center, Department of Anesthesia and Pain Medicine, Houston, TX
| | - Megan L Uhelski
- University of Texas MD Anderson Cancer Center, Department of Anesthesia and Pain Medicine, Houston, TX
| | - Patrick M. Dougherty
- University of Texas MD Anderson Cancer Center, Department of Anesthesia and Pain Medicine, Houston, TX
| | - Theodore J Price
- University of Texas at Dallas, Department of Neuroscience and Center for Advanced Pain Studies, Richardson, TX
| |
Collapse
|
126
|
Vermeiren S, Bellefroid EJ, Desiderio S. Vertebrate Sensory Ganglia: Common and Divergent Features of the Transcriptional Programs Generating Their Functional Specialization. Front Cell Dev Biol 2020; 8:587699. [PMID: 33195244 PMCID: PMC7649826 DOI: 10.3389/fcell.2020.587699] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 09/08/2020] [Indexed: 12/13/2022] Open
Abstract
Sensory fibers of the peripheral nervous system carry sensation from specific sense structures or use different tissues and organs as receptive fields, and convey this information to the central nervous system. In the head of vertebrates, each cranial sensory ganglia and associated nerves perform specific functions. Sensory ganglia are composed of different types of specialized neurons in which two broad categories can be distinguished, somatosensory neurons relaying all sensations that are felt and visceral sensory neurons sensing the internal milieu and controlling body homeostasis. While in the trunk somatosensory neurons composing the dorsal root ganglia are derived exclusively from neural crest cells, somato- and visceral sensory neurons of cranial sensory ganglia have a dual origin, with contributions from both neural crest and placodes. As most studies on sensory neurogenesis have focused on dorsal root ganglia, our understanding of the molecular mechanisms underlying the embryonic development of the different cranial sensory ganglia remains today rudimentary. However, using single-cell RNA sequencing, recent studies have made significant advances in the characterization of the neuronal diversity of most sensory ganglia. Here we summarize the general anatomy, function and neuronal diversity of cranial sensory ganglia. We then provide an overview of our current knowledge of the transcriptional networks controlling neurogenesis and neuronal diversification in the developing sensory system, focusing on cranial sensory ganglia, highlighting specific aspects of their development and comparing it to that of trunk sensory ganglia.
Collapse
Affiliation(s)
- Simon Vermeiren
- ULB Neuroscience Institute, Université Libre de Bruxelles, Gosselies, Belgium
| | - Eric J Bellefroid
- ULB Neuroscience Institute, Université Libre de Bruxelles, Gosselies, Belgium
| | - Simon Desiderio
- Institute for Neurosciences of Montpellier, INSERM U1051, University of Montpellier, Montpellier, France
| |
Collapse
|
127
|
Liu BL, Cao QL, Zhao X, Liu HZ, Zhang YQ. Inhibition of TRPV1 by SHP-1 in nociceptive primary sensory neurons is critical in PD-L1 analgesia. JCI Insight 2020; 5:137386. [PMID: 32960817 PMCID: PMC7605531 DOI: 10.1172/jci.insight.137386] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 09/10/2020] [Indexed: 12/15/2022] Open
Abstract
Recently programmed death-ligand 1 (PD-L1) receptor PD-1 was found in dorsal root ganglion (DRG) neurons, and PD-L1 activates PD-1 to inhibit inflammatory and neuropathic pain by modulating neuronal excitability. However, the downstream signaling of PD-1 in sensory neurons remains unclear. Here, we show that PD-L1 activated Src homology 2 domain-containing tyrosine phosphatase-1 (SHP-1) to downregulate transient receptor potential vanilloid 1 (TRPV1) in DRG neurons and inhibit bone cancer pain in mice. Local injection of PD-L1 produced analgesia. PD-1 in DRG neurons colocalized with TRPV1 and SHP-1. PD-L1 induced the phosphorylation of SHP-1 in DRG TRPV1 neurons and inhibited TRPV1 currents. Loss of TRPV1 in mice abolished bone cancer–induced thermal hyperalgesia and PD-L1 analgesia. Conditioned deletion of SHP-1 in NaV1.8+ neurons aggravated bone cancer pain and diminished the inhibition of PD-L1 on TRPV1 currents and pain. Together, our findings suggest that PD-L1/PD-1 signaling suppresses bone cancer pain via inhibition of TRPV1 activity. Our results also suggest that SHP-1 in sensory neurons is an endogenous pain inhibitor and delays the development of bone cancer pain via suppressing TRPV1 function. PD-L1/PD-1 signaling suppresses TRPV1 activity and alleviates pain-like behaviors via phosphorylation of SHP-1 in nociceptive primary sensory neurons in a mouse bone cancer model.
Collapse
Affiliation(s)
- Ben-Long Liu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Qi-Lai Cao
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Xin Zhao
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Hui-Zhu Liu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yu-Qiu Zhang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, Institutes of Brain Science, Fudan University, Shanghai, China.,Institutes of Integrative Medicine, Fudan University, Shanghai, China
| |
Collapse
|
128
|
Mecklenburg J, Zou Y, Wangzhou A, Garcia D, Lai Z, Tumanov AV, Dussor G, Price TJ, Akopian AN. Transcriptomic sex differences in sensory neuronal populations of mice. Sci Rep 2020; 10:15278. [PMID: 32943709 PMCID: PMC7499251 DOI: 10.1038/s41598-020-72285-z] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 08/24/2020] [Indexed: 12/24/2022] Open
Abstract
Many chronic pain conditions show sex differences in their epidemiology. This could be attributed to sex-dependent differential expression of genes (DEGs) involved in nociceptive pathways, including sensory neurons. This study aimed to identify sex-dependent DEGs in estrous female versus male sensory neurons, which were prepared by using different approaches and ganglion types. RNA-seq on non-purified sensory neuronal preparations, such as whole dorsal root ganglion (DRG) and hindpaw tissues, revealed only a few sex-dependent DEGs. Sensory neuron purification increased numbers of sex-dependent DEGs. These DEG sets were substantially influenced by preparation approaches and ganglion types [DRG vs trigeminal ganglia (TG)]. Percoll-gradient enriched DRG and TG neuronal fractions produced distinct sex-dependent DEG groups. We next isolated a subset of sensory neurons by sorting DRG neurons back-labeled from paw and thigh muscle. These neurons have a unique sex-dependent DEG set, yet there is similarity in biological processes linked to these different groups of sex-dependent DEGs. Female-predominant DEGs in sensory neurons relate to inflammatory, synaptic transmission and extracellular matrix reorganization processes that could exacerbate neuro-inflammation severity, especially in TG. Male-selective DEGs were linked to oxidative phosphorylation and protein/molecule metabolism and production. Our findings catalog preparation-dependent sex differences in neuronal gene expressions in sensory ganglia.
Collapse
Affiliation(s)
- Jennifer Mecklenburg
- Department of Endodontics, University of Texas Health Science Center at San Antonio (UTHSCSA), San Antonio, TX, 78229, USA
| | - Yi Zou
- Greehey Children's Cancer Research Institute, UTHSCSA, San Antonio, TX, USA
| | - Andi Wangzhou
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas School of Behavioral and Brain Sciences, Richardson, TX, 75080, USA
| | - Dawn Garcia
- Greehey Children's Cancer Research Institute, UTHSCSA, San Antonio, TX, USA
| | - Zhao Lai
- Greehey Children's Cancer Research Institute, UTHSCSA, San Antonio, TX, USA
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio (UTHSCSA), San Antonio, TX, 78229, USA
| | - Alexei V Tumanov
- Departments of Microbiology, Immunology & Molecular Genetics, University of Texas Health Science Center at San Antonio (UTHSCSA), San Antonio, TX, 78229, USA
| | - Gregory Dussor
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas School of Behavioral and Brain Sciences, Richardson, TX, 75080, USA
| | - Theodore J Price
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas School of Behavioral and Brain Sciences, Richardson, TX, 75080, USA
| | - Armen N Akopian
- Department of Endodontics, University of Texas Health Science Center at San Antonio (UTHSCSA), San Antonio, TX, 78229, USA.
- Department of Pharmacology, The School of Dentistry, University of Texas Health Science Center at San Antonio (UTHSCSA), 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA.
| |
Collapse
|
129
|
Barrantes FJ. Central Nervous System Targets and Routes for SARS-CoV-2: Current Views and New Hypotheses. ACS Chem Neurosci 2020; 11:2793-2803. [PMID: 32845609 PMCID: PMC7460807 DOI: 10.1021/acschemneuro.0c00434] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 08/07/2020] [Indexed: 01/08/2023] Open
Abstract
As the coronavirus disease 2019 (COVID-19) pandemic unfolds, neurological signs and symptoms reflect the involvement of targets beyond the primary lung effects. The etiological agent of COVID-19, the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), exhibits neurotropism for central and peripheral nervous systems. Various infective mechanisms and paths can be exploited by the virus to reach the central nervous system, some of which bypass the blood-brain barrier; others alter its integrity. Numerous studies have established beyond doubt that the membrane-bound metalloprotease angiotensin-converting enzyme 2 (ACE2) performs the role of SARS-CoV-2 host-cell receptor. Histochemical studies and more recently transcriptomics of mRNA have dissected the cellular localization of the ACE2 enzyme in various tissues, including the central nervous system. Epithelial cells lining the nasal mucosae, the upper respiratory tract, and the oral cavity, bronchoalveolar cells type II in the pulmonary parenchyma, and intestinal enterocytes display ACE2 binding sites at their cell surfaces, making these epithelial mucosae the most likely viral entry points. Neuronal and glial cells and endothelial cells in the central nervous system also express ACE2. This short review analyzes the known entry points and routes followed by the SARS-CoV-2 to reach the central nervous system and postulates new hypothetical pathways stemming from the enterocytes lining the intestinal lumen.
Collapse
Affiliation(s)
- Francisco J. Barrantes
- Institute of Biomedical Research (BIOMED),
UCA-CONICET, Av. Alicia Moreau de
Justo 1600, C1107AFF Buenos Aires, Argentina
| |
Collapse
|
130
|
Meerschaert KA, Adelman PC, Friedman RL, Albers KM, Koerber HR, Davis BM. Unique Molecular Characteristics of Visceral Afferents Arising from Different Levels of the Neuraxis: Location of Afferent Somata Predicts Function and Stimulus Detection Modalities. J Neurosci 2020; 40:7216-7228. [PMID: 32817244 PMCID: PMC7534907 DOI: 10.1523/jneurosci.1426-20.2020] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 07/30/2020] [Accepted: 08/07/2020] [Indexed: 02/07/2023] Open
Abstract
Viscera receive innervation from sensory ganglia located adjacent to multiple levels of the brainstem and spinal cord. Here we examined whether molecular profiling could be used to identify functional clusters of colon afferents from thoracolumbar (TL), lumbosacral (LS), and nodose ganglia (NG) in male and female mice. Profiling of TL and LS bladder afferents was also performed. Visceral afferents were back-labeled using retrograde tracers injected into proximal and distal regions of colon or bladder, followed by single-cell qRT-PCR and analysis via an automated hierarchical clustering method. Genes were chosen for assay (32 for bladder; 48 for colon) based on their established role in stimulus detection, regulation of sensitivity/function, or neuroimmune interaction. A total of 132 colon afferents (from NG, TL, and LS ganglia) and 128 bladder afferents (from TL and LS ganglia) were analyzed. Retrograde labeling from the colon showed that NG and TL afferents innervate proximal and distal regions of the colon, whereas 98% of LS afferents only project to distal regions. There were clusters of colon and bladder afferents, defined by mRNA profiling, that localized to either TL or LS ganglia. Mixed TL/LS clustering also was found. In addition, transcriptionally, NG colon afferents were almost completely segregated from colon TL and LS neurons. Furthermore, colon and bladder afferents expressed genes at similar levels, although different gene combinations defined the clusters. These results indicate that genes implicated in both homeostatic regulation and conscious sensations are found at all anatomic levels, suggesting that afferents from different portions of the neuraxis have overlapping functions.SIGNIFICANCE STATEMENT Visceral organs are innervated by sensory neurons whose cell bodies are located in multiple ganglia associated with the brainstem and spinal cord. For the colon, this overlapping innervation is proposed to facilitate visceral sensation and homeostasis, where sensation and pain are mediated by spinal afferents and fear and anxiety (the affective aspects of visceral pain) are the domain of nodose afferents. The transcriptomic analysis performed here reveals that genes implicated in both homeostatic regulation and pain are found in afferents across all ganglia types, suggesting that conscious sensation and homeostatic regulation are the result of convergence, and not segregation, of sensory input.
Collapse
Affiliation(s)
- Kimberly A Meerschaert
- Department of Neurobiology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
- Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| | | | - Robert L Friedman
- Department of Neurobiology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
- Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| | - Kathryn M Albers
- Department of Neurobiology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
- Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| | - H Richard Koerber
- Department of Neurobiology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
- Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| | - Brian M Davis
- Department of Neurobiology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
- Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| |
Collapse
|
131
|
Kumar V, Mahajan N, Khare P, Kondepudi KK, Bishnoi M. Role of TRPV1 in colonic mucin production and gut microbiota profile. Eur J Pharmacol 2020; 888:173567. [PMID: 32946867 DOI: 10.1016/j.ejphar.2020.173567] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 09/12/2020] [Accepted: 09/14/2020] [Indexed: 12/16/2022]
Abstract
This study focuses on exploring the role of sensory cation channel Transient Receptor Potential channel subfamily Vanilloid 1 (TRPV1) in gut health, specifically mucus production and microflora profile in gut. We employed resiniferatoxin (ultrapotent TRPV1 agonist) induced chemo-denervation model in rats and studied the effects of TRPV1 ablation on colonic mucus secretion patterns. Histological and transcriptional analysis showed substantial decrease in mucus production as well as in expression of genes involved in goblet cell differentiation, mucin production and glycosylation. 16S metagenome analysis revealed changes in abundance of various gut bacteria, including decrease in beneficial bacteria like Lactobacillus spp and Clostridia spp. Also, TRPV1 ablation significantly decreased the levels of short chain fatty acids, i.e. acetate and butyrate. The present study provides first evidence that systemic TRPV1 ablation leads to impairment in mucus production and causes dysbiosis in gut. Further, it suggests to address mucin production and gut microbiota related adverse effects during the development of TRPV1 antagonism/ablation-based therapeutic and preventive strategies.
Collapse
Affiliation(s)
- Vijay Kumar
- National Agri-Food Biotechnology Institute (NABI), Knowledge City-Sector 81, SAS Nagar, Punjab, 140306, India; Department of Biotechnology, Panjab University, Sector-25, Chandigarh, 160014, India
| | - Neha Mahajan
- National Agri-Food Biotechnology Institute (NABI), Knowledge City-Sector 81, SAS Nagar, Punjab, 140306, India; Regional Centre for Biotechnology, Faridabad-Gurgaon expressway, Faridabad, Haryana, 121001, India
| | - Pragyanshu Khare
- National Agri-Food Biotechnology Institute (NABI), Knowledge City-Sector 81, SAS Nagar, Punjab, 140306, India
| | - Kanthi Kiran Kondepudi
- National Agri-Food Biotechnology Institute (NABI), Knowledge City-Sector 81, SAS Nagar, Punjab, 140306, India
| | - Mahendra Bishnoi
- National Agri-Food Biotechnology Institute (NABI), Knowledge City-Sector 81, SAS Nagar, Punjab, 140306, India.
| |
Collapse
|
132
|
Ramirez V, Swain S, Murray K, Reardon C. Neural Immune Communication in the Control of Host-Bacterial Pathogen Interactions in the Gastrointestinal Tract. Infect Immun 2020; 88:e00928-19. [PMID: 32341116 PMCID: PMC7440759 DOI: 10.1128/iai.00928-19] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The orchestration of host immune responses to enteric bacterial pathogens is a complex process involving the integration of numerous signals, including from the nervous system. Despite the recent progress in understanding the contribution of neuroimmune interactions in the regulation of inflammation, the mechanisms and effects of this communication during enteric bacterial infection are only beginning to be characterized. As part of this neuroimmune communication, neurons specialized to detect painful or otherwise noxious stimuli can respond to bacterial pathogens. Highlighting the complexity of these systems, the immunological consequences of sensory neuron activation can be either host adaptive or maladaptive, depending on the pathogen and organ system. These are but one of many types of neuroimmune circuits, with the vagus nerve and sympathetic innervation of numerous organs now known to modulate immune cell function and therefore dictate immunological outcomes during health and disease. Here, we review the evidence for neuroimmune communication in response to bacterial pathogens, and then discuss the consequences to host morbidity and mortality during infection of the gastrointestinal tract.
Collapse
Affiliation(s)
- Valerie Ramirez
- Department. of Anatomy, Physiology, and Cell Biology, UC Davis School of Veterinary Medicine, Davis, California, USA
| | - Samantha Swain
- Department. of Anatomy, Physiology, and Cell Biology, UC Davis School of Veterinary Medicine, Davis, California, USA
| | - Kaitlin Murray
- Department. of Anatomy, Physiology, and Cell Biology, UC Davis School of Veterinary Medicine, Davis, California, USA
| | - Colin Reardon
- Department. of Anatomy, Physiology, and Cell Biology, UC Davis School of Veterinary Medicine, Davis, California, USA
| |
Collapse
|
133
|
Chakrabarti S, Pattison LA, Doleschall B, Rickman RH, Blake H, Callejo G, Heppenstall PA, Smith ESJ. Intraarticular Adeno-Associated Virus Serotype AAV-PHP.S-Mediated Chemogenetic Targeting of Knee-Innervating Dorsal Root Ganglion Neurons Alleviates Inflammatory Pain in Mice. Arthritis Rheumatol 2020; 72:1749-1758. [PMID: 32418284 DOI: 10.1002/art.41314] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 05/12/2020] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Joint pain is the major clinical symptom of arthritis that affects millions of people. Controlling the excitability of knee-innervating dorsal root ganglion (DRG) neurons (knee neurons) could potentially provide pain relief. We undertook this study to evaluate whether the newly engineered adeno-associated virus (AAV) serotype, AAV-PHP.S, can deliver functional artificial receptors to control knee neuron excitability following intraarticular knee injection. METHODS The AAV-PHP.S virus, packaged with dTomato fluorescent protein and either excitatory (Gq ) or inhibitory (Gi ) designer receptors exclusively activated by designer drugs (DREADDs), was injected into the knee joints of adult mice. Labeling of DRG neurons with AAV-PHP.S from the knee was evaluated using immunohistochemistry. The functionality of Gq - and Gi -DREADDs was evaluated using whole-cell patch clamp electrophysiology on acutely cultured DRG neurons. Pain behavior in mice was assessed using a digging assay, dynamic weight bearing, and rotarod performance, before and after intraperitoneal administration of the DREADD activator, Compound 21. RESULTS We showed that AAV-PHP.S can deliver functional genes into ~7% of lumbar DRG neurons when injected into the knee joint in a similar manner to the well-established retrograde tracer, fast blue. Short-term activation of AAV-PHP.S-delivered Gq -DREADD increased excitability of knee neurons in vitro (P = 0.02 by unpaired t-test), without inducing overt pain in mice when activated in vivo. By contrast, in vivo Gi -DREADD activation alleviated digging deficits induced by Freund's complete adjuvant-mediated knee inflammation (P = 0.0002 by repeated-measures analysis of variance [ANOVA] followed by Holm-Sidak multiple comparisons test). A concomitant decrease in knee neuron excitability was observed in vitro (P = 0.005 by ANOVA followed by Holm-Sidak multiple comparisons test). CONCLUSION We describe an AAV-mediated chemogenetic approach to specifically control joint pain, which may be utilized in translational arthritic pain research.
Collapse
|
134
|
Development of a Mouse Reporter Strain for the Purinergic P2X 2 Receptor. eNeuro 2020; 7:ENEURO.0203-20.2020. [PMID: 32669344 PMCID: PMC7418537 DOI: 10.1523/eneuro.0203-20.2020] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/23/2020] [Accepted: 07/03/2020] [Indexed: 02/06/2023] Open
Abstract
The ATP-sensitive P2X2 ionotropic receptor plays a critical role in a number of signal processes including taste and hearing, carotid body detection of hypoxia, the exercise pressor reflex and sensory transduction of mechanical stimuli in the airways and bladder. Elucidation of the role of P2X2 has been hindered by the lack of selective tools. In particular, detection of P2X2 using established pharmacological and biochemical techniques yields dramatically different expression patterns, particularly in the peripheral and central nervous systems. Here, we have developed a knock-in P2X2-cre mouse, which we crossed with a cre-sensitive tdTomato reporter mouse to determine P2X2 expression. P2X2 was found in more than 80% of nodose vagal afferent neurons, but not in jugular vagal afferent neurons. Reporter expression correlated in vagal neurons with sensitivity to α,β methylene ATP (αβmATP). P2X2 was expressed in 75% of petrosal afferents, but only 12% and 4% of dorsal root ganglia (DRG) and trigeminal afferents, respectively. P2X2 expression was limited to very few cell types systemically. Together with the central terminals of P2X2-expressing afferents, reporter expression in the CNS was mainly found in brainstem neurons projecting mossy fibers to the cerebellum, with little expression in the hippocampus or cortex. The structure of peripheral terminals of P2X2-expressing afferents was demonstrated in the tongue (taste buds), carotid body, trachea and esophagus. P2X2 was observed in hair cells and support cells in the cochlear, but not in spiral afferent neurons. This mouse strain provides a novel approach to the identification and manipulation of P2X2-expressing cell types.
Collapse
|
135
|
Chakrabarti S, Ai M, Henson FM, Smith ESJ. Peripheral mechanisms of arthritic pain: A proposal to leverage large animals for in vitro studies. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2020; 8:100051. [PMID: 32817908 PMCID: PMC7426561 DOI: 10.1016/j.ynpai.2020.100051] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/22/2020] [Accepted: 07/22/2020] [Indexed: 04/14/2023]
Abstract
Pain arising from musculoskeletal disorders such as arthritis is one of the leading causes of disability. Whereas the past 20-years has seen an increase in targeted therapies for rheumatoid arthritis (RA), other arthritis conditions, especially osteoarthritis, remain poorly treated. Although modulation of central pain pathways occurs in chronic arthritis, multiple lines of evidence indicate that peripherally driven pain is important in arthritic pain. To understand the peripheral mechanisms of arthritic pain, various in vitro and in vivo models have been developed, largely in rodents. Although rodent models provide numerous advantages for studying arthritis pathogenesis and treatment, the anatomy and biomechanics of rodent joints differ considerably to those of humans. By contrast, the anatomy and biomechanics of joints in larger animals, such as dogs, show greater similarity to human joints and thus studying them can provide novel insight for arthritis research. The purpose of this article is firstly to review models of arthritis and behavioral outcomes commonly used in large animals. Secondly, we review the existing in vitro models and assays used to study arthritic pain, primarily in rodents, and discuss the potential for adopting these strategies, as well as likely limitations, in large animals. We believe that exploring peripheral mechanisms of arthritic pain in vitro in large animals has the potential to reduce the veterinary burden of arthritis in commonly afflicted species like dogs, as well as to improve translatability of pain research into the clinic.
Collapse
Affiliation(s)
- Sampurna Chakrabarti
- Department of Neuroscience, Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin, Germany
- Department of Pharmacology, University of Cambridge, UK
| | - Minji Ai
- Department of Veterinary Medicine, University of Cambridge, UK
| | | | | |
Collapse
|
136
|
Browe BM, Olsen AR, Ramirez C, Rickman RH, Smith ESJ, Park TJ. The naked mole-rat has a functional purinergic pain pathway despite having a non-functional peptidergic pain pathway. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2020; 8:100047. [PMID: 32478202 PMCID: PMC7248424 DOI: 10.1016/j.ynpai.2020.100047] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 04/29/2020] [Accepted: 05/01/2020] [Indexed: 01/08/2023]
Abstract
Naked mole-rats (Heterocephalus glaber) have adaptations within their pain pathway that are beneficial to survival in large colonies within poorly ventilated burrow systems, with lower O2 and higher CO2 ambient levels than ground-level environments. These adaptations ultimately lead to a partial disruption of the C-fiber pain pathway, which enables naked mole-rats to not feel pain from the acidosis associated with CO2 accumulation. One hallmark of this disruption is that naked mole-rats do not express neuropeptides, such as Substance P and calcitonin gene-related peptide in their cutaneous C-fibers, effectively making the peptidergic pain pathway hypofunctional. One C-fiber pathway that remains unstudied in the naked mole-rat is the non-peptidergic, purinergic pathway, despite this being a key pathway for inflammatory pain. The current study aimed to establish the functionality of the purinergic pathway in naked mole-rats and the effectiveness of cannabinoids in attenuating pain through this pathway. Cannabinoids can manage chronic inflammatory pain in both humans and mouse models, and studies suggest a major downstream role for the purinergic receptor, P2X3, in this treatment. Here we used Ca2+-imaging of cultured dorsal root ganglion neurons and in vivo behavioral testing to demonstrate that the P2X3 pathway is functional in naked mole-rats. Additionally, formalin-induced inflammatory pain was reduced by the cannabinoid receptor agonist, WIN55 (inflammatory, but not acute phase) and the P2X3 receptor antagonist A-317491 (acute and inflammatory phases). This study establishes that the purinergic C-fiber pathway is present and functional in naked mole-rats and that cannabinoid-mediated analgesia occurs in this species.
Collapse
Affiliation(s)
- Brigitte M. Browe
- Laboratory of Integrative Neuroscience, Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Abigail R. Olsen
- Laboratory of Integrative Neuroscience, Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Cesar Ramirez
- Laboratory of Integrative Neuroscience, Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Rebecca H. Rickman
- Department of Pharmacology, University of Cambridge, Cambridge CB2 1PD, UK
| | | | - Thomas J. Park
- Laboratory of Integrative Neuroscience, Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
137
|
Islam M, Chen B, Spraggins JM, Kelly RT, Lau KS. Use of Single-Cell -Omic Technologies to Study the Gastrointestinal Tract and Diseases, From Single Cell Identities to Patient Features. Gastroenterology 2020; 159:453-466.e1. [PMID: 32417404 PMCID: PMC7484006 DOI: 10.1053/j.gastro.2020.04.073] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 02/29/2020] [Accepted: 04/04/2020] [Indexed: 02/07/2023]
Abstract
Single cells are the building blocks of tissue systems that determine organ phenotypes, behaviors, and functions. Understanding the differences between cell types and their activities might provide us with insights into normal tissue physiology, development of disease, and new therapeutic strategies. Although -omic level single-cell technologies are a relatively recent development that have been used only in research settings, these approaches might eventually be used in the clinic. We review the prospects of applying single-cell genome, transcriptome, epigenome, proteome, and metabolome analyses to gastroenterology and hepatology research. Combining data from multi-omic platforms coupled to rapid technological development could lead to new diagnostic, prognostic, and therapeutic approaches.
Collapse
Affiliation(s)
- Mirazul Islam
- Epithelial Biology Center and Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Bob Chen
- Chemical and Physical Biology Program, Vanderbilt University, Nashville, Tennessee
| | - Jeffrey M Spraggins
- Mass Spectrometry Research Center, Departments of Biochemistry and Chemistry, Vanderbilt University, Nashville, Tennessee
| | - Ryan T Kelly
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah
| | - Ken S Lau
- Epithelial Biology Center and Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee; Chemical and Physical Biology Program, Vanderbilt University, Nashville, Tennessee.
| |
Collapse
|
138
|
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
|
139
|
Mayer F, Gunawan AL, Tso P, Aponte GW. Glucagon-like peptide 1 and glucose-dependent insulinotropic polypeptide stimulate release of substance P from TRPV1- and TRPA1-expressing sensory nerves. Am J Physiol Gastrointest Liver Physiol 2020; 319:G23-G35. [PMID: 32421358 PMCID: PMC7468754 DOI: 10.1152/ajpgi.00189.2019] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) are released from enteroendocrine cells (EECs) in response to nutrient ingestion and lower blood glucose levels by stimulation of insulin secretion and thus are defined as incretins. GLP-1 receptor (GLP-1R) expression has been identified on enteric neurons that include intrinsic afferent neurons, extrinsic spinal, and vagal sensory afferents but has not been shown to have an incretin effect through these nerves. GLP-1 and GIP enter the mesenteric lymphatic fluid (MLF) after a meal via the interstitial fluid (IF) from local tissue secretion and/or blood capillaries. We tested if MLF could induce diet-dependent intransient increases in intracellular calcium ([Ca2+]i) in cultured sensory neurons. Postprandial rat MLF, collected from the superior mesenteric lymphatic duct, induced a significant twofold higher intransient increase in [Ca2+]i in primary-cultured sensory neurons than MLF from fasted rats. Inhibition of transient receptor potential vanilloid 1 (TRPV1) and TRPV1 and ankyrin 1 cation channels (TRPA1) with ruthenium red eliminated the difference. Substance P (SP) (a peptide that stimulates insulin secretion) sensor cells cocultured with sensory neurons showed both the GLP-1R agonist exendin-4 (Ex-4) and GIP induced transient increases in [Ca2+]i directly coupled to SP secretion in the sensory nerves. Ex-4-induced release of SP required expression of either TRPA1 or TRPV1. These data identify unrecognized actions of GLP-1 and GIP as incretins by acting as neurolymphocrines and suggest a mechanism for sensory nerves to respond to the postprandial state through MLF.NEW & NOTEWORTHY Glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) are secreted upon eating to lower blood sugar. GLP-1 and GIP were found to induce the secretion of substance P (SP) from cultured sensory nerves. SP enhances insulin secretion. Mesenteric lymphatic fluid (MLF) also stimulates sensory neurons in a diet-dependent manner. These studies identify new actions of GLP-1 and GIP as incretins and suggest a mechanism for sensory nerves to respond to diet through MLF.
Collapse
Affiliation(s)
- Fahima Mayer
- 1Department of Nutritional Sciences and Toxicology, Graduate Program in Metabolic Biology, University of California, Berkeley, California
| | - Amanda L. Gunawan
- 1Department of Nutritional Sciences and Toxicology, Graduate Program in Metabolic Biology, University of California, Berkeley, California
| | - Patrick Tso
- 2Department of Pathobiology and Molecular Medicine, University of Cincinnati, Reading, Ohio
| | - Gregory W. Aponte
- 1Department of Nutritional Sciences and Toxicology, Graduate Program in Metabolic Biology, University of California, Berkeley, California
| |
Collapse
|
140
|
Chakrabarti S, Jadon DR, Bulmer DC, Smith ESJ. Human osteoarthritic synovial fluid increases excitability of mouse dorsal root ganglion sensory neurons: an in-vitro translational model to study arthritic pain. Rheumatology (Oxford) 2020; 59:662-667. [PMID: 31410487 DOI: 10.1093/rheumatology/kez331] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 07/05/2019] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVES Knee OA is a leading global cause of morbidity. This study investigates the effects of knee SF from patients with OA on the activity of dorsal root ganglion sensory neurons that innervate the knee (knee neurons) as a novel translational model of disease-mediated nociception in human OA. METHODS Dissociated cultures of mouse knee neurons were incubated overnight or acutely stimulated with OA-SF (n = 4) and fluid from healthy donors (n = 3, Ctrl-SF). Electrophysiology and Ca2+-imaging determined changes in electrical excitability and transient receptor potential channel function, respectively. RESULTS Incubation with OA-SF induced knee neuron hyperexcitability compared to Ctrl-SF: the resting membrane potential significantly increased (F(2, 92) = 5.6, P = 0.005, ANOVA) and the action potential threshold decreased (F(2, 92) = 8.8, P = 0.0003, ANOVA); TRPV1 (F(2, 445) = 3.7, P = 0.02) and TRPM8 (F(2, 174) = 11.1, P < 0.0001, ANOVA) channel activity also increased. Acute application of Ctrl-SF and OA-SF increased intracellular Ca2+ concentration via intra- and extracellular Ca2+ sources. CONCLUSION Human OA-SF acutely activated knee neurons and induced hyperexcitability indicating that mediators present in OA-SF stimulate sensory nerve activity and thereby give rise to knee pain. Taken together, this study provides proof-of-concept for a new method to study the ability of mediators present in joints of patients with arthritis to stimulate nociceptor activity and hence identify clinically relevant drug targets for treating knee pain.
Collapse
Affiliation(s)
| | - Deepak R Jadon
- Department of Medicine, University of Cambridge, Cambridge, UK.,Department of Rheumatology, Cambridge University Hospitals NHSFT, Cambridge, UK
| | | | | |
Collapse
|
141
|
Hassler SN, Kume M, Mwirigi JM, Ahmad A, Shiers S, Wangzhou A, Ray PR, Belugin SN, Naik DK, Burton MD, Vagner J, Boitano S, Akopian AN, Dussor G, Price TJ. The cellular basis of protease-activated receptor 2-evoked mechanical and affective pain. JCI Insight 2020; 5:137393. [PMID: 32352932 PMCID: PMC7308051 DOI: 10.1172/jci.insight.137393] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 04/24/2020] [Indexed: 12/31/2022] Open
Abstract
Protease-activated receptor 2 (PAR2) has long been implicated in inflammatory and visceral pain, but the cellular basis of PAR2-evoked pain has not been delineated. Although PAR2-evoked pain has been attributed to sensory neuron expression, RNA-sequencing experiments show ambiguous F2rl1 mRNA detection. Moreover, many pharmacological tools for PAR2 are nonspecific, acting also on the Mas-related GPCR family (Mrg) that are highly enriched in sensory neurons. We sought to clarify the cellular basis of PAR2-evoked pain. We developed a PAR2-conditional knockout mouse and specifically deleted PAR2 in all sensory neurons using the PirtCre mouse line. Our behavioral findings show that PAR2 agonist-evoked mechanical hyperalgesia and facial grimacing, but not thermal hyperalgesia, are dependent on PAR2 expression in sensory neurons that project to the hind paw in male and female mice. F2rl1 mRNA is expressed in a discrete population (~4%) of mostly small-diameter sensory neurons that coexpress the Nppb and IL31ra genes. This cell population has been implicated in itch, but our work shows that PAR2 activation in these cells causes clear pain-related behaviors from the skin. Our findings show that a discrete population of DRG sensory neurons mediate PAR2-evoked pain.
Collapse
Affiliation(s)
- Shayne N. Hassler
- School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, University of Texas at Dallas, Dallas, Texas, USA
| | - Moeno Kume
- School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, University of Texas at Dallas, Dallas, Texas, USA
| | - Juliet M. Mwirigi
- School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, University of Texas at Dallas, Dallas, Texas, USA
| | - Ayesha Ahmad
- School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, University of Texas at Dallas, Dallas, Texas, USA
| | - Stephanie Shiers
- School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, University of Texas at Dallas, Dallas, Texas, USA
| | - Andi Wangzhou
- School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, University of Texas at Dallas, Dallas, Texas, USA
| | - Pradipta R. Ray
- School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, University of Texas at Dallas, Dallas, Texas, USA
| | - Serge N. Belugin
- Department of Endodontics, School of Dentistry, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - Dhananjay K. Naik
- School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, University of Texas at Dallas, Dallas, Texas, USA
| | - Michael D. Burton
- School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, University of Texas at Dallas, Dallas, Texas, USA
| | | | - Scott Boitano
- BIO5 Research Institute and
- Department of Physiology, Asthma & Airway Disease Research Center, University of Arizona, Tucson, Arizona, USA
| | - Armen N. Akopian
- Department of Endodontics, School of Dentistry, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - Gregory Dussor
- School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, University of Texas at Dallas, Dallas, Texas, USA
| | - Theodore J. Price
- School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, University of Texas at Dallas, Dallas, Texas, USA
| |
Collapse
|
142
|
Mouse intestinal tuft cells express advillin but not villin. Sci Rep 2020; 10:8877. [PMID: 32483224 PMCID: PMC7264147 DOI: 10.1038/s41598-020-65469-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Accepted: 03/06/2020] [Indexed: 12/26/2022] Open
Abstract
Tuft (or brush) cells are solitary chemosensory cells scattered throughout the epithelia of the respiratory and alimentary tract. The actin-binding protein villin (Vil1) is used as a marker of tuft cells and the villin promoter is frequently used to drive expression of the Cre recombinase in tuft cells. While there is widespread agreement about the expression of villin in tuft cells there are several disagreements related to tuft cell lineage commitment and function. We now show that many of these inconsistencies could be resolved by our surprising finding that intestinal tuft cells, in fact, do not express villin protein. Furthermore, we show that a related actin-binding protein, advillin which shares 75% homology with villin, has a tuft cell restricted expression in the gastrointestinal epithelium. Our study identifies advillin as a marker of tuft cells and provides a mechanism for driving gene expression in tuft cells but not in other epithelial cells of the gastrointestinal tract. Our findings fundamentally change the way we identify and study intestinal tuft cells.
Collapse
|
143
|
Smith ESJ, Park TJ, Lewin GR. Independent evolution of pain insensitivity in African mole-rats: origins and mechanisms. J Comp Physiol A Neuroethol Sens Neural Behav Physiol 2020; 206:313-325. [PMID: 32206859 PMCID: PMC7192887 DOI: 10.1007/s00359-020-01414-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 02/10/2020] [Accepted: 02/27/2020] [Indexed: 12/21/2022]
Abstract
The naked mole-rat (Heterocephalus glaber) is famous for its longevity and unusual physiology. This eusocial species that lives in highly ordered and hierarchical colonies with a single breeding queen, also discovered secrets enabling somewhat pain-free living around 20 million years ago. Unlike most mammals, naked mole-rats do not feel the burn of chili pepper's active ingredient, capsaicin, nor the sting of acid. Indeed, by accumulating mutations in genes encoding proteins that are only now being exploited as targets for new pain therapies (the nerve growth factor receptor TrkA and voltage-gated sodium channel, NaV1.7), this species mastered the art of analgesia before humans evolved. Recently, we have identified pain insensitivity as a trait shared by several closely related African mole-rat species. One of these African mole-rats, the Highveld mole-rat (Cryptomys hottentotus pretoriae), is uniquely completely impervious and pain free when confronted with electrophilic compounds that activate the TRPA1 ion channel. The Highveld mole-rat has evolved a biophysical mechanism to shut down the activation of sensory neurons that drive pain. In this review, we will show how mole-rats have evolved pain insensitivity as well as discussing what the proximate factors may have been that led to the evolution of pain-free traits.
Collapse
Affiliation(s)
- Ewan St John Smith
- Department of Pharmacology, University of Cambridge, Cambridge, CB2 1PD, UK
| | - Thomas J Park
- Laboratory of Integrative Neuroscience, Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Gary R Lewin
- Molecular Physiology of Somatic Sensation, Max Delbrück Center for Molecular Medicine, Robert-Rössle Str. 10, D-13125, Berlin, Germany.
| |
Collapse
|
144
|
Esquerre N, Basso L, Defaye M, Vicentini FA, Cluny N, Bihan D, Hirota SA, Schick A, Jijon HB, Lewis IA, Geuking MB, Sharkey KA, Altier C, Nasser Y. Colitis-Induced Microbial Perturbation Promotes Postinflammatory Visceral Hypersensitivity. Cell Mol Gastroenterol Hepatol 2020; 10:225-244. [PMID: 32289500 PMCID: PMC7301239 DOI: 10.1016/j.jcmgh.2020.04.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 04/01/2020] [Accepted: 04/03/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Despite achieving endoscopic remission, more than 20% of inflammatory bowel disease patients experience chronic abdominal pain. These patients have increased rectal transient receptor potential vanilloid-1 receptor (TRPV1) expression, a key transducer of inflammatory pain. Because inflammatory bowel disease patients in remission exhibit dysbiosis and microbial manipulation alters TRPV1 function, our goal was to examine whether microbial perturbation modulated transient receptor potential function in a mouse model. METHODS Mice were given dextran sodium sulfate (DSS) to induce colitis and were allowed to recover. The microbiome was perturbed by using antibiotics as well as fecal microbial transplant (FMT). Visceral and somatic sensitivity were assessed by recording visceromotor responses to colorectal distention and using hot plate/automated Von Frey tests, respectively. Calcium imaging of isolated dorsal root ganglia neurons was used as an in vitro correlate of nociception. The microbiome composition was evaluated via 16S rRNA gene variable region V4 amplicon sequencing, whereas fecal short-chain fatty acids (SCFAs) were assessed by using targeted mass spectrometry. RESULTS Postinflammatory DSS mice developed visceral and somatic hyperalgesia. Antibiotic administration during DSS recovery induced visceral, but not somatic, hyperalgesia independent of inflammation. FMT of postinflammatory DSS stool into antibiotic-treated mice increased visceral hypersensitivity, whereas FMT of control stool reversed antibiotics' sensitizing effects. Postinflammatory mice exhibited both increased SCFA-producing species and fecal acetate/butyrate content compared with controls. Capsaicin-evoked calcium responses were increased in naive dorsal root ganglion neurons incubated with both sodium butyrate/propionate alone and with colonic supernatants derived from postinflammatory mice. CONCLUSIONS The microbiome plays a central role in postinflammatory visceral hypersensitivity. Microbial-derived SCFAs can sensitize nociceptive neurons and may contribute to the pathogenesis of postinflammatory visceral pain.
Collapse
Affiliation(s)
- Nicolas Esquerre
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary
| | - Lilian Basso
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary; Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary
| | - Manon Defaye
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary; Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary
| | - Fernando A Vicentini
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary; Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary; Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary
| | - Nina Cluny
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary; Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary; Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary
| | | | - Simon A Hirota
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary; Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary
| | - Alana Schick
- International Microbiome Centre, Cumming School of Medicine, University of Calgary
| | - Humberto B Jijon
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary; Division of Gastroenterology and Hepatology, Department of Medicine, University of Calgary
| | - Ian A Lewis
- Department of Biological Sciences, University of Calgary
| | - Markus B Geuking
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary; Department of Microbiology, Immunity and Infectious Diseases, Cumming School of Medicine, University of Calgary
| | - Keith A Sharkey
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary; Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary; Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary
| | - Christophe Altier
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary; Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary; Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Yasmin Nasser
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary; Division of Gastroenterology and Hepatology, Department of Medicine, University of Calgary.
| |
Collapse
|
145
|
Jameson KG, Olson CA, Kazmi SA, Hsiao EY. Toward Understanding Microbiome-Neuronal Signaling. Mol Cell 2020; 78:577-583. [PMID: 32275853 DOI: 10.1016/j.molcel.2020.03.006] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 02/04/2020] [Accepted: 03/02/2020] [Indexed: 12/19/2022]
Abstract
Host-associated microbiomes are emerging as important modifiers of brain activity and behavior. Metabolic, immune, and neuronal pathways are proposed to mediate communication across the so-called microbiota-gut-brain axis. However, strong mechanistic evidence, especially for direct signaling between microbes and sensory neurons, is lacking. Here, we discuss microbial regulation of short-chain fatty acids, neurotransmitters, as-yet-uncharacterized biochemicals, and derivatives of neuromodulatory drugs as important areas for assessing microbial interactions with the nervous system.
Collapse
Affiliation(s)
- K G Jameson
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - C A Olson
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - S A Kazmi
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - E Y Hsiao
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
146
|
Identification of a Sacral, Visceral Sensory Transcriptome in Embryonic and Adult Mice. eNeuro 2020; 7:ENEURO.0397-19.2019. [PMID: 31996391 PMCID: PMC7036621 DOI: 10.1523/eneuro.0397-19.2019] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 12/08/2019] [Accepted: 12/19/2019] [Indexed: 02/08/2023] Open
Abstract
Visceral sensory neurons encode distinct sensations from healthy organs and initiate pain states that are resistant to common analgesics. Transcriptome analysis is transforming our understanding of sensory neuron subtypes but has generally focused on somatic sensory neurons or the total population of neurons in which visceral neurons form the minority. Our aim was to define transcripts specifically expressed by sacral visceral sensory neurons, as a step towards understanding the unique biology of these neurons and potentially leading to identification of new analgesic targets for pelvic visceral pain. Our strategy was to identify genes differentially expressed between sacral dorsal root ganglia (DRG) that include somatic neurons and sacral visceral neurons, and adjacent lumbar DRG that comprise exclusively of somatic sensory neurons. This was performed in adult and E18.5 male and female mice. By developing a method to restrict analyses to nociceptive Trpv1 neurons, a larger group of genes were detected as differentially expressed between spinal levels. We identified many novel genes that had not previously been associated with pelvic visceral sensation or nociception. Limited sex differences were detected across the transcriptome of sensory ganglia, but more were revealed in sacral levels and especially in Trpv1 nociceptive neurons. These data will facilitate development of new tools to modify mature and developing sensory neurons and nociceptive pathways.
Collapse
|
147
|
Basso L, Benamar M, Mas-Orea X, Deraison C, Blanpied C, Cenac N, Saoudi A, Dietrich G. Endogenous control of inflammatory visceral pain by T cell-derived opioids in IL-10-deficient mice. Neurogastroenterol Motil 2020; 32:e13743. [PMID: 31588671 DOI: 10.1111/nmo.13743] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 09/17/2019] [Accepted: 09/18/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND The opioid-mediated analgesic activity of mucosal CD4+ T lymphocytes in colitis has been reported in immunocompetent mice so far. Here, we investigated whether CD4+ T lymphocytes alleviate from inflammation-induced abdominal pain in mice with defective immune regulation. METHODS Endogenous control of visceral pain by opioids locally produced in inflamed mucosa was assessed in IL-10-deficient mice. KEY RESULTS CD4+ T lymphocytes but not F4/80+ macrophages isolated from the lamina propria of IL-10-deficient mice with colitis express enkephalin-containing opioid peptides as assessed by cytofluorometry. Colitis in IL-10-/- mice was not associated with abdominal pain. Intraperitoneal injection of naloxone-methiodide, a peripheral opioid receptor antagonist, induced abdominal hypersensitivity in IL-10-/- mice with colitis. CONCLUSION AND INFERENCES Opioid-mediated analgesic activity of mucosal T lymphocytes remains operating in IL-10-/- mice with impaired immune regulation. The data suggest that endogenous T cell-derived opioids might reduce inflammation-induced abdominal pain in inflammatory bowel diseases associated with homozygous "loss of function mutations" in interleukin-10.
Collapse
Affiliation(s)
- Lilian Basso
- IRSD, INSERM, INRA, ENVT, UPS, Université de Toulouse, Toulouse, France
| | - Mehdi Benamar
- Centre de Physiopathologie de Toulouse Purpan (CPTP), UPS, INSERM, CNRS, Université de Toulouse, Toulouse, France
| | - Xavier Mas-Orea
- IRSD, INSERM, INRA, ENVT, UPS, Université de Toulouse, Toulouse, France
| | - Céline Deraison
- IRSD, INSERM, INRA, ENVT, UPS, Université de Toulouse, Toulouse, France
| | | | - Nicolas Cenac
- IRSD, INSERM, INRA, ENVT, UPS, Université de Toulouse, Toulouse, France
| | - Abdelhadi Saoudi
- Centre de Physiopathologie de Toulouse Purpan (CPTP), UPS, INSERM, CNRS, Université de Toulouse, Toulouse, France
| | - Gilles Dietrich
- IRSD, INSERM, INRA, ENVT, UPS, Université de Toulouse, Toulouse, France
| |
Collapse
|
148
|
Najjar SA, Davis BM, Albers KM. Epithelial-Neuronal Communication in the Colon: Implications for Visceral Pain. Trends Neurosci 2020; 43:170-181. [PMID: 31983457 DOI: 10.1016/j.tins.2019.12.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 12/18/2019] [Accepted: 12/30/2019] [Indexed: 02/07/2023]
Abstract
Visceral hypersensitivity and pain result, at least in part, from increased excitability of primary afferents that innervate the colon. In addition to intrinsic changes in these neurons, emerging evidence indicates that changes in lining epithelial cells may also contribute to increased excitability. Here we review recent studies on how colon epithelial cells communicate directly with colon afferents. Specifically, anatomical studies revealed specialized synaptic connections between epithelial cells and nerve fibers and studies using optogenetic activation of the epithelium showed initiation of pain-like responses. We review the possible mechanisms of epithelial-neuronal communication and provide an overview of the possible neurotransmitters and receptors involved. Understanding the biology of this interface and how it changes in pathological conditions may provide new treatments for visceral pain conditions.
Collapse
Affiliation(s)
- Sarah A Najjar
- Center for Neuroscience at the University of Pittsburgh, Pittsburgh Center for Pain Research and Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Brian M Davis
- Center for Neuroscience at the University of Pittsburgh, Pittsburgh Center for Pain Research and Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Kathryn M Albers
- Center for Neuroscience at the University of Pittsburgh, Pittsburgh Center for Pain Research and Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
149
|
Lai NY, Musser MA, Pinho-Ribeiro FA, Baral P, Jacobson A, Ma P, Potts DE, Chen Z, Paik D, Soualhi S, Yan Y, Misra A, Goldstein K, Lagomarsino VN, Nordstrom A, Sivanathan KN, Wallrapp A, Kuchroo VK, Nowarski R, Starnbach MN, Shi H, Surana NK, An D, Wu C, Huh JR, Rao M, Chiu IM. Gut-Innervating Nociceptor Neurons Regulate Peyer's Patch Microfold Cells and SFB Levels to Mediate Salmonella Host Defense. Cell 2020; 180:33-49.e22. [PMID: 31813624 PMCID: PMC6954329 DOI: 10.1016/j.cell.2019.11.014] [Citation(s) in RCA: 234] [Impact Index Per Article: 46.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 09/08/2019] [Accepted: 11/12/2019] [Indexed: 12/30/2022]
Abstract
Gut-innervating nociceptor sensory neurons respond to noxious stimuli by initiating protective responses including pain and inflammation; however, their role in enteric infections is unclear. Here, we find that nociceptor neurons critically mediate host defense against the bacterial pathogen Salmonella enterica serovar Typhimurium (STm). Dorsal root ganglia nociceptors protect against STm colonization, invasion, and dissemination from the gut. Nociceptors regulate the density of microfold (M) cells in ileum Peyer's patch (PP) follicle-associated epithelia (FAE) to limit entry points for STm invasion. Downstream of M cells, nociceptors maintain levels of segmentous filamentous bacteria (SFB), a gut microbe residing on ileum villi and PP FAE that mediates resistance to STm infection. TRPV1+ nociceptors directly respond to STm by releasing calcitonin gene-related peptide (CGRP), a neuropeptide that modulates M cells and SFB levels to protect against Salmonella infection. These findings reveal a major role for nociceptor neurons in sensing and defending against enteric pathogens.
Collapse
Affiliation(s)
- Nicole Y Lai
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Melissa A Musser
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA; Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
| | | | - Pankaj Baral
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Amanda Jacobson
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Pingchuan Ma
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - David E Potts
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Zuojia Chen
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA; Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Donggi Paik
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Salima Soualhi
- Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
| | - Yiqing Yan
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Aditya Misra
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Kaitlin Goldstein
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | | | - Anja Nordstrom
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Kisha N Sivanathan
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Antonia Wallrapp
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Vijay K Kuchroo
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Roni Nowarski
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | | | - Hailian Shi
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA; Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Neeraj K Surana
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA; Department of Pediatrics, Duke University, Durham, NC 27710, USA; Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27710, USA; Department of Immunology, Duke University, Durham, NC 27710, USA
| | - Dingding An
- Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
| | - Chuan Wu
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA; Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jun R Huh
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Meenakshi Rao
- Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
| | - Isaac M Chiu
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
150
|
Jarret A, Jackson R, Duizer C, Healy ME, Zhao J, Rone JM, Bielecki P, Sefik E, Roulis M, Rice T, Sivanathan KN, Zhou T, Solis AG, Honcharova-Biletska H, Vélez K, Hartner S, Low JS, Qu R, de Zoete MR, Palm NW, Ring AM, Weber A, Moor AE, Kluger Y, Nowarski R, Flavell RA. Enteric Nervous System-Derived IL-18 Orchestrates Mucosal Barrier Immunity. Cell 2020; 180:50-63.e12. [PMID: 31923399 PMCID: PMC7339937 DOI: 10.1016/j.cell.2019.12.016] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 11/01/2019] [Accepted: 12/12/2019] [Indexed: 12/18/2022]
Abstract
Mucosal barrier immunity is essential for the maintenance of the commensal microflora and combating invasive bacterial infection. Although immune and epithelial cells are thought to be the canonical orchestrators of this complex equilibrium, here, we show that the enteric nervous system (ENS) plays an essential and non-redundant role in governing the antimicrobial protein (AMP) response. Using confocal microscopy and single-molecule fluorescence in situ mRNA hybridization (smFISH) studies, we observed that intestinal neurons produce the pleiotropic cytokine IL-18. Strikingly, deletion of IL-18 from the enteric neurons alone, but not immune or epithelial cells, rendered mice susceptible to invasive Salmonella typhimurium (S.t.) infection. Mechanistically, unbiased RNA sequencing and single-cell sequencing revealed that enteric neuronal IL-18 is specifically required for homeostatic goblet cell AMP production. Together, we show that neuron-derived IL-18 signaling controls tissue-wide intestinal immunity and has profound consequences on the mucosal barrier and invasive bacterial killing.
Collapse
Affiliation(s)
- Abigail Jarret
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Ruaidhrí Jackson
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA.
| | - Coco Duizer
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Marc E Healy
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich 8091, Switzerland; Institute of Molecular Cancer Research, University of Zurich, Zurich 8057, Switzerland
| | - Jun Zhao
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA; Program of Computational Biology and Bioinformatics, Yale University, New Haven, CT 06520, USA; Department of Pathology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Joseph M Rone
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Piotr Bielecki
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Esen Sefik
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Manolis Roulis
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Tyler Rice
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Kisha N Sivanathan
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Ting Zhou
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Angel G Solis
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Hanna Honcharova-Biletska
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich 8091, Switzerland
| | - Karelia Vélez
- Institute of Molecular Cancer Research, University of Zurich, Zurich 8057, Switzerland
| | - Saskia Hartner
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA; University of Vienna, Universitätsring 1, Wien 1010, Austria
| | - Jun Siong Low
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Rihao Qu
- Program of Computational Biology and Bioinformatics, Yale University, New Haven, CT 06520, USA; Department of Pathology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Marcel R de Zoete
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Noah W Palm
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Aaron M Ring
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Achim Weber
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich 8091, Switzerland; Institute of Molecular Cancer Research, University of Zurich, Zurich 8057, Switzerland
| | - Andreas E Moor
- Institute of Molecular Cancer Research, University of Zurich, Zurich 8057, Switzerland
| | - Yuval Kluger
- Program of Computational Biology and Bioinformatics, Yale University, New Haven, CT 06520, USA; Department of Pathology, Yale School of Medicine, New Haven, CT 06510, USA; Applied Mathematics Program, Yale University, New Haven, CT 06511, USA
| | - Roni Nowarski
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA.
| | - Richard A Flavell
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA; Howard Hughes Medical Institute, Yale University, New Haven, CT 06520, USA.
| |
Collapse
|