1
|
Zhang X, Hartung JE, Gold MS. Persistent (Na v 1.9) sodium currents in human dorsal root ganglion neurons. Pain 2025; 166:448-459. [PMID: 39297710 PMCID: PMC11723807 DOI: 10.1097/j.pain.0000000000003394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 07/07/2024] [Indexed: 10/12/2024]
Abstract
ABSTRACT Na v 1.9 is of interest to the pain community for a number of reasons, including the human mutations in the gene encoding Na v 1.9, SCN11a , that are associated with both pain and loss of pain phenotypes. However, because much of what we know about the biophysical properties of Na v 1.9 has been learned through the study of rodent sensory neurons, and there is only 76% identity between human and rodent homologs of SCN11a , there is reason to suggest that there may be differences in the biophysical properties of the channels in human and rodent sensory neurons, and consequently, the contribution of these channels to the control of sensory neuron excitability, if not pain. Thus, the purpose of this study was to characterize Na v 1.9 currents in human sensory neurons and compare the properties of these currents with those in rat sensory neurons recorded under identical conditions. Whole-cell patch clamp techniques were used to record Na v 1.9 currents in isolated sensory neurons in vitro. Our results indicate that several of the core biophysical properties of the currents, including persistence and a low threshold for activation, are conserved across species. However, we noted a number of potentially important differences between the currents in human and rat sensory neurons including a lower threshold for activation, higher threshold for inactivation, slower deactivation, and faster recovery from slow inactivation. Human Na v 1.9 was inhibited by inflammatory mediators, whereas rat Na v 1.9 was potentiated. Our results may have implications for the role of Na v 1.9 in sensory, if not nociceptive signaling.
Collapse
Affiliation(s)
- Xiulin Zhang
- Department of Neurobiology, and the Pittsburgh Center for Pain Research University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Urology, the Second Hospital of Shandong University, 250032, P.R. China
| | - Jane E Hartung
- Department of Neurobiology, and the Pittsburgh Center for Pain Research University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Michael S Gold
- Department of Neurobiology, and the Pittsburgh Center for Pain Research University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
2
|
Bertomeu JB, Fioravanço LP, Ramis TR, Godinho DB, Nascimento AS, Lima GC, Furian AF, Oliveira MS, Fighera MR, Royes LFF. The Role of Ion-Transporting Proteins on Crosstalk Between the Skeletal Muscle and Central Nervous Systems Elicited by Physical Exercise. Mol Neurobiol 2024:10.1007/s12035-024-04613-7. [PMID: 39578339 DOI: 10.1007/s12035-024-04613-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 11/05/2024] [Indexed: 11/24/2024]
Abstract
A paradigm shift in the understanding of bidirectional interactions between peripheral and central nervous systems is essential for development of rehabilitation and preventive interventions based on physical exercise. Although a causal relationship has not been completely established, modulation of voltage-dependent ion channels (Ca2+, Cl-, K+, Na+, lactate-, H+) in skeletal and neuronal cells provides opportunities to maintain force production during exercise and reduce the risk of disease. However, there are caveats to consider when interpreting the effects of physical exercise on this bidirectional axis, since exercise protocol details (e.g., duration and intensity) have variable effects on this crosstalk. Therefore, an integrative perspective of the skeletal muscle and brain's communication pathway is discussed, and the role of physical exercise on such communication highway is explained in this review.
Collapse
Affiliation(s)
- Judit Borràs Bertomeu
- Graduate Program in Biological Sciences: Toxicological Biochemistry, Center in Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS, Brazil
- Exercise Biochemistry Laboratory (BIOEX), Department of Sports Methods and Techniques, Physical Education and Sports Center, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Letícia Paiva Fioravanço
- Graduate Program in Biological Sciences: Toxicological Biochemistry, Center in Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS, Brazil
- Exercise Biochemistry Laboratory (BIOEX), Department of Sports Methods and Techniques, Physical Education and Sports Center, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Thiago Rozales Ramis
- Graduate Program in Biological Sciences: Toxicological Biochemistry, Center in Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS, Brazil
- Exercise Biochemistry Laboratory (BIOEX), Department of Sports Methods and Techniques, Physical Education and Sports Center, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Douglas Buchmann Godinho
- Graduate Program in Biological Sciences: Toxicological Biochemistry, Center in Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS, Brazil
- Exercise Biochemistry Laboratory (BIOEX), Department of Sports Methods and Techniques, Physical Education and Sports Center, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Alexandre Seixas Nascimento
- Graduate Program in Biological Sciences: Toxicological Biochemistry, Center in Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS, Brazil
- Exercise Biochemistry Laboratory (BIOEX), Department of Sports Methods and Techniques, Physical Education and Sports Center, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Gabriel Corrêa Lima
- Graduate Program in Biological Sciences: Toxicological Biochemistry, Center in Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS, Brazil
- Exercise Biochemistry Laboratory (BIOEX), Department of Sports Methods and Techniques, Physical Education and Sports Center, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Ana Flavia Furian
- Graduate Program in Pharmacology, Health Sciences Center, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Mauro Schneider Oliveira
- Graduate Program in Pharmacology, Health Sciences Center, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Michele Rechia Fighera
- Graduate Program in Biological Sciences: Toxicological Biochemistry, Center in Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS, Brazil
- Exercise Biochemistry Laboratory (BIOEX), Department of Sports Methods and Techniques, Physical Education and Sports Center, Federal University of Santa Maria, Santa Maria, RS, Brazil
- Laboratory of Experimental and Clinical Neuropsychiatry, Department of Neuropsychiatry, Health Sciences Center, Federal University of Santa Maria, Santa Maria, RS, 97105-900, Brazil
| | - Luiz Fernando Freire Royes
- Graduate Program in Biological Sciences: Toxicological Biochemistry, Center in Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS, Brazil.
- Exercise Biochemistry Laboratory (BIOEX), Department of Sports Methods and Techniques, Physical Education and Sports Center, Federal University of Santa Maria, Santa Maria, RS, Brazil.
- Department of Sports Methods and Techniques, Center of Physical Education and , Sports, Federal University of Santa Maria - UFSM, Santa Maria, RS, 97105-900, Brazil.
| |
Collapse
|
3
|
Xu S, Wang Y. Transient Receptor Potential Channels: Multiple Modulators of Peripheral Neuropathic Pain in Several Rodent Models. Neurochem Res 2024; 49:872-886. [PMID: 38281247 DOI: 10.1007/s11064-023-04087-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 11/22/2023] [Accepted: 12/16/2023] [Indexed: 01/30/2024]
Abstract
Neuropathic pain, a prevalent chronic condition in clinical settings, has attracted widespread societal attention. This condition is characterized by a persistent pain state accompanied by affective and cognitive disruptions, significantly impacting patients' quality of life. However, current clinical therapies fall short of addressing its complexity. Thus, exploring the underlying molecular mechanism of neuropathic pain and identifying potential targets for intervention is highly warranted. The transient receptor potential (TRP) receptors, a class of widely distributed channel proteins, in the nervous system, play a crucial role in sensory signaling, cellular calcium regulation, and developmental influences. TRP ion channels are also responsible for various sensory responses including heat, cold, pain, and stress. This review highlights recent advances in understanding TRPs in various rodent models of neuropathic pain, aiming to uncover potential therapeutic targets for clinical management.
Collapse
Affiliation(s)
- Songchao Xu
- Department of Anesthesiology, Beijing Friendship Hospital, Capital Medical University, No. 95, Yong'an Road, Xicheng District, Beijing, 100050, China
| | - Yun Wang
- Department of Anesthesiology, Beijing Friendship Hospital, Capital Medical University, No. 95, Yong'an Road, Xicheng District, Beijing, 100050, China.
| |
Collapse
|
4
|
Moore AA, Nelson M, Wickware C, Choi S, Moon G, Xiong E, Orta L, Brideau-Andersen A, Brin MF, Broide RS, Liedtke W, Moore C. OnabotulinumtoxinA effects on trigeminal nociceptors. Cephalalgia 2023; 43:3331024221141683. [PMID: 36751871 PMCID: PMC10652784 DOI: 10.1177/03331024221141683] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
BACKGROUND OnabotulinumtoxinA (onabotA) is approved globally for prevention of chronic migraine; however, the classical mechanism of action of onabotA in motor and autonomic neurons cannot fully explain the effectiveness of onabotulinumtoxinA in this sensory neurological disease. We sought to explore the direct effects of onabotulinumtoxinA on mouse trigeminal ganglion sensory neurons using an inflammatory soup-based model of sensitization. METHODS Primary cultured trigeminal ganglion neurons were pre-treated with inflammatory soup, then treated with onabotulinumtoxinA (2.75 pM). Treated neurons were used to examine transient receptor potential vanilloid subtype 1 and transient receptor potential ankyrin 1 cell-surface expression, calcium influx, and neuropeptide release. RESULTS We found that onabotulinumtoxinA cleaved synaptosomal-associated protein-25 kDa in cultured trigeminal ganglion neurons; synaptosomal-associated protein-25 kDa cleavage was enhanced by inflammatory soup pre-treatment, suggesting greater uptake of toxin under sensitized conditions. OnabotulinumtoxinA also prevented inflammatory soup-mediated increases in TRPV1 and TRPA1 cell-surface expression, without significantly altering TRPV1 or TRPA1 protein expression in unsensitized conditions. We observed similar inhibitory effects of onabotulinumtoxinA on TRP-mediated calcium influx and TRPV1- and TRPA1-mediated release of calcitonin gene-related peptide and prostaglandin 2 under sensitized, but not unsensitized control, conditions. CONCLUSIONS Our data deepen the understanding of the sensory mechanism of action of onabotulinumtoxinA and support the notion that, once endocytosed, the cytosolic light chain of onabotulinumtoxinA cleaves synaptosomal-associated protein-25 kDa to prevent soluble N-ethylmaleimide-sensitive factor attachment protein receptor-mediated processes more generally in motor, autonomic, and sensory neurons.
Collapse
Affiliation(s)
- Ashley A Moore
- Department of Neurology, Duke University, Durham, NC, USA
| | | | | | - Shinbe Choi
- Department of Neurology, Duke University, Durham, NC, USA
| | - Gene Moon
- Department of Neurology, Duke University, Durham, NC, USA
| | - Emma Xiong
- Department of Neurology, Duke University, Durham, NC, USA
| | - Lily Orta
- Department of Neurology, Duke University, Durham, NC, USA
| | | | - Mitchell F Brin
- Allergan, an AbbVie company, Irvine, CA, USA
- Department of Neurology, University of California, Irvine, Irvine, CA, USA
| | | | - Wolfgang Liedtke
- Department of Neurology, Duke University, Durham, NC, USA
- Department of Molecular Pathobiology – Dental Pain Research, New York University College of Dentistry, New York, NY, USA
| | - Carlene Moore
- Department of Neurology, Duke University, Durham, NC, USA
| |
Collapse
|
5
|
Tiwari MN, Hall BE, Ton AT, Ghetti R, Terse A, Amin N, Chung MK, Kulkarni AB. Activation of cyclin-dependent kinase 5 broadens action potentials in human sensory neurons. Mol Pain 2023; 19:17448069231218353. [PMID: 37982142 PMCID: PMC10687939 DOI: 10.1177/17448069231218353] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 11/06/2023] [Accepted: 11/10/2023] [Indexed: 11/21/2023] Open
Abstract
Chronic pain is one of the most devastating and unpleasant conditions, associated with many pathological states. Tissue or nerve injuries induce extensive neurobiological plasticity in nociceptive neurons, which leads to chronic pain. Recent studies suggest that cyclin-dependent kinase 5 (CDK5) in primary afferents is a key neuronal kinase that modulates nociception through phosphorylation under pathological conditions. However, the impact of the CDK5 on nociceptor activity especially in human sensory neurons is not known. To determine the CDK5-mediated regulation of human dorsal root ganglia (hDRG) neuronal properties, we have performed the whole-cell patch clamp recordings in neurons dissociated from hDRG. CDK5 activation induced by overexpression of p35 depolarized the resting membrane potential (RMP) and reduced the rheobase currents as compared to the control neurons. CDK5 activation changed the shape of the action potential (AP) by increasing AP -rise time, -fall time, and -half width. The application of a prostaglandin E2 (PG) and bradykinin (BK) cocktail in control hDRG neurons induced the depolarization of RMP and the reduction of rheobase currents along with increased AP rise time. However, PG and BK applications failed to induce any significant changes in the p35-overexpressing group. We conclude that, in dissociated hDRGs neurons, CDK5 activation through the overexpression of p35 broadens the AP and that CDK5 may play important roles in the modulation of AP properties in human primary afferents under the condition in which CDK5 is upregulated, contributing to chronic pain.
Collapse
Affiliation(s)
- Manindra Nath Tiwari
- Department of Neural and Pain Sciences, School of Dentistry, Program in Neuroscience, Center to Advance Chronic Pain Research, The University of Maryland, Baltimore, MD, United States
| | - Bradford E Hall
- Functional Genomics Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, United States
| | | | - Re Ghetti
- AnaBios, San Diego, CA, United States
| | - Anita Terse
- Functional Genomics Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, United States
| | - Niranjana Amin
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Man-Kyo Chung
- Department of Neural and Pain Sciences, School of Dentistry, Program in Neuroscience, Center to Advance Chronic Pain Research, The University of Maryland, Baltimore, MD, United States
| | - Ashok B Kulkarni
- Functional Genomics Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
6
|
Meerschaert KA, Edwards BS, Epouhe AY, Jefferson B, Friedman R, Babyok OL, Moy JK, Kehinde F, Liu C, Workman CJ, Vignali DAA, Albers KM, Koerber HR, Gold MS, Davis BM, Scheff NN, Saloman JL. Neuronally expressed PDL1, not PD1, suppresses acute nociception. Brain Behav Immun 2022; 106:233-246. [PMID: 36089217 PMCID: PMC10343937 DOI: 10.1016/j.bbi.2022.09.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 08/21/2022] [Accepted: 09/03/2022] [Indexed: 11/20/2022] Open
Abstract
PDL1 is a protein that induces immunosuppression by binding to PD1 expressed on immune cells. In line with historical studies, we found that membrane-bound PD1 expression was largely restricted to immune cells; PD1 was not detectable at either the mRNA or protein level in peripheral neurons using single neuron qPCR, immunolabeling and flow cytometry. However, we observed widespread expression of PDL1 in both sensory and sympathetic neurons that could have important implications for patients receiving immunotherapies targeting this pathway that include unexpected autonomic and sensory related effects. While signaling pathways downstream of PD1 are well established, little to no information is available regarding the intracellular signaling downstream of membrane-bound PDL1 (also known as reverse signaling). Here, we administered soluble PD1 to engage neuronally expressed PDL1 and found that PD1 significantly reduced nocifensive behaviors evoked by algogenic capsaicin. We used calcium imaging to examine the underlying neural mechanism of this reduction and found that exogenous PD1 diminished TRPV1-dependent calcium transients in dissociated sensory neurons. Furthermore, we observed a reduction in membrane expression of TRPV1 following administration of PD1. Exogenous PD1 had no effect on pain-related behaviors in sensory neuron specific PDL1 knockout mice. These data indicate that neuronal PDL1 activation is sufficient to modulate sensitivity to noxious stimuli and as such, may be an important homeostatic mechanism for regulating acute nociception.
Collapse
Affiliation(s)
- Kimberly A Meerschaert
- Pittsburgh Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Center for Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Brian S Edwards
- Pittsburgh Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Ariel Y Epouhe
- Pittsburgh Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Center for Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Bahiyyah Jefferson
- Pittsburgh Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Robert Friedman
- Pittsburgh Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Olivia L Babyok
- Pittsburgh Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Center for Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Jamie K Moy
- Pittsburgh Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Center for Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Faith Kehinde
- Pittsburgh Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Chang Liu
- Department of Immunology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States; Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, United States
| | - Creg J Workman
- Department of Immunology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States; Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, United States
| | - Dario A A Vignali
- Department of Immunology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States; Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, United States; Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, United States
| | - Kathryn M Albers
- Pittsburgh Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Center for Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - H Richard Koerber
- Pittsburgh Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Center for Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Michael S Gold
- Pittsburgh Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Center for Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Brian M Davis
- Pittsburgh Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Center for Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Nicole N Scheff
- Pittsburgh Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Center for Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Biobehavioral Cancer Control Program, UPMC Hillman Cancer Center, Pittsburgh, PA, United States
| | - Jami L Saloman
- Pittsburgh Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Center for Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Biobehavioral Cancer Control Program, UPMC Hillman Cancer Center, Pittsburgh, PA, United States; Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.
| |
Collapse
|
7
|
Pre-Synaptic GABAA in NaV1.8+ Primary Afferents Is Required for the Development of Punctate but Not Dynamic Mechanical Allodynia following CFA Inflammation. Cells 2022; 11:cells11152390. [PMID: 35954234 PMCID: PMC9368720 DOI: 10.3390/cells11152390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/26/2022] [Accepted: 07/27/2022] [Indexed: 01/27/2023] Open
Abstract
Hypersensitivity to mechanical stimuli is a cardinal symptom of neuropathic and inflammatory pain. A reduction in spinal inhibition is generally considered a causal factor in the development of mechanical hypersensitivity after injury. However, the extent to which presynaptic inhibition contributes to altered spinal inhibition is less well established. Here, we used conditional deletion of GABAA in NaV1.8-positive sensory neurons (Scn10aCre;Gabrb3fl/fl) to manipulate selectively presynaptic GABAergic inhibition. Behavioral testing showed that the development of inflammatory punctate allodynia was mitigated in mice lacking pre-synaptic GABAA. Dorsal horn cellular circuits were visualized in single slices using stimulus-tractable dual-labelling of c-fos mRNA for punctate and the cognate c-Fos protein for dynamic mechanical stimulation. This revealed a substantial reduction in the number of cells activated by punctate stimulation in mice lacking presynaptic GABAA and an approximate 50% overlap of the punctate with the dynamic circuit, the relative percentage of which did not change following inflammation. The reduction in dorsal horn cells activated by punctate stimuli was equally prevalent in parvalbumin- and calretinin-positive cells and across all laminae I–V, indicating a generalized reduction in spinal input. In peripheral DRG neurons, inflammation following complete Freund’s adjuvant (CFA) led to an increase in axonal excitability responses to GABA, suggesting that presynaptic GABA effects in NaV1.8+ afferents switch from inhibition to excitation after CFA. In the days after inflammation, presynaptic GABAA in NaV1.8+ nociceptors constitutes an “open gate” pathway allowing mechanoreceptors responding to punctate mechanical stimulation access to nociceptive dorsal horn circuits.
Collapse
|
8
|
Sivakumar D, Ramli R. GABAergic signalling in modulation of dental pain. Eur J Pharmacol 2022; 924:174958. [DOI: 10.1016/j.ejphar.2022.174958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/07/2022] [Accepted: 04/11/2022] [Indexed: 11/30/2022]
|
9
|
Sadler KE, Mogil JS, Stucky CL. Innovations and advances in modelling and measuring pain in animals. Nat Rev Neurosci 2022; 23:70-85. [PMID: 34837072 PMCID: PMC9098196 DOI: 10.1038/s41583-021-00536-7] [Citation(s) in RCA: 84] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/20/2021] [Indexed: 12/12/2022]
Abstract
Best practices in preclinical algesiometry (pain behaviour testing) have shifted over the past decade as a result of technological advancements, the continued dearth of translational progress and the emphasis that funding institutions and journals have placed on rigour and reproducibility. Here we describe the changing trends in research methods by analysing the methods reported in preclinical pain publications from the past 40 years, with a focus on the last 5 years. We also discuss how the status quo may be hampering translational success. This discussion is centred on four fundamental decisions that apply to every pain behaviour experiment: choice of subject (model organism), choice of assay (pain-inducing injury), laboratory environment and choice of outcome measures. Finally, we discuss how human tissues, which are increasingly accessible, can be used to validate the translatability of targets and mechanisms identified in animal pain models.
Collapse
Affiliation(s)
- Katelyn E Sadler
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Jeffrey S Mogil
- Department of Psychology, McGill University, Montreal, QC, Canada
- Department of Anesthesia, McGill University, Montreal, QC, Canada
| | - Cheryl L Stucky
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
10
|
Hornung RS, Raut NGR, Cantu DJ, Lockhart LM, Averitt DL. Sigma-1 receptors and progesterone metabolizing enzymes in nociceptive sensory neurons of the female rat trigeminal ganglia: A neural substrate for the antinociceptive actions of progesterone. Mol Pain 2022; 18:17448069211069255. [PMID: 35040378 PMCID: PMC8777333 DOI: 10.1177/17448069211069255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Orofacial pain disorders are predominately experienced by women. Progesterone, a major ovarian hormone, is neuroprotective and antinociceptive. We recently reported that progesterone attenuates estrogen-exacerbated orofacial pain behaviors, yet it remains unclear what anatomical substrate underlies progesterone's activity in the trigeminal system. Progesterone has been reported to exert protective effects through actions at intracellular progesterone receptors (iPR), membrane-progesterone receptors (mPR), or sigma 1 receptors (Sig-1R). Of these, the iPR and Sig-1R have been reported to have a role in pain. Progesterone can also have antinociceptive effects through its metabolite, allopregnanolone. Two enzymes, 5α-reductase and 3α-hydroxysteroid dehydrogenase (3α-HSD), are required for the metabolism of progesterone to allopregnanolone. Both progesterone and allopregnanolone rapidly attenuate pain sensitivity, implicating action of either progesterone at Sig-1R and/or conversion to allopregnanolone which targets GABAA receptors. In the present study, we investigated whether Sig-1 Rs are expressed in nociceptors within the trigeminal ganglia of cycling female rats and whether the two enzymes required for progesterone metabolism to allopregnanolone, 5α-reductase and 3α-hydroxysteroid dehydrogenase, are also present. Adult female rats from each stage of the estrous cycle were rapidly decapitated and the trigeminal ganglia collected. Trigeminal ganglia were processed by either fluorescent immunochemistry or western blotting to for visualization and quantification of Sig-1R, 5α-reductase, and 3α-hydroxysteroid dehydrogenase. Here we report that Sig-1Rs and both enzymes involved in progesterone metabolism are highly expressed in a variety of nociceptive sensory neuron populations in the female rat trigeminal ganglia at similar levels across the four stages of the estrous cycle. These data indicate that trigeminal sensory neurons are an anatomical substrate for the reported antinociceptive activity of progesterone via Sig-1R and/or conversion to allopregnanolone.
Collapse
Affiliation(s)
| | | | - Daisy J Cantu
- Division of Biology, School of the Sciences, Texas Woman’s
University, Denton, TX, USA
| | - Lauren M Lockhart
- Division of Biology, School of the Sciences, Texas Woman’s
University, Denton, TX, USA
| | - Dayna L Averitt
- Division of Biology, School of the Sciences, Texas Woman’s
University, Denton, TX, USA
| |
Collapse
|
11
|
Bonalume V, Caffino L, Castelnovo LF, Faroni A, Liu S, Hu J, Milanese M, Bonanno G, Sohns K, Hoffmann T, De Col R, Schmelz M, Fumagalli F, Magnaghi V, Carr R. Axonal GABA A stabilizes excitability in unmyelinated sensory axons secondary to NKCC1 activity. J Physiol 2021; 599:4065-4084. [PMID: 34174096 DOI: 10.1113/jp279664] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 06/08/2021] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS GABA depolarized sural nerve axons and increased the electrical excitability of C-fibres via GABAA receptor. Axonal excitability responses to GABA increased monotonically with the rate of action potential firing. Action potential activity in unmyelinated C-fibres is coupled to Na-K-Cl cotransporter type 1 (NKCC1) loading of axonal chloride. Activation of axonal GABAA receptor stabilized C-fibre excitability during prolonged low frequency (2.5 Hz) firing. NKCC1 maintains intra-axonal chloride to provide feed-forward stabilization of C-fibre excitability and thus support sustained firing. ABSTRACT GABAA receptor (GABAA R)-mediated depolarization of dorsal root ganglia (DRG) axonal projections in the spinal dorsal horn is implicated in pre-synaptic inhibition. Inhibition, in this case, is predicated on an elevated intra-axonal chloride concentration and a depolarizing GABA response. In the present study, we report that the peripheral axons of DRG neurons are also depolarized by GABA and this results in an increase in the electrical excitability of unmyelinated C-fibre axons. GABAA R agonists increased axonal excitability, whereas GABA excitability responses were blocked by GABAA R antagonists and were absent in mice lacking the GABAA R β3 subunit selectively in DRG neurons (AdvillinCre or snsCre ). Under control conditions, excitability responses to GABA became larger at higher rates of electrical stimulation (0.5-2.5 Hz). However, during Na-K-Cl cotransporter type 1 (NKCC1) blockade, the electrical stimulation rate did not affect GABA response size, suggesting that NKCC1 regulation of axonal chloride is coupled to action potential firing. To examine this, activity-dependent conduction velocity slowing (activity-dependent slowing; ADS) was used to quantify C-fibre excitability loss during a 2.5 Hz challenge. ADS was reduced by GABAA R agonists and exacerbated by either GABAA R antagonists, β3 deletion or NKCC1 blockade. This illustrates that activation of GABAA R stabilizes C-fibre excitability during sustained firing. We posit that NKCC1 acts in a feed-forward manner to maintain an elevated intra-axonal chloride in C-fibres during ongoing firing. The resulting chloride gradient can be utilized by GABAA R to stabilize axonal excitability. The data imply that therapeutic strategies targeting axonal chloride regulation at peripheral loci of pain and itch may curtail aberrant firing in C-fibres.
Collapse
Affiliation(s)
- Veronica Bonalume
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Lucia Caffino
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Luca F Castelnovo
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
- Marine Science Institute, University of Texas at Austin, Port Aransas, TX, USA
| | - Alessandro Faroni
- Blond McIndoe Laboratories, Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Sheng Liu
- Institute of Pharmacology, Heidelberg University, Mannheim, Germany
| | - Jing Hu
- Institute of Pharmacology, Heidelberg University, Mannheim, Germany
| | - Marco Milanese
- Department of Pharmacy (DIFAR), Pharmacology and Toxicology Unit, Università degli Studi di Genova, Genova, Italy
| | - Giambattista Bonanno
- Department of Pharmacy (DIFAR), Pharmacology and Toxicology Unit, Università degli Studi di Genova, Genova, Italy
- Ospedale Policlinico San Martino, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Genova, Italy
| | - Kyra Sohns
- Experimental Pain Research, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Tal Hoffmann
- Institute for Physiology and Pathophysiology, Friedrich-Alexander University, Erlangen, Germany
| | - Roberto De Col
- Experimental Pain Research, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Martin Schmelz
- Experimental Pain Research, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Fabio Fumagalli
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Valerio Magnaghi
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Richard Carr
- Experimental Pain Research, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
12
|
Chrysostomidou L, Cooper AH, Weir GA. Cellular models of pain: New technologies and their potential to progress preclinical research. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2021; 10:100063. [PMID: 34977426 PMCID: PMC8683679 DOI: 10.1016/j.ynpai.2021.100063] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 05/12/2021] [Accepted: 05/15/2021] [Indexed: 01/16/2023]
Abstract
Human sensory neurons can reduce the translational gap in analgesic development. Access to dorsal root ganglion (hDRG) neurons is increasing. Diverse sensory neuron subtypes can now be generated via stem cell technology. Advances of these technologies will improve our understanding of human nociception.
In vitro models fill a vital niche in preclinical pain research, allowing detailed study of molecular pathways, and in the case of humanised systems, providing a translational bridge between in vivo animal models and human patients. Significant advances in cellular technology available to basic pain researchers have occurred in the last decade, including developing protocols to differentiate sensory neuron-like cells from stem cells and greater access to human dorsal root ganglion tissue. In this review, we discuss the use of both models in preclinical pain research: What can a human sensory neuron in a dish tell us that rodent in vivo models cannot? How similar are these models to their endogenous counterparts, and how should we judge them? What limitations do we need to consider? How can we leverage cell models to improve translational success? In vitro human sensory neuron models equip pain researchers with a valuable tool to investigate human nociception. With continual development, consideration for their advantages and limitations, and effective integration with other experimental strategies, they could become a driving force for the pain field's advancement.
Collapse
Affiliation(s)
- Lina Chrysostomidou
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Andrew H Cooper
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Greg A Weir
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
13
|
Wang C, Hao H, He K, An Y, Pu Z, Gamper N, Zhang H, Du X. Neuropathic Injury-Induced Plasticity of GABAergic System in Peripheral Sensory Ganglia. Front Pharmacol 2021; 12:702218. [PMID: 34385921 PMCID: PMC8354334 DOI: 10.3389/fphar.2021.702218] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 06/25/2021] [Indexed: 12/30/2022] Open
Abstract
GABA is a major inhibitory neurotransmitter in the mammalian central nervous system (CNS). Inhibitory GABAA channel circuits in the dorsal spinal cord are the gatekeepers of the nociceptive input from the periphery to the CNS. Weakening of these spinal inhibitory mechanisms is a hallmark of chronic pain. Yet, recent studies have suggested the existence of an earlier GABAergic “gate” within the peripheral sensory ganglia. In this study, we performed systematic investigation of plastic changes of the GABA-related proteins in the dorsal root ganglion (DRG) in the process of neuropathic pain development. We found that chronic constriction injury (CCI) induced general downregulation of most GABAA channel subunits and the GABA-producing enzyme, glutamate decarboxylase, consistent with the weakening of the GABAergic inhibition at the periphery. Strikingly, the α5 GABAA subunit was consistently upregulated. Knock-down of the α5 subunit in vivo moderately alleviated neuropathic hyperalgesia. Our findings suggest that while the development of neuropathic pain is generally accompanied by weakening of the peripheral GABAergic system, the α5 GABAA subunit may have a unique pro-algesic role and, hence, might represent a new therapeutic target.
Collapse
Affiliation(s)
- Caixue Wang
- The Key Laboratory of Neural and Vascular Biology, The Key Laboratory of New Drug Pharmacology and Toxicology, Department of Pharmacology, Ministry of Education, Hebei Medical University, Shijiazhuang, China
| | - Han Hao
- The Key Laboratory of Neural and Vascular Biology, The Key Laboratory of New Drug Pharmacology and Toxicology, Department of Pharmacology, Ministry of Education, Hebei Medical University, Shijiazhuang, China
| | - Kaitong He
- The Key Laboratory of Neural and Vascular Biology, The Key Laboratory of New Drug Pharmacology and Toxicology, Department of Pharmacology, Ministry of Education, Hebei Medical University, Shijiazhuang, China
| | - Yating An
- The Key Laboratory of Neural and Vascular Biology, The Key Laboratory of New Drug Pharmacology and Toxicology, Department of Pharmacology, Ministry of Education, Hebei Medical University, Shijiazhuang, China
| | - Zeyao Pu
- The Key Laboratory of Neural and Vascular Biology, The Key Laboratory of New Drug Pharmacology and Toxicology, Department of Pharmacology, Ministry of Education, Hebei Medical University, Shijiazhuang, China
| | - Nikita Gamper
- The Key Laboratory of Neural and Vascular Biology, The Key Laboratory of New Drug Pharmacology and Toxicology, Department of Pharmacology, Ministry of Education, Hebei Medical University, Shijiazhuang, China.,Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Hailin Zhang
- The Key Laboratory of Neural and Vascular Biology, The Key Laboratory of New Drug Pharmacology and Toxicology, Department of Pharmacology, Ministry of Education, Hebei Medical University, Shijiazhuang, China
| | - Xiaona Du
- The Key Laboratory of Neural and Vascular Biology, The Key Laboratory of New Drug Pharmacology and Toxicology, Department of Pharmacology, Ministry of Education, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
14
|
Middleton SJ, Barry AM, Comini M, Li Y, Ray PR, Shiers S, Themistocleous AC, Uhelski ML, Yang X, Dougherty PM, Price TJ, Bennett DL. Studying human nociceptors: from fundamentals to clinic. Brain 2021; 144:1312-1335. [PMID: 34128530 PMCID: PMC8219361 DOI: 10.1093/brain/awab048] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 11/26/2020] [Accepted: 12/08/2020] [Indexed: 12/14/2022] Open
Abstract
Chronic pain affects one in five of the general population and is the third most important cause of disability-adjusted life-years globally. Unfortunately, treatment remains inadequate due to poor efficacy and tolerability. There has been a failure in translating promising preclinical drug targets into clinic use. This reflects challenges across the whole drug development pathway, from preclinical models to trial design. Nociceptors remain an attractive therapeutic target: their sensitization makes an important contribution to many chronic pain states, they are located outside the blood-brain barrier, and they are relatively specific. The past decade has seen significant advances in the techniques available to study human nociceptors, including: the use of corneal confocal microscopy and biopsy samples to observe nociceptor morphology, the culture of human nociceptors (either from surgical or post-mortem tissue or using human induced pluripotent stem cell derived nociceptors), the application of high throughput technologies such as transcriptomics, the in vitro and in vivo electrophysiological characterization through microneurography, and the correlation with pain percepts provided by quantitative sensory testing. Genome editing in human induced pluripotent stem cell-derived nociceptors enables the interrogation of the causal role of genes in the regulation of nociceptor function. Both human and rodent nociceptors are more heterogeneous at a molecular level than previously appreciated, and while we find that there are broad similarities between human and rodent nociceptors there are also important differences involving ion channel function, expression, and cellular excitability. These technological advances have emphasized the maladaptive plastic changes occurring in human nociceptors following injury that contribute to chronic pain. Studying human nociceptors has revealed new therapeutic targets for the suppression of chronic pain and enhanced repair. Cellular models of human nociceptors have enabled the screening of small molecule and gene therapy approaches on nociceptor function, and in some cases have enabled correlation with clinical outcomes. Undoubtedly, challenges remain. Many of these techniques are difficult to implement at scale, current induced pluripotent stem cell differentiation protocols do not generate the full diversity of nociceptor populations, and we still have a relatively poor understanding of inter-individual variation in nociceptors due to factors such as age, sex, or ethnicity. We hope our ability to directly investigate human nociceptors will not only aid our understanding of the fundamental neurobiology underlying acute and chronic pain but also help bridge the translational gap.
Collapse
Affiliation(s)
- Steven J Middleton
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Allison M Barry
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Maddalena Comini
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Yan Li
- Department of Anesthesia and Pain Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Pradipta R Ray
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Stephanie Shiers
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Andreas C Themistocleous
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK.,Brain Function Research Group, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2000, South Africa
| | - Megan L Uhelski
- Department of Anesthesia and Pain Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Xun Yang
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Patrick M Dougherty
- Brain Function Research Group, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2000, South Africa
| | - Theodore J Price
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX 75080, USA
| | - David L Bennett
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| |
Collapse
|
15
|
Abstract
Preclinical evidence has highlighted the importance of the μ-opioid peptide (MOP) receptor on primary afferents for both the analgesic actions of MOP receptor agonists, as well as the development of tolerance, if not opioid-induced hyperalgesia. There is also growing interest in targeting other opioid peptide receptor subtypes (δ-opioid peptide [DOP], κ-opioid peptide [KOP], and nociceptin/orphanin-FQ opioid peptide [NOP]) on primary afferents, as alternatives to MOP receptors, which may not be associated with as many deleterious side effects. Nevertheless, results from several recent studies of human sensory neurons indicate that although there are many similarities between rodent and human sensory neurons, there may also be important differences. Thus, the purpose of this study was to assess the distribution of opioid receptor subtypes among human sensory neurons. A combination of pharmacology, patch-clamp electrophysiology, Ca imaging, and single-cell semiquantitative polymerase chain reaction was used. Our results suggest that functional MOP-like receptors are present in approximately 50% of human dorsal root ganglion neurons. δ-opioid peptide-like receptors were detected in a subpopulation largely overlapping that with MOP-like receptors. Furthermore, KOP-like and NOP-like receptors are detected in a large proportion (44% and 40%, respectively) of human dorsal root ganglion neurons with KOP receptors also overlapping with MOP receptors at a high rate (83%). Our data confirm that all 4 opioid receptor subtypes are present and functional in human sensory neurons, where the overlap of DOP, KOP, and NOP receptors with MOP receptors suggests that activation of these other opioid receptor subtypes may also have analgesic efficacy.
Collapse
|
16
|
Nickolls AR, Lee MM, Espinoza DF, Szczot M, Lam RM, Wang Q, Beers J, Zou J, Nguyen MQ, Solinski HJ, AlJanahi AA, Johnson KR, Ward ME, Chesler AT, Bönnemann CG. Transcriptional Programming of Human Mechanosensory Neuron Subtypes from Pluripotent Stem Cells. Cell Rep 2021; 30:932-946.e7. [PMID: 31968264 PMCID: PMC7059559 DOI: 10.1016/j.celrep.2019.12.062] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 09/17/2019] [Accepted: 12/16/2019] [Indexed: 12/17/2022] Open
Abstract
Efficient and homogeneous in vitro generation of peripheral sensory neurons may provide a framework for novel drug screening platforms and disease models of touch and pain. We discover that, by ovesssrexpressing NGN2 and BRN3A, human pluripotent stem cells can be transcriptionally programmed to differentiate into a surprisingly uniform culture of cold- and mechano-sensing neurons. Although such a neuronal subtype is not found in mice, we identify molecular evidence for its existence in human sensory ganglia. Combining NGN2 and BRN3A programming with neural crest patterning, we produce two additional populations of sensory neurons, including a specialized touch receptor neuron subtype. Finally, we apply this system to model a rare inherited sensory disorder of touch and proprioception caused by inactivating mutations in PIEZO2. Together, these findings establish an approach to specify distinct sensory neuron subtypes in vitro, underscoring the utility of stem cell technology to capture human-specific features of physiology and disease. Nickolls et al. develop a method, using human stem cells, to generate specific types of sensory neurons that detect cold temperature and mechanical force. This approach uncovers a class of neuron found in humans, but not mice, and enables the modeling of a rare sensory disorder of touch and proprioception.
Collapse
Affiliation(s)
- Alec R Nickolls
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA; Department of Neuroscience, Brown University, Providence, RI 02912, USA
| | - Michelle M Lee
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - David F Espinoza
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Marcin Szczot
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ruby M Lam
- Department of Neuroscience, Brown University, Providence, RI 02912, USA; National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Qi Wang
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jeanette Beers
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jizhong Zou
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Minh Q Nguyen
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Hans J Solinski
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Aisha A AlJanahi
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kory R Johnson
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Michael E Ward
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Alexander T Chesler
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Carsten G Bönnemann
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
17
|
Colciago A, Bonalume V, Melfi V, Magnaghi V. Genomic and Non-genomic Action of Neurosteroids in the Peripheral Nervous System. Front Neurosci 2020; 14:796. [PMID: 32848567 PMCID: PMC7403499 DOI: 10.3389/fnins.2020.00796] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 07/07/2020] [Indexed: 01/12/2023] Open
Abstract
Since the former evidence of biologic actions of neurosteroids in the central nervous system, also the peripheral nervous system (PNS) was reported as a structure affected by these substances. Indeed, neurosteroids are synthesized and active in the PNS, exerting many important actions on the different cell types of this system. PNS is a target for neurosteroids, in their native form or as metabolites. In particular, old and recent evidence indicates that the progesterone metabolite allopregnanolone possesses important functions in the PNS, thus contributing to its physiologic processes. In this review, we will survey the more recent findings on the genomic and non-genomic actions of neurosteroids in nerves, ganglia, and cells forming the PNS, focusing on the mechanisms regulating the peripheral neuron-glial crosstalk. Then, we will refer to the physiopathological significance of the neurosteroid signaling disturbances in the PNS, in to identify new molecular targets for promising pharmacotherapeutic approaches.
Collapse
Affiliation(s)
- Alessandra Colciago
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Veronica Bonalume
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Valentina Melfi
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Valerio Magnaghi
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
18
|
Dehkordi FM, Kaboutari J, Zendehdel M, Javdani M. The antinociceptive effect of artemisinin on the inflammatory pain and role of GABAergic and opioidergic systems. Korean J Pain 2019; 32:160-167. [PMID: 31257824 PMCID: PMC6615442 DOI: 10.3344/kjp.2019.32.3.160] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 03/11/2019] [Accepted: 03/19/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Pain is a complex mechanism which involves different systems, including the opioidergic and GABAergic systems. Due to the side effects of chemical analgesic agents, attention toward natural agents have been increased. Artemisinin is an herbal compound with widespread modern and traditional therapeutic indications, which its interaction with the GABAergic system and antinoniceptive effects on neuropathic pain have shown. Therefore, this study was designed to evaluate the antinociceptive effects of artemisinin during inflammatory pain and interaction with the GABAergic and opioidergic systems by using a writhing response test. METHODS On the whole, 198 adult male albino mice were used in 4 experiments, including 9 groups (n = 6) each with three replicates, by intraperitoneal (i.p.) administration of artemisinin (2.5, 5, and 10 mg/kg), naloxone (2 mg/kg), bicuculline (2 mg/kg), saclofen (2 mg/kg), indomethacin (5 mg/kg), and ethanol (10 mL/kg). Writhing test responses were induced by i.p. injection of 10 mL/kg of 0.6% acetic acid, and the percentage of writhing inhibition was recorded. RESULTS Results showed significant dose dependent anti-nociceptive effects from artemisinin which, at a 10 mg/kg dose, was statistically similar to indomethacin. Neither saclofen nor naloxone had antinociceptive effects and did not antagonize antinociceptive effects of artemisinin, whereas bicuculline significantly inhibited the antinocicptive effect of artemisinin. CONCLUSIONS It seems that antinocicptive effects of artemisinin are mediated by GABAA receptors.
Collapse
Affiliation(s)
- Faraz Mahdian Dehkordi
- Department of Basic Sciences, Faculty of Veterinary Medicine, Shahrekord University, Shahrekord,
Iran
| | - Jahangir Kaboutari
- Department of Basic Sciences, Faculty of Veterinary Medicine, Shahrekord University, Shahrekord,
Iran
| | - Morteza Zendehdel
- Department of Physiology, Faculty of Veterinary Medicine, University of Tehran, Tehran,
Iran
| | - Moosa Javdani
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Shahrekord University, Shahrekord,
Iran
| |
Collapse
|
19
|
Haberberger RV, Barry C, Dominguez N, Matusica D. Human Dorsal Root Ganglia. Front Cell Neurosci 2019; 13:271. [PMID: 31293388 PMCID: PMC6598622 DOI: 10.3389/fncel.2019.00271] [Citation(s) in RCA: 142] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 06/04/2019] [Indexed: 12/14/2022] Open
Abstract
Sensory neurons with cell bodies situated in dorsal root ganglia convey information from external or internal sites of the body such as actual or potential harm, temperature or muscle length to the central nervous system. In recent years, large investigative efforts have worked toward an understanding of different types of DRG neurons at transcriptional, translational, and functional levels. These studies most commonly rely on data obtained from laboratory animals. Human DRG, however, have received far less investigative focus over the last 30 years. Nevertheless, knowledge about human sensory neurons is critical for a translational research approach and future therapeutic development. This review aims to summarize both historical and emerging information about the size and location of human DRG, and highlight advances in the understanding of the neurochemical characteristics of human DRG neurons, in particular nociceptive neurons.
Collapse
Affiliation(s)
- Rainer Viktor Haberberger
- Pain and Pulmonary Neurobiology Laboratory, Centre for Neuroscience, Anatomy and Histology, Flinders University, Adelaide, SA, Australia.,Órama Institute, Flinders University, Adelaide, SA, Australia
| | - Christine Barry
- Pain and Pulmonary Neurobiology Laboratory, Centre for Neuroscience, Anatomy and Histology, Flinders University, Adelaide, SA, Australia
| | - Nicholas Dominguez
- Pain and Pulmonary Neurobiology Laboratory, Centre for Neuroscience, Anatomy and Histology, Flinders University, Adelaide, SA, Australia
| | - Dusan Matusica
- Pain and Pulmonary Neurobiology Laboratory, Centre for Neuroscience, Anatomy and Histology, Flinders University, Adelaide, SA, Australia.,Órama Institute, Flinders University, Adelaide, SA, Australia
| |
Collapse
|
20
|
Zhang X, Hartung JE, Friedman RL, Koerber HR, Belfer I, Gold MS. Nicotine Evoked Currents in Human Primary Sensory Neurons. THE JOURNAL OF PAIN 2019; 20:810-818. [PMID: 30659887 DOI: 10.1016/j.jpain.2019.01.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 12/18/2018] [Accepted: 01/03/2019] [Indexed: 11/28/2022]
Abstract
Sensory neuron nicotinic acetylcholine receptors (nAChRs) contribute to pain associated with tissue injury. However, there are marked differences between rats and mice with respect to both the properties and distribution of nAChR currents in sensory neurons. Because both species are used to understand pain signaling in humans, we sought to determine whether the currents present in either species was reflective of those present in human sensory neurons. Neurons from the L4/L5 dorsal root ganglia were obtained from adult male and female organ donors. Nicotine evoked currents were detected in 40 of 47 neurons (85%). In contrast with the naïve mouse, in which almost all nAChR currents are transient, or the rat, in which both mouse-like transient and more slowly activating and inactivating currents are detected, all the currents in human DRG neurons were slow, but slower than those in the rat. Currents were blocked by the nAChR antagonists mecamylamine (30 µmol/L), but not by the TRPA1 selective antagonist HC-030031 (10 µmol/L). Single cell polymerase chain reaction analysis of nicotinic receptor subunit expression in human DRG neurons are consistent with functional data indicating that receptor expression is detected 85 ± 2.1% of neurons assessed (n = 48, from 4 donors). The most prevalent coexpression pattern was α3/β2 (95 ± 4% of neurons with subunits), but α7 subunits were detected in 70 ± 3.4% of neurons. These results suggest that there are not only species differences in the sensory neuron distribution of nAChR currents between rodent and human, but that the subunit composition of the channel underlying human nAChR currents may be different from those in the mouse or rat. PERSPECTIVE: The properties and distribution of nicotine evoked currents in human sensory neurons were markedly different from those previously observed in mice and rats. These observations add additional support to the suggestion that human sensory neurons may be an essential screening tool for those considering moving novel therapeutics targeting primary afferents into clinical trials.
Collapse
Affiliation(s)
| | - Jane E Hartung
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Robert L Friedman
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - H Richard Koerber
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | | | - Michael S Gold
- Department of Anesthesiology; Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.
| |
Collapse
|
21
|
Hockley JRF, Smith ESJ, Bulmer DC. Human visceral nociception: findings from translational studies in human tissue. Am J Physiol Gastrointest Liver Physiol 2018; 315:G464-G472. [PMID: 29848022 DOI: 10.1152/ajpgi.00398.2017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Peripheral sensitization of nociceptors during disease has long been recognized as a leading cause of inflammatory pain. However, a growing body of data generated over the last decade has led to the increased understanding that peripheral sensitization is also an important mechanism driving abdominal pain in highly prevalent functional bowel disorders, in particular, irritable bowel syndrome (IBS). As such, the development of drugs that target pain-sensing nerves innervating the bowel has the potential to be a successful analgesic strategy for the treatment of abdominal pain in both organic and functional gastrointestinal diseases. Despite the success of recent peripherally restricted approaches for the treatment of IBS, not all drugs that have shown efficacy in animal models of visceral pain have reduced pain end points in clinical trials of IBS patients, suggesting innate differences in the mechanisms of pain processing between rodents and humans and, in particular, how we model disease states. To address this gap in our understanding of peripheral nociception from the viscera and the body in general, several groups have developed experimental systems to study nociception in isolated human tissue and neurons, the findings of which we discuss in this review. Studies of human tissue identify a repertoire of human primary afferent subtypes comparable to rodent models including a nociceptor population, the targeting of which will shape future analgesic development efforts. Detailed mechanistic studies in human sensory neurons combined with unbiased RNA-sequencing approaches have revealed fundamental differences in not only receptor/channel expression but also peripheral pain pathways.
Collapse
Affiliation(s)
- James R F Hockley
- Department of Pharmacology, University of Cambridge , Cambridge , United Kingdom
| | - Ewan St John Smith
- Department of Pharmacology, University of Cambridge , Cambridge , United Kingdom
| | - David C Bulmer
- Department of Pharmacology, University of Cambridge , Cambridge , United Kingdom
| |
Collapse
|
22
|
Snyder LM, Chiang MC, Loeza-Alcocer E, Omori Y, Hachisuka J, Sheahan TD, Gale JR, Adelman PC, Sypek EI, Fulton SA, Friedman RL, Wright MC, Duque MG, Lee YS, Hu Z, Huang H, Cai X, Meerschaert KA, Nagarajan V, Hirai T, Scherrer G, Kaplan DH, Porreca F, Davis BM, Gold MS, Koerber HR, Ross SE. Kappa Opioid Receptor Distribution and Function in Primary Afferents. Neuron 2018; 99:1274-1288.e6. [PMID: 30236284 PMCID: PMC6300132 DOI: 10.1016/j.neuron.2018.08.044] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 07/06/2018] [Accepted: 08/21/2018] [Indexed: 02/02/2023]
Abstract
Primary afferents are known to be inhibited by kappa opioid receptor (KOR) signaling. However, the specific types of somatosensory neurons that express KOR remain unclear. Here, using a newly developed KOR-cre knockin allele, viral tracing, single-cell RT-PCR, and ex vivo recordings, we show that KOR is expressed in several populations of primary afferents: a subset of peptidergic sensory neurons, as well as low-threshold mechanoreceptors that form lanceolate or circumferential endings around hair follicles. We find that KOR acts centrally to inhibit excitatory neurotransmission from KOR-cre afferents in laminae I and III, and this effect is likely due to KOR-mediated inhibition of Ca2+ influx, which we observed in sensory neurons from both mouse and human. In the periphery, KOR signaling inhibits neurogenic inflammation and nociceptor sensitization by inflammatory mediators. Finally, peripherally restricted KOR agonists selectively reduce pain and itch behaviors, as well as mechanical hypersensitivity associated with a surgical incision. These experiments provide a rationale for the use of peripherally restricted KOR agonists for therapeutic treatment.
Collapse
Affiliation(s)
- Lindsey M Snyder
- Department of Neurobiology and the Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Michael C Chiang
- Department of Neurobiology and the Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Emanuel Loeza-Alcocer
- Department of Neurobiology and the Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Yu Omori
- Department of Neurobiology and the Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Junichi Hachisuka
- Department of Neurobiology and the Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Tayler D Sheahan
- Department of Neurobiology and the Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Jenna R Gale
- Department of Neurobiology and the Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Peter C Adelman
- Department of Neurobiology and the Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Elizabeth I Sypek
- Department of Anesthesiology, Perioperative, and Pain Medicine, Department of Molecular and Cellular Physiology, and Department of Neurosurgery, Stanford Neurosciences Institute, Stanford University, Stanford, CA 94305, USA
| | - Stephanie A Fulton
- Department of Neurobiology and the Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Robert L Friedman
- Department of Neurobiology and the Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Margaret C Wright
- Department of Neurobiology and the Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Melissa Giraldo Duque
- Department of Neurobiology and the Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Yeon Sun Lee
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ 85721, USA
| | - Zeyu Hu
- Department of Neurobiology and the Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Huizhen Huang
- Department of Neurobiology and the Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213, USA; Tsinghua University School of Medicine Beijing, Beijing 100084, China
| | - Xiaoyun Cai
- Department of Neurobiology and the Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Kimberly A Meerschaert
- Department of Neurobiology and the Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Vidhya Nagarajan
- Department of Neurobiology and the Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Toshiro Hirai
- Departments of Dermatology and Immunology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Gregory Scherrer
- Department of Anesthesiology, Perioperative, and Pain Medicine, Department of Molecular and Cellular Physiology, and Department of Neurosurgery, Stanford Neurosciences Institute, Stanford University, Stanford, CA 94305, USA; New York Stem Cell Foundation-Robertson Investigator, Stanford University, Palo Alto, CA 94304, USA
| | - Daniel H Kaplan
- Departments of Dermatology and Immunology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Frank Porreca
- Department of Pharmacology, University of Arizona, Tucson, AZ 85719, USA
| | - Brian M Davis
- Department of Neurobiology and the Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Michael S Gold
- Department of Neurobiology and the Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| | - H Richard Koerber
- Department of Neurobiology and the Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| | - Sarah E Ross
- Department of Neurobiology and the Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Anesthesiology, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| |
Collapse
|
23
|
Chakrabarti S, Pattison LA, Singhal K, Hockley JRF, Callejo G, Smith ESJ. Acute inflammation sensitizes knee-innervating sensory neurons and decreases mouse digging behavior in a TRPV1-dependent manner. Neuropharmacology 2018; 143:49-62. [PMID: 30240782 PMCID: PMC6277850 DOI: 10.1016/j.neuropharm.2018.09.014] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 09/06/2018] [Accepted: 09/11/2018] [Indexed: 01/20/2023]
Abstract
Ongoing, spontaneous pain is characteristic of inflammatory joint pain and reduces an individual's quality of life. To understand the neural basis of inflammatory joint pain, we made a unilateral knee injection of complete Freund's adjuvant (CFA) in mice, which reduced their natural digging behavior. We hypothesized that sensitization of knee-innervating dorsal root ganglion (DRG) neurons underlies this altered behavior. To test this hypothesis, we performed electrophysiological recordings on retrograde labeled knee-innervating primary DRG neuron cultures and measured their responses to a number of electrical and chemical stimuli. We found that 24-h after CFA-induced knee inflammation, knee neurons show a decreased action potential generation threshold, as well as increased GABA and capsaicin sensitivity, but have unaltered acid sensitivity. The inflammation-induced sensitization of knee neurons persisted for 24-h in culture, but was not observed after 48-h in culture. Through immunohistochemistry, we showed that the increased knee neuron capsaicin sensitivity correlated with enhanced expression of the capsaicin receptor, transient receptor potential vanilloid 1 (TRPV1) in knee-innervating neurons of the CFA-injected side. We also observed an increase in the co-expression of TRPV1 with tropomyosin receptor kinase A (TrkA), which is the receptor for nerve growth factor (NGF), suggesting that NGF partially induces the increased TRPV1 expression. Lastly, we found that systemic administration of the TRPV1 antagonist, A-425619, reversed the decrease in digging behavior induced by CFA injection, further confirming the role of TRPV1, expressed by knee neurons, in acute inflammatory joint pain. Knee inflammation decreases digging behavior in mice. Knee-innervating dorsal root ganglion neurons are hyperexcitable after inflammation. NGF-mediated increase in TRPV1 expression is observed in knee-innervating neurons. Systemic TRPV1 antagonist administration normalises digging behavior in mice.
Collapse
Affiliation(s)
| | - Luke A Pattison
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Kaajal Singhal
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | | | - Gerard Callejo
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | | |
Collapse
|
24
|
Peripheral GABA A receptor-mediated signaling facilitates persistent inflammatory hypersensitivity. Neuropharmacology 2018; 135:572-580. [PMID: 29634983 DOI: 10.1016/j.neuropharm.2018.04.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 03/30/2018] [Accepted: 04/06/2018] [Indexed: 01/26/2023]
Abstract
Unlike in the central nervous system (CNS), in the adult peripheral nervous system (PNS), activation of GABAA receptors (GABAAR) is excitatory because of the relatively high concentration of intracellular chloride in these neurons. Indeed, exogenous GABA and muscimol, a GABAAR agonist, exacerbate acute inflammatory hypersensitivity in rodents. However, it remains unclear whether peripheral GABAAR and the endogenous GABA play an important role in persistent inflammatory hypersensitivity. In this study, we thus investigated how peripheral GABAAR affects pain hypersensitivity by using the complete Freund's adjuvant (CFA)-induced persistent inflammatory pain mouse model. We found that intraplantar (i.pl.) administration of GABAAR antagonists, picrotoxin, and 1(S),9(R)-(-)-bicuculline methiodide significantly inhibited both spontaneous nociceptive (paw licking and flinching) behavior and mechanical hypersensitivity in CFA-injected mice at day 3 (D3), but not in naïve mice. Interestingly, CFA-induced mechanical hypersensitivity was significantly reversed by anti-GABA antibody (anti-GABA, i.pl.). In addition, RT-qPCR revealed that glutamate decarboxylase Gad1 (GAD 67) and Gad2 (GAD 65) mRNA expression was also upregulated in the ipsilateral hind paw of CFA-injected mice at D3. Finally, 5α-pregnan-3α-ol-20-one (3α,5α-THP), a selective positive allosteric modulator of GABAAR, produced mechanical hypersensitivity in naïve mice in a dose-dependent manner. Taken together, our results indicate that peripheral GABAAR and endogenous GABA, possibly produced by the inflamed tissue, potentiate CFA-induced persistent inflammatory hypersensitivity, suggesting that they can be used as a therapeutic target for alleviating inflammatory pain.
Collapse
|
25
|
Sheahan TD, Valtcheva MV, McIlvried LA, Pullen MY, Baranger DA, Gereau RW. Metabotropic Glutamate Receptor 2/3 (mGluR2/3) Activation Suppresses TRPV1 Sensitization in Mouse, But Not Human, Sensory Neurons. eNeuro 2018; 5:ENEURO.0412-17.2018. [PMID: 29662945 PMCID: PMC5898698 DOI: 10.1523/eneuro.0412-17.2018] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 02/06/2018] [Accepted: 02/07/2018] [Indexed: 01/08/2023] Open
Abstract
The use of human tissue to validate putative analgesic targets identified in rodents is a promising strategy for improving the historically poor translational record of preclinical pain research. We recently demonstrated that in mouse and human sensory neurons, agonists for metabotropic glutamate receptors 2 and 3 (mGluR2/3) reduce membrane hyperexcitability produced by the inflammatory mediator prostaglandin E2 (PGE2). Previous rodent studies indicate that mGluR2/3 can also reduce peripheral sensitization by suppressing inflammation-induced sensitization of TRPV1. Whether this observation similarly translates to human sensory neurons has not yet been tested. We found that activation of mGluR2/3 with the agonist APDC suppressed PGE2-induced sensitization of TRPV1 in mouse, but not human, sensory neurons. We also evaluated sensory neuron expression of the gene transcripts for mGluR2 (Grm2), mGluR3 (Grm3), and TRPV1 (Trpv1). The majority of Trpv1+ mouse and human sensory neurons expressed Grm2 and/or Grm3, and in both mice and humans, Grm2 was expressed in a greater percentage of sensory neurons than Grm3. Although we demonstrated a functional difference in the modulation of TRPV1 sensitization by mGluR2/3 activation between mouse and human, there were no species differences in the gene transcript colocalization of mGluR2 or mGluR3 with TRPV1 that might explain this functional difference. Taken together with our previous work, these results suggest that mGluR2/3 activation suppresses only some aspects of human sensory neuron sensitization caused by PGE2. These differences have implications for potential healthy human voluntary studies or clinical trials evaluating the analgesic efficacy of mGluR2/3 agonists or positive allosteric modulators.
Collapse
Affiliation(s)
- Tayler D. Sheahan
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri 63110
- Washington University Program in Neuroscience, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Manouela V. Valtcheva
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri 63110
- Washington University Program in Neuroscience, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Lisa A. McIlvried
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Melanie Y. Pullen
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri 63110
| | - David A.A. Baranger
- Washington University Program in Neuroscience, Washington University School of Medicine, St. Louis, Missouri 63110
- BRAIN Laboratory, Department of Psychological and Brain Sciences, Washington University in St. Louis, St. Louis, Missouri 63130
| | - Robert W. Gereau
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri 63110
| |
Collapse
|
26
|
Zhang X, Priest BT, Belfer I, Gold MS. Voltage-gated Na + currents in human dorsal root ganglion neurons. eLife 2017; 6. [PMID: 28508747 PMCID: PMC5433841 DOI: 10.7554/elife.23235] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2016] [Accepted: 04/11/2017] [Indexed: 12/15/2022] Open
Abstract
Available evidence indicates voltage-gated Na+ channels (VGSCs) in peripheral sensory neurons are essential for the pain and hypersensitivity associated with tissue injury. However, our understanding of the biophysical and pharmacological properties of the channels in sensory neurons is largely based on the study of heterologous systems or rodent tissue, despite evidence that both expression systems and species differences influence these properties. Therefore, we sought to determine the extent to which the biophysical and pharmacological properties of VGSCs were comparable in rat and human sensory neurons. Whole cell patch clamp techniques were used to study Na+ currents in acutely dissociated neurons from human and rat. Our results indicate that while the two major current types, generally referred to as tetrodotoxin (TTX)-sensitive and TTX-resistant were qualitatively similar in neurons from rats and humans, there were several differences that have important implications for drug development as well as our understanding of pain mechanisms. DOI:http://dx.doi.org/10.7554/eLife.23235.001
Collapse
Affiliation(s)
- Xiulin Zhang
- Department of Urology, The Second Hospital of Shandong University, Jinan Shi, China
| | | | - Inna Belfer
- Office of Research on Women's Health, National Institutes of Health, Bethesda, United States
| | - Michael S Gold
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, United States
| |
Collapse
|
27
|
Du X, Hao H, Yang Y, Huang S, Wang C, Gigout S, Ramli R, Li X, Jaworska E, Edwards I, Deuchars J, Yanagawa Y, Qi J, Guan B, Jaffe DB, Zhang H, Gamper N. Local GABAergic signaling within sensory ganglia controls peripheral nociceptive transmission. J Clin Invest 2017; 127:1741-1756. [PMID: 28375159 PMCID: PMC5409786 DOI: 10.1172/jci86812] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 02/02/2017] [Indexed: 01/05/2023] Open
Abstract
The integration of somatosensory information is generally assumed to be a function of the central nervous system (CNS). Here we describe fully functional GABAergic communication within rodent peripheral sensory ganglia and show that it can modulate transmission of pain-related signals from the peripheral sensory nerves to the CNS. We found that sensory neurons express major proteins necessary for GABA synthesis and release and that sensory neurons released GABA in response to depolarization. In vivo focal infusion of GABA or GABA reuptake inhibitor to sensory ganglia dramatically reduced acute peripherally induced nociception and alleviated neuropathic and inflammatory pain. In addition, focal application of GABA receptor antagonists to sensory ganglia triggered or exacerbated peripherally induced nociception. We also demonstrated that chemogenetic or optogenetic depolarization of GABAergic dorsal root ganglion neurons in vivo reduced acute and chronic peripherally induced nociception. Mechanistically, GABA depolarized the majority of sensory neuron somata, yet produced a net inhibitory effect on the nociceptive transmission due to the filtering effect at nociceptive fiber T-junctions. Our findings indicate that peripheral somatosensory ganglia represent a hitherto underappreciated site of somatosensory signal integration and offer a potential target for therapeutic intervention.
Collapse
Affiliation(s)
- Xiaona Du
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, China; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province; Shijiazhuang, China
| | - Han Hao
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, China; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province; Shijiazhuang, China
| | - Yuehui Yang
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, China; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province; Shijiazhuang, China
| | - Sha Huang
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, China; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province; Shijiazhuang, China
| | - Caixue Wang
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, China; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province; Shijiazhuang, China
| | - Sylvain Gigout
- Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Rosmaliza Ramli
- Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
- School of Dental Sciences, Universiti Sains Malaysia, Kelantan, Malaysia
| | - Xinmeng Li
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, China; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province; Shijiazhuang, China
| | - Ewa Jaworska
- Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Ian Edwards
- Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Jim Deuchars
- Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Yuchio Yanagawa
- Department of Genetic and Behavioral Neuroscience, Gunma University Graduate School of Medicine and Japan Science and Technology Agency, CREST, Maebashi, Japan
| | - Jinlong Qi
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, China; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province; Shijiazhuang, China
| | - Bingcai Guan
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, China; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province; Shijiazhuang, China
| | - David B. Jaffe
- Department of Biology, UTSA Neurosciences Institute, University of Texas at San Antonio, San Antonio, Texas, USA
| | - Hailin Zhang
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, China; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province; Shijiazhuang, China
| | - Nikita Gamper
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, China; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province; Shijiazhuang, China
- Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
28
|
Valtcheva MV, Copits BA, Davidson S, Sheahan TD, Pullen MY, McCall JG, Dikranian K, Gereau RW. Surgical extraction of human dorsal root ganglia from organ donors and preparation of primary sensory neuron cultures. Nat Protoc 2016; 11:1877-88. [PMID: 27606776 PMCID: PMC5082842 DOI: 10.1038/nprot.2016.111] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Primary cultures of rodent sensory neurons are widely used to investigate the cellular and molecular mechanisms involved in pain, itch, nerve injury and regeneration. However, translation of these preclinical findings may be greatly improved by direct validation in human tissues. We have developed an approach to extract and culture human sensory neurons in collaboration with a local organ procurement organization (OPO). Here we describe the surgical procedure for extraction of human dorsal root ganglia (hDRG) and the necessary modifications to existing culture techniques to prepare viable adult human sensory neurons for functional studies. Dissociated sensory neurons can be maintained in culture for >10 d, and they are amenable to electrophysiological recording, calcium imaging and viral gene transfer. The entire process of extraction and culturing can be completed in <7 h, and it can be performed by trained graduate students. This approach can be applied at any institution with access to organ donors consenting to tissue donation for research, and is an invaluable resource for improving translational research.
Collapse
Affiliation(s)
- Manouela V. Valtcheva
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, 660 South Euclid Ave, Campus Box 8054, St. Louis, MO 63110
- Medical Scientist Training Program, Washington University in St. Louis, 660 South Euclid Ave, St. Louis, MO 63110
- Neurosciences Program, Washington University in St. Louis, 660 South Euclid Ave, St. Louis, MO 63110
| | - Bryan A. Copits
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, 660 South Euclid Ave, Campus Box 8054, St. Louis, MO 63110
| | - Steve Davidson
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, 660 South Euclid Ave, Campus Box 8054, St. Louis, MO 63110
- Pain Research Center, Department of Anesthesiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267
| | - Tayler D. Sheahan
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, 660 South Euclid Ave, Campus Box 8054, St. Louis, MO 63110
- Neurosciences Program, Washington University in St. Louis, 660 South Euclid Ave, St. Louis, MO 63110
| | - Melanie Y. Pullen
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, 660 South Euclid Ave, Campus Box 8054, St. Louis, MO 63110
- Developmental, Regenerative, and Stem Cell Biology Program, Washington University in St. Louis, 660 South Euclid Ave, St. Louis, MO 63110
| | - Jordan G. McCall
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, 660 South Euclid Ave, Campus Box 8054, St. Louis, MO 63110
| | - Krikor Dikranian
- Department of Anatomy and Neurobiology, Washington University School of Medicine, 660 South Euclid Ave, St. Louis, Missouri, 63110
| | - Robert W. Gereau
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, 660 South Euclid Ave, Campus Box 8054, St. Louis, MO 63110
| |
Collapse
|