1
|
Abd El Hay MY, Kamm GB, Tlaie Boria A, Siemens J. Diverging roles of TRPV1 and TRPM2 in warm-temperature detection. eLife 2025; 13:RP95618. [PMID: 40215103 PMCID: PMC11991700 DOI: 10.7554/elife.95618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2025] Open
Abstract
The perception of innocuous temperatures is crucial for thermoregulation. The TRP ion channels TRPV1 and TRPM2 have been implicated in warmth detection, yet their precise roles remain unclear. A key challenge is the low prevalence of warmth-sensitive sensory neurons, comprising fewer than 10% of rodent dorsal root ganglion (DRG) neurons. Using calcium imaging of >20,000 cultured mouse DRG neurons, we uncovered distinct contributions of TRPV1 and TRPM2 to warmth sensitivity. TRPV1's absence - and to a lesser extent absence of TRPM2 - reduces the number of neurons responding to warmth. Additionally, TRPV1 mediates the rapid, dynamic response to a warmth challenge. Behavioural tracking in a whole-body thermal preference assay revealed that these cellular differences shape nuanced thermal behaviours. Drift diffusion modelling of decision-making in mice exposed to varying temperatures showed that TRPV1 deletion impairs evidence accumulation, reducing the precision of thermal choice, while TRPM2 deletion increases overall preference for warmer environments that wildtype mice avoid. It remains unclear whether TRPM2 in DRG sensory neurons or elsewhere mediates thermal preference. Our findings suggest that different aspects of thermal information, such as stimulation speed and temperature magnitude, are encoded by distinct TRP channel mechanisms.
Collapse
Affiliation(s)
- Muad Y Abd El Hay
- Department of Pharmacology, Heidelberg UniversityHeidelbergGermany
- Ernst Strüngmann Institute for Neuroscience in Cooperation with the Max Planck SocietyFrankfurt am MainGermany
| | - Gretel B Kamm
- Department of Pharmacology, Heidelberg UniversityHeidelbergGermany
| | - Alejandro Tlaie Boria
- Ernst Strüngmann Institute for Neuroscience in Cooperation with the Max Planck SocietyFrankfurt am MainGermany
| | - Jan Siemens
- Department of Pharmacology, Heidelberg UniversityHeidelbergGermany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory (EMBL)HeidelbergGermany
| |
Collapse
|
2
|
Yi J, Yang L, Widman AJ, Toliver A, Bertels Z, Del Rosario JS, Slivicki RA, Payne M, Dourson AJ, Li JN, Kumar R, Gupta P, Mwirigi JM, Chamessian A, Lemen J, Copits BA, Gereau RW. Human sensory neurons exhibit cell-type-specific, pain-associated differences in intrinsic excitability and expression of SCN9A and SCN10A. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.25.645367. [PMID: 40196681 PMCID: PMC11974934 DOI: 10.1101/2025.03.25.645367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Despite the prevalence of chronic pain, the approval of novel, non-opioid therapeutics has been slow. A major translational challenge in analgesic development is the difference in gene expression and functional properties between human and rodent dorsal root ganglia (DRG) sensory neurons. Extensive work in rodents suggests that sensitization of nociceptors in the DRG is essential for the pathogenesis and persistence of pain; however, direct evidence demonstrating similar physiological sensitization in humans is limited. Here, we examine whether pain history is associated with nociceptor hyperexcitability in human DRG (hDRG). We identified three electrophysiologically distinct clusters (E-types) of hDRG neurons based on firing properties and membrane excitability. Combining electrophysiological recordings and single-cell RNA-sequencing ("Patch-seq"), we linked these E-types to specific transcriptionally defined nociceptor subpopulations. Comparing hDRG neurons from donors with and without evident pain history revealed cluster-specific, pain history-associated differences in hDRG excitability. Finally, we found that hDRG from donors with pain history express higher levels of transcripts encoding voltage-gated sodium channel 1.7 (NaV1.7) and 1.8 (NaV1.8) which specifically regulate nociceptor excitability. These findings suggest that donors with pain history exhibit distinct hDRG electrophysiological profiles compared to those without pain history and further validate NaV1.7 and 1.8 as targets for analgesic development.
Collapse
Affiliation(s)
- Jiwon Yi
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, United States
- Neuroscience Graduate Program, Division of Biology & Biomedical Sciences, Washington University School of Medicine, St. Louis, MO, United States
| | - Lite Yang
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, United States
- Neuroscience Graduate Program, Division of Biology & Biomedical Sciences, Washington University School of Medicine, St. Louis, MO, United States
| | - Allie J. Widman
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, United States
| | - Alexa Toliver
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, United States
| | - Zachariah Bertels
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, United States
| | - John Smith Del Rosario
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, United States
| | - Richard A. Slivicki
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, United States
| | - Maria Payne
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, United States
| | - Adam J. Dourson
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, United States
| | - Jun-Nan Li
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, United States
| | - Rakesh Kumar
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, United States
| | - Prashant Gupta
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, United States
| | - Juliet M. Mwirigi
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, United States
| | - Alexander Chamessian
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, United States
| | - John Lemen
- Mid-America Transplant, St. Louis, MO, United States
| | - Bryan A. Copits
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, United States
| | - Robert W. Gereau
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, United States
- Department of Neuroscience, Washington University, St. Louis, MO, United States
| |
Collapse
|
3
|
Barry AM, Sondermann JR, Lesnak JB, Xian F, Franco-Enzástiga Ú, O'Brien JA, Gomez-Varela D, Schackmuth MK, Shiers S, Price TJ, Schmidt M. Multi-omic integration with human DRG proteomics highlights TNFα signalling as a relevant sexually dimorphic pathway. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.06.626968. [PMID: 39713351 PMCID: PMC11661068 DOI: 10.1101/2024.12.06.626968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
The peripheral nervous system (PNS) plays a critical role in pathological conditions, including chronic pain disorders, that manifest differently in men and women. To investigate this sexual dimorphism at the molecular level, we integrated quantitative proteomic profiling of human dorsal root ganglia (hDRG) and peripheral nerve tissue into the expanding omics framework of the PNS. Using data-independent acquisition (DIA) mass spectrometry, we characterized a comprehensive proteomic profile, validating tissue-specific differences between the hDRG and peripheral nerve. Through multi-omic analyses and in vitro functional assays, we identified sex-specific molecular differences, with TNFα signalling emerging as a key sexually dimorphic pathway with higher prominence in males. Genetic evidence from genome-wide association studies (GWAS) further supports the functional relevance of TNFα signalling in the periphery, while clinical trial data and meta-analyses indicate a sex-dependent response to TNFα inhibitors. Collectively, these findings underscore a functionally sexual dimorphism in the PNS, with direct implications for sensory and pain-related clinical translation.
Collapse
Affiliation(s)
- Allison M Barry
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX 75080, USA
- Systems Biology of Pain, Division of Pharmacology & Toxicology, Department of Pharmaceutical Sciences, University of Vienna, Austria
| | - Julia R Sondermann
- Systems Biology of Pain, Division of Pharmacology & Toxicology, Department of Pharmaceutical Sciences, University of Vienna, Austria
| | - Joseph B Lesnak
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Feng Xian
- Systems Biology of Pain, Division of Pharmacology & Toxicology, Department of Pharmaceutical Sciences, University of Vienna, Austria
| | - Úrzula Franco-Enzástiga
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Jayden A O'Brien
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX 75080, USA
| | - David Gomez-Varela
- Systems Biology of Pain, Division of Pharmacology & Toxicology, Department of Pharmaceutical Sciences, University of Vienna, Austria
| | - Morgan K Schackmuth
- 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
| | - Theodore J Price
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Manuela Schmidt
- Systems Biology of Pain, Division of Pharmacology & Toxicology, Department of Pharmaceutical Sciences, University of Vienna, Austria
| |
Collapse
|
4
|
Chamessian A, Tavares-Ferriera D, Payne M, Govindarajan R, Pestronk A, Bertels Z, Widman AJ, Slivicki RA, Del Rosario JS, Yi J, Copits B, Ornitz DM, Price TJ, Gereau RW. Expression of Fibroblast Growth Factor Receptor 3 (FGFR3) in the Human Peripheral Nervous System: Implications for the Putative Pathogenic Role of FGFR3 Autoantibodies in Neuropathy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.03.639509. [PMID: 40093173 PMCID: PMC11908139 DOI: 10.1101/2025.03.03.639509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Introduction Autoantibodies to the Fibroblast Growth Factor Receptor 3 (FGFR3-AAbs) have been associated with idiopathic sensory-predominant neuropathy. The pathogenicity of FGFR3-AAbs in this disorder is unknown. Pathogenic mechanisms of autoantibodies in other dysimmune neuropathies commonly involve their direct binding to antigens on either neurons or glia. The expression of FGFR3 in the human peripheral nervous system is unknown. Therefore, as an initial step toward clarifying the pathogenicity of FGFR3-AAbs, we characterized the expression of FGFR3 in nerve (hNerve), dorsal root ganglia (hDRG) and spinal cord (hSC) in human. Methods FGFR3 mRNA was assayed via in situ hybridization (ISH) on post-mortem sections of hNerve, hDRG and hSC, and by re-analysis of RNA-sequencing data from hDRG. FGFR3 protein was assayed in these tissues using capillary electrophoretic immunoassays (CEIA) with several validated anti-FGFR3 antibodies. Results FGFR3 mRNA was not detected in hNerve or hDRG but was abundant in hSC by ISH. FGFR3 protein was absent from hNerve and hDRG by CEIA but was moderately expressed in hSC. Discussion A direct pathogenic mechanism of FGFR3-AAbs in sensory neuropathy would require the expression of FGFR3 in either neurons or non-neuronal cells in nerve or DRG. Using multiple methods, we did not detect FGFR3 expression at the mRNA or protein levels in these tissues. Given the absence of FGFR3 from hNerve and hDRG, it is improbable that FGFR3-AAbs cause direct damage to the neural components involved in neuropathy and thus are unlikely to be pathogenic, although indirect mechanisms via non-neural cells cannot be excluded.
Collapse
Affiliation(s)
- Alexander Chamessian
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Diana Tavares-Ferriera
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson TX 75080, US
| | - Maria Payne
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Raghav Govindarajan
- Hospital Sisters Health System, Division of Neurology, Edwardsville, IL 62025
| | - Alan Pestronk
- Washington University Department of Neurology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Zach Bertels
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Allie J Widman
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Richard A Slivicki
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - John S Del Rosario
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Jiwon Yi
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Bryan Copits
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - David M Ornitz
- Department of Developmental Biology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Theodore J Price
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson TX 75080, US
| | - Robert W Gereau
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| |
Collapse
|
5
|
Yang J, Xie YF, Smith R, Ratté S, Prescott SA. Discordance between preclinical and clinical testing of Na V 1.7-selective inhibitors for pain. Pain 2025; 166:481-501. [PMID: 39928833 PMCID: PMC11808711 DOI: 10.1097/j.pain.0000000000003425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/19/2024] [Accepted: 08/13/2024] [Indexed: 10/26/2024]
Abstract
ABSTRACT The voltage-gated sodium channel Na V 1.7 plays an important role in pain processing according to genetic data. Those data made Na V 1.7 a popular drug target, especially since its relatively selective expression in nociceptors promised pain relief without the adverse effects associated with broader sodium channel blockade. Despite encouraging preclinical data in rodents, Na V 1.7-selective inhibitors have not yet proven effective in clinical trials. Discrepancies between preclinical and clinical results should raise alarms. We reviewed preclinical and clinical reports on the analgesic efficacy of Na V 1.7-selective inhibitors and found critical differences in several factors. Putting aside species differences, most preclinical studies tested young male rodents with limited genetic variability, inconsistent with the clinical population. Inflammatory pain was the most common preclinical chronic pain model whereas nearly all clinical trials focused on neuropathic pain despite some evidence suggesting Na V 1.7 channels are not essential for neuropathic pain. Preclinical studies almost exclusively measured evoked pain whereas most clinical trials assessed average pain intensity without distinguishing between evoked and spontaneous pain. Nearly all preclinical studies gave a single dose of drug unlike the repeat dosing used clinically, thus precluding preclinical data from demonstrating whether tolerance or other slow processes occur. In summary, preclinical testing of Na V 1.7-selective inhibitors aligned poorly with clinical testing. Beyond issues that have already garnered widespread attention in the pain literature, our results highlight the treatment regimen and choice of pain model as areas for improvement.
Collapse
Affiliation(s)
- Jane Yang
- Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Yu-Feng Xie
- Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, Canada
| | - Russell Smith
- Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, Canada
| | - Stéphanie Ratté
- Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, Canada
| | - Steven A. Prescott
- Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
6
|
Gupta RA, Higham JP, Pearce A, Urriola-Muñoz P, Barker KH, Paine L, Ghooraroo J, Raine T, Hockley JRF, Rahman T, St John Smith E, Brown AJH, Ladds G, Suzuki R, Bulmer DC. GPR35 agonists inhibit TRPA1-mediated colonic nociception through suppression of substance P release. Pain 2025; 166:596-613. [PMID: 39382322 PMCID: PMC11808708 DOI: 10.1097/j.pain.0000000000003399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 07/12/2024] [Accepted: 07/25/2024] [Indexed: 10/10/2024]
Abstract
ABSTRACT The development of nonopioid analgesics for the treatment of abdominal pain is a pressing clinical problem. To address this, we examined the expression of G i/o -coupled receptors, which typically inhibit nociceptor activation, in colonic sensory neurons. This led to the identification of the orphan receptor GPR35 as a visceral analgesic drug target because of its marked coexpression with transient receptor potential ankyrin 1 (TRPA1), a mediator of noxious mechanotransduction in the bowel. Building on in silico docking simulations, we confirmed that the mast cell stabiliser, cromolyn (CS), and phosphodiesterase inhibitor, zaprinast, are agonists at mouse GPR35, promoting the activation of different G i/o subunits. Pretreatment with either CS or zaprinast significantly attenuated TRPA1-mediated colonic nociceptor activation and prevented TRPA1-mediated mechanosensitisation. These effects were lost in tissue from GPR35 -/- mice and were shown to be mediated by inhibition of TRPA1-evoked substance P (SP) release. This observation highlights the pronociceptive effect of SP and its contribution to TRPA1-mediated colonic nociceptor activation and sensitisation. Consistent with this mechanism of action, we confirmed that TRPA1-mediated colonic contractions evoked by SP release were abolished by CS pretreatment in a GPR35-dependent manner. Our data demonstrate that GPR35 agonists prevent the activation and sensitisation of colonic nociceptors through the inhibition of TRPA1-mediated SP release. These findings highlight the potential of GPR35 agonists to deliver nonopioid analgesia for the treatment of abdominal pain.
Collapse
Affiliation(s)
- Rohit A. Gupta
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, United Kingdom
| | - James P. Higham
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, United Kingdom
| | - Abigail Pearce
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, United Kingdom
| | - Paulina Urriola-Muñoz
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, United Kingdom
| | - Katie H. Barker
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, United Kingdom
| | - Luke Paine
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, United Kingdom
| | - Joshua Ghooraroo
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, United Kingdom
| | - Tim Raine
- Department of Gastroenterology, Addenbrookes Hospital, Cambridge University Teaching Hospitals, Cambridge, United Kingdom
| | - James R. F. Hockley
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, United Kingdom
| | - Taufiq Rahman
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, United Kingdom
| | - Ewan St John Smith
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, United Kingdom
| | - Alastair J. H. Brown
- Nxera, Steinmetz Building, Granta Park Great Abington, Cambridge, United Kingdom
| | - Graham Ladds
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, United Kingdom
| | - Rie Suzuki
- Nxera, Steinmetz Building, Granta Park Great Abington, Cambridge, United Kingdom
| | - David C. Bulmer
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, United Kingdom
| |
Collapse
|
7
|
Sankaranarayanan I, Kume M, Mohammed A, Mwirigi JM, Inturi NN, Munro G, Petersen KA, Tavares-Ferreira D, Price TJ. Persistent changes in the dorsal root ganglion nociceptor translatome governs hyperalgesic priming in mice: roles of GPR88 and Meteorin. Pain 2025:00006396-990000000-00811. [PMID: 39878635 DOI: 10.1097/j.pain.0000000000003523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 10/08/2024] [Indexed: 01/31/2025]
Abstract
ABSTRACT Hyperalgesic priming is a model system that has been widely used to understand plasticity in painful stimulus-detecting sensory neurons, called nociceptors. A key feature of this model system is that following priming, stimuli that do not normally cause hyperalgesia now readily provoke this state. We hypothesized that hyperalgesic priming occurs because of reorganization of translation of mRNA in nociceptors. To test this hypothesis, we used paclitaxel treatment as the priming stimulus and translating ribosome affinity purification to measure persistent changes in mRNA translation in Nav1.8+ nociceptors. Translating ribosome affinity purification sequencing revealed 161 genes with persistently altered mRNA translation in the primed state. Among these genes, we identified Gpr88 as upregulated and Metrn as downregulated. To provide functional evidence for these changes in hyperalgesic priming in a related priming model, we used the interleukin-6 priming model. A GPR88 agonist injection into the paw had no effect in naive mice but caused mechanical hypersensitivity and grimacing responses in female primed mice. Systemic Meteorin treatment in primed mice completely reversed established hyperalgesic priming mechanical hypersensitivity and grimacing responses to prostaglandin E2 in female mice. Our work demonstrates that altered nociceptor translatomes are causative in producing hyperalgesic priming in multiple models in female mice.
Collapse
Affiliation(s)
- Ishwarya Sankaranarayanan
- Department of Neuroscience, Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX
| | - Moeno Kume
- Department of Neuroscience, Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX
| | - Ayaan Mohammed
- Department of Neuroscience, Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX
| | - Juliet M Mwirigi
- Department of Neuroscience, Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX
| | - Nikhil Nageswar Inturi
- Department of Neuroscience, Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX
| | | | | | - Diana Tavares-Ferreira
- Department of Neuroscience, Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX
| | - Theodore J Price
- Department of Neuroscience, Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX
| |
Collapse
|
8
|
Inyang KE, Sim J, Clark KB, Geron M, Monahan K, Evans C, O'Connell P, Laumet S, Peng B, Ma J, Heijnen CJ, Dantzer R, Scherrer G, Kavelaars A, Bernard M, Aldhamen YA, Folger JK, Bavencoffe A, Laumet G. Upregulation of delta opioid receptor by meningeal interleukin-10 prevents relapsing pain. Brain Behav Immun 2025; 123:399-410. [PMID: 39349285 PMCID: PMC11624093 DOI: 10.1016/j.bbi.2024.09.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 09/23/2024] [Accepted: 09/27/2024] [Indexed: 10/02/2024] Open
Abstract
Chronic pain often includes periods of transient amelioration and even remission that alternate with severe relapsing pain. While most research on chronic pain has focused on pain development and maintenance, there is a critical unmet need to better understand the mechanisms that underlie pain remission and relapse. We found that interleukin (IL)-10, a pain resolving cytokine, is produced by resident macrophages in the spinal meninges during remission from pain and signaled to IL-10 receptor-expressing sensory neurons. Using unbiased RNA-sequencing, we identified that IL-10 upregulated expression and antinociceptive activity of δ-opioid receptor (δOR) in the dorsal root ganglion. Genetic or pharmacological inhibition of either IL-10 signaling or δOR triggered relapsing pain. Overall, our findings, from electrophysiology, genetic manipulation, flow cytometry, pharmacology, and behavioral approaches, indicate that remission of pain is not simply a return to the naïve state. Instead, remission is an adapted homeostatic state associated with lasting pain vulnerability resulting from persisting neuroimmune interactions within the nociceptive system. Broadly, this sheds light on the elusive mechanisms underlying recurrence a common aspect across various chronic pain conditions.
Collapse
Affiliation(s)
| | - Jaewon Sim
- Department of Physiology, Michigan State University, East Lansing, MI, USA; Cell and Molecular Biology Graduate Program, Michigan State University, East Lansing, MI, USA
| | - Kimberly B Clark
- Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Matan Geron
- Department of Cell Biology and Physiology, Department of Pharmacology, UNC Neuroscience Center, University of North Carolina, Chapel Hill, NC, USA
| | - Karli Monahan
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Christine Evans
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Patrick O'Connell
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, USA
| | - Sophie Laumet
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Bo Peng
- Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jiacheng Ma
- Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Cobi J Heijnen
- Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Robert Dantzer
- Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Grégory Scherrer
- Department of Cell Biology and Physiology, Department of Pharmacology, UNC Neuroscience Center, University of North Carolina, Chapel Hill, NC, USA; New York Stem Cell Foundation - Robertson Investigator, University of North Carolina, Chapel Hill, NC, USA
| | - Annemieke Kavelaars
- Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Matthew Bernard
- Flow Cytometry Core, Michigan State University, East Lansing, MI, USA
| | - Yasser A Aldhamen
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, USA
| | - Joseph K Folger
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Alexis Bavencoffe
- Department of Integrative Biology and Pharmacology, McGovern Medical School at UTHealth, Houston, TX, USA
| | - Geoffroy Laumet
- Department of Physiology, Michigan State University, East Lansing, MI, USA; Cell and Molecular Biology Graduate Program, Michigan State University, East Lansing, MI, USA; Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
9
|
Leisengang S. Pain research in a petri dish? Advantages and limitations of neuro-glial primary cell cultures from structures of the nociceptive system. Brain Behav Immun Health 2024; 41:100854. [PMID: 39308957 PMCID: PMC11415590 DOI: 10.1016/j.bbih.2024.100854] [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: 02/16/2024] [Revised: 06/26/2024] [Accepted: 09/03/2024] [Indexed: 09/25/2024] Open
Abstract
How can we learn more about pain without causing pain in humans or animals? This short review focuses on neuro-glial primary cell cultures as models to study neuro-immune interactions in the context of pain and discusses their advantages and limitations. The field of basic pain research places scientists in an ethical dilemma. We aim to understand underlying mechanisms of pain for an improved pain therapy for humans and animals. At the same time, this regularly includes the induction of pain in model animals. Within the field of psychoneuroimmunology, the examination of the complexity of neuro-immune interactions in health and disease as well as the bi-directional communication between the brain and the periphery make animal experiments an inevitable part of pain research. To address ethical and legal considerations as well as the growing societal awareness for animal welfare, scientists push for the identification and characterization of complementary methods to implement the 3R principle of Russel and Burch. As such, methods to replace animal studies, reduce the number of animals used, and refine experiments are tested. Neuro-glial primary cell cultures of structures of the nociceptive system, such as dorsal root ganglia (DRG) or the spinal dorsal horn (SDH) represent useful in vitro tools, when research comes to a cellular and molecular level. They allow for studying mechanisms of neuronal sensitization, glial cell activation, or the role of specific inflammatory mediators and intracellular signaling cascades involved in the development of inflammatory and neuropathic pain. Moreover, DRG/SDH-cultures provide the opportunity to test novel strategies for interventions, such as pharmaceuticals or cell-based therapies targeting neuroinflammatory processes. Thereby, in vitro models contribute to a better understanding of neuron-glia-immune communication in the context of pain and in the advancement of pain therapies. However, this can only be one piece in a large puzzle. Our knowledge about the complexity of pain will depend on studies in humans and animals applied in vitro and in vivo and will benefit from clear and open-minded interdisciplinary communication and transparency in public outreach.
Collapse
Affiliation(s)
- Stephan Leisengang
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University Giessen, Frankfurter Strasse 100, 35392 Giessen, Germany
- Translational Neuroscience Network Giessen (TNNG), Justus Liebig University Giessen, Germany
- Center for Mind, Brain and Behavior (CMBB), Philipps University Marburg & Justus Liebig University Giessen, Germany
| |
Collapse
|
10
|
Arendt-Tranholm A, Mwirigi JM, Price TJ. RNA isoform expression landscape of the human dorsal root ganglion generated from long-read sequencing. Pain 2024; 165:2468-2481. [PMID: 38809314 PMCID: PMC11511651 DOI: 10.1097/j.pain.0000000000003255] [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: 11/07/2023] [Accepted: 02/14/2024] [Indexed: 05/30/2024]
Abstract
ABSTRACT Splicing is a posttranscriptional RNA processing mechanism that enhances genomic complexity by creating multiple isoforms from the same gene. We aimed to characterize the isoforms expressed in the human peripheral nervous system, with the goal of creating a resource to identify novel isoforms of functionally relevant genes associated with somatosensation and nociception. We used long-read sequencing to document isoform expression in the human dorsal root ganglia from 3 organ donors and validated in silico by confirming expression in short-read sequencing from 3 independent organ donors. Nineteen thousand five hundred forty-seven isoforms of protein-coding genes were detected and validated. We identified 763 isoforms with at least one previously undescribed splice junction. Previously unannotated isoforms of multiple pain-associated genes, including ASIC3 , MRGPRX1 , and HNRNPK , were identified. In the novel isoforms of ASIC3 , a region comprising approximately 35% of the 5'UTR was excised. By contrast, a novel splice junction was used in isoforms of MRGPRX1 to include an additional exon upstream of the start codon, consequently adding a region to the 5'UTR. Novel isoforms of HNRNPK were identified, which used previously unannotated splice sites to both excise exon 14 and include a sequence in the 3' end of exon 13. This novel insertion is predicted to introduce a tyrosine phosphorylation site potentially phosphorylated by SRC. We also independently confirm a recently reported DRG-specific splicing event in WNK1 that gives insight into how painless peripheral neuropathy occurs when this gene is mutated. Our findings give a clear overview of mRNA isoform diversity in the human dorsal root ganglia obtained using long-read sequencing.
Collapse
Affiliation(s)
- Asta Arendt-Tranholm
- School of Behavioral and Brain Sciences, Department of Neuroscience and Center for Advanced Pain Studies, The University of Texas at Dallas, 800 W Campbell Rd, Richardson, Texas, 75080
| | - Juliet M. Mwirigi
- School of Behavioral and Brain Sciences, Department of Neuroscience and Center for Advanced Pain Studies, The University of Texas at Dallas, 800 W Campbell Rd, Richardson, Texas, 75080
| | - Theodore J. Price
- School of Behavioral and Brain Sciences, Department of Neuroscience and Center for Advanced Pain Studies, The University of Texas at Dallas, 800 W Campbell Rd, Richardson, Texas, 75080
| |
Collapse
|
11
|
Zebochin I, Denk F, Nochi Z. Modeling neuropathic pain in a dish. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2024; 179:233-278. [PMID: 39580214 DOI: 10.1016/bs.irn.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2024]
Abstract
The study of pain mechanisms has advanced significantly with the development of innovative in vitro models. This chapter explores those already used in or potentially useful for neuropathic pain research, emphasizing the complementary roles of animal and human cellular models to enhance translational success. Traditional animal models have provided foundational insights into the neurobiology of pain and remain invaluable for understanding complex pain pathways. However, integrating human cellular models addresses the need for better replication of human nociceptors. The chapter details methodologies for culturing rodent and human primary sensory neurons, including isolation and culture techniques, advantages, and limitations. It highlights the application of these models in neuropathic pain research, such as identifying pain-associated receptors and ion channels. Recent advancements in using induced pluripotent stem cell (iPSC)-derived sensory neurons are also discussed. Finally, the chapter explores advanced in vitro models, including 2D co-cultures and 3D organoids, and their implications for studying neuropathic pain. These models offer significant advantages for drug screening and ethical research practices, providing a more accurate representation of human pain pathways and paving the way for innovative therapeutic strategies. Despite challenges such as limited access to viable human tissue and variability between samples, these in vitro models, alongside traditional animal models, are indispensable for advancing our understanding of neuropathic pain and developing effective treatments.
Collapse
Affiliation(s)
- Irene Zebochin
- Wolfson Sensory Pain and Regeneration Centre (SPaRC), King's College London
| | - Franziska Denk
- Wolfson Sensory Pain and Regeneration Centre (SPaRC), King's College London
| | - Zahra Nochi
- Danish Pain Research Centre, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
12
|
Asiri YI, Moni SS, Ramar M, Chidambaram K. Advancing Pain Understanding and Drug Discovery: Insights from Preclinical Models and Recent Research Findings. Pharmaceuticals (Basel) 2024; 17:1439. [PMID: 39598351 PMCID: PMC11597627 DOI: 10.3390/ph17111439] [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: 07/21/2024] [Revised: 10/19/2024] [Accepted: 10/21/2024] [Indexed: 11/29/2024] Open
Abstract
Despite major advancements in our understanding of its fundamental causes, pain-both acute and chronic-remains a serious health concern. Various preclinical investigations utilizing diverse animal, cellular, and alternative models are required and frequently demanded by regulatory approval bodies to bridge the gap between the lab and the clinic. Investigating naturally occurring painful disorders can speed up medication development at the preclinical and clinical levels by illuminating molecular pathways. A wide range of animal models related to pain have been developed to elucidate pathophysiological mechanisms and aid in identifying novel targets for treatment. Pain sometimes drugs fail clinically, causing high translational costs due to poor selection and the use of preclinical tools and reporting. To improve the study of pain in a clinical context, researchers have been creating innovative models over the past few decades that better represent pathological pain conditions. In this paper, we provide a summary of traditional animal models, including rodents, cellular models, human volunteers, and alternative models, as well as the specific characteristics of pain diseases they model. However, a more rigorous approach to preclinical research and cutting-edge analgesic technologies may be necessary to successfully create novel analgesics. The research highlights from this review emphasize new opportunities to develop research that includes animals and non-animals using proven methods pertinent to comprehending and treating human suffering. This review highlights the value of using a variety of modern pain models in animals before human trials. These models can help us understand the different mechanisms behind various pain types. This will ultimately lead to the development of more effective pain medications.
Collapse
Affiliation(s)
- Yahya I. Asiri
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha 62521, Saudi Arabia;
| | - Sivakumar S. Moni
- Health Research Centre, Jazan University, Jazan 45142, Saudi Arabia;
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Mohankumar Ramar
- Department of Pharmaceutical Sciences, UConn School of Pharmacy, University of Connecticut, Storrs, CT 06269, USA;
| | - Kumarappan Chidambaram
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha 62521, Saudi Arabia;
| |
Collapse
|
13
|
Flayer CH, Kernin IJ, Matatia PR, Zeng X, Yarmolinsky DA, Han C, Naik PR, Buttaci DR, Aderhold PA, Camire RB, Zhu X, Tirard AJ, McGuire JT, Smith NP, McKimmie CS, McAlpine CS, Swirski FK, Woolf CJ, Villani AC, Sokol CL. A γδ T cell-IL-3 axis controls allergic responses through sensory neurons. Nature 2024; 634:440-446. [PMID: 39232162 DOI: 10.1038/s41586-024-07869-0] [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: 01/19/2023] [Accepted: 07/23/2024] [Indexed: 09/06/2024]
Abstract
In naive individuals, sensory neurons directly detect and respond to allergens, leading to both the sensation of itch and the activation of local innate immune cells, which initiate the allergic immune response1,2. In the setting of chronic allergic inflammation, immune factors prime sensory neurons, causing pathologic itch3-7. Although these bidirectional neuroimmune circuits drive responses to allergens, whether immune cells regulate the set-point for neuronal activation by allergens in the naive state is unknown. Here we describe a γδ T cell-IL-3 signalling axis that controls the allergen responsiveness of cutaneous sensory neurons. We define a poorly characterized epidermal γδ T cell subset8, termed GD3 cells, that produces its hallmark cytokine IL-3 to promote allergic itch and the initiation of the allergic immune response. Mechanistically, IL-3 acts on Il3ra-expressing sensory neurons in a JAK2-dependent manner to lower their threshold for allergen activation without independently eliciting itch. This γδ T cell-IL-3 signalling axis further acts by means of STAT5 to promote neuropeptide production and the initiation of allergic immunity. These results reveal an endogenous immune rheostat that sits upstream of and governs sensory neuronal responses to allergens on first exposure. This pathway may explain individual differences in allergic susceptibility and opens new therapeutic avenues for treating allergic diseases.
Collapse
Affiliation(s)
- Cameron H Flayer
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Isabela J Kernin
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Peri R Matatia
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Xiangsunze Zeng
- FM Kirby Center, Boston Children's Hospital and Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - David A Yarmolinsky
- FM Kirby Center, Boston Children's Hospital and Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Cai Han
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Parth R Naik
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Dean R Buttaci
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Pamela A Aderhold
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ryan B Camire
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Xueping Zhu
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Alice J Tirard
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - John T McGuire
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Neal P Smith
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Clive S McKimmie
- Virus Host Interaction Team, Skin Research Centre, University of York, York, UK
| | - Cameron S McAlpine
- Cardiovascular Research Institute and the Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute and the Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Filip K Swirski
- Cardiovascular Research Institute and the Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Clifford J Woolf
- FM Kirby Center, Boston Children's Hospital and Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Alexandra-Chloe Villani
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Caroline L Sokol
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
14
|
Sankaranarayanan I, Kume M, Mohammed A, Mwirigi JM, Inturi NN, Munro G, Petersen KA, Tavares-Ferreira D, Price TJ. Persistent changes in nociceptor translatomes govern hyperalgesic priming in mouse models. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.07.606891. [PMID: 39149295 PMCID: PMC11326310 DOI: 10.1101/2024.08.07.606891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Hyperalgesic priming is a model system that has been widely used to understand plasticity in painful stimulus-detecting sensory neurons, called nociceptors. A key feature of this model system is that following priming, stimuli that do not normally cause hyperalgesia now readily provoke this state. We hypothesized that hyperalgesic priming occurs due to reorganization of translation of mRNA in nociceptors. To test this hypothesis, we used paclitaxel treatment as the priming stimulus and translating ribosome affinity purification (TRAP) to measure persistent changes in mRNA translation in Nav1.8+ nociceptors. TRAP sequencing revealed 161 genes with persistently altered mRNA translation in the primed state. We identified Gpr88 as upregulated and Metrn as downregulated. We confirmed a functional role for these genes, wherein a GPR88 agonist causes pain only in primed mice and established hyperalgesic priming is reversed by Meteorin. Our work demonstrates that altered nociceptor translatomes are causative in producing hyperalgesic priming.
Collapse
Affiliation(s)
- Ishwarya Sankaranarayanan
- Pain Neurobiology Research Group, Department of Neuroscience, Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas
| | - Moeno Kume
- Pain Neurobiology Research Group, Department of Neuroscience, Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas
| | - Ayaan Mohammed
- Pain Neurobiology Research Group, Department of Neuroscience, Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas
| | - Juliet M Mwirigi
- Pain Neurobiology Research Group, Department of Neuroscience, Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas
| | - Nikhil Nageswar Inturi
- Pain Neurobiology Research Group, Department of Neuroscience, Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas
| | | | | | - Diana Tavares-Ferreira
- Pain Neurobiology Research Group, Department of Neuroscience, Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas
| | - Theodore J Price
- Pain Neurobiology Research Group, Department of Neuroscience, Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas
| |
Collapse
|
15
|
Higham JP, Bhebhe CN, Gupta RA, Tranter MM, Barakat FM, Dogra H, Bab N, Wozniak E, Barker KH, Wilson CH, Mein CA, Raine T, Cox JJ, Wood JN, Croft NM, Wright PD, Bulmer DC. Transcriptomic profiling reveals a pronociceptive role for angiotensin II in inflammatory bowel disease. Pain 2024; 165:1592-1604. [PMID: 38293826 PMCID: PMC11190897 DOI: 10.1097/j.pain.0000000000003159] [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: 07/26/2023] [Revised: 10/20/2023] [Accepted: 10/29/2023] [Indexed: 02/01/2024]
Abstract
ABSTRACT Visceral pain is a leading cause of morbidity in inflammatory bowel disease (IBD), contributing significantly to reduced quality of life. Currently available analgesics often lack efficacy or have intolerable side effects, driving the need for a more complete understanding of the mechanisms causing pain. Whole transcriptome gene expression analysis was performed by bulk RNA sequencing of colonic biopsies from patients with ulcerative colitis (UC) and Crohn's disease (CD) reporting abdominal pain and compared with noninflamed control biopsies. Potential pronociceptive mediators were identified based on gene upregulation in IBD biopsy tissue and cognate receptor expression in murine colonic sensory neurons. Pronociceptive activity of identified mediators was assessed in assays of sensory neuron and colonic afferent activity. RNA sequencing analysis highlighted a 7.6-fold increase in the expression of angiotensinogen transcripts, Agt , which encode the precursor to angiotensin II (Ang II), in samples from UC patients ( P = 3.2 × 10 -8 ). Consistent with the marked expression of the angiotensin AT 1 receptor in colonic sensory neurons, Ang II elicited an increase in intracellular Ca 2+ in capsaicin-sensitive, voltage-gated sodium channel subtype Na V 1.8-positive sensory neurons. Ang II also evoked action potential discharge in high-threshold colonic nociceptors. These effects were inhibited by the AT 1 receptor antagonist valsartan. Findings from our study identify AT 1 receptor-mediated colonic nociceptor activation as a novel pathway of visceral nociception in patients with UC. This work highlights the potential utility of angiotensin receptor blockers, such as valsartan, as treatments for pain in IBD.
Collapse
Affiliation(s)
- James P. Higham
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
| | - Charity N. Bhebhe
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
| | - Rohit A. Gupta
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
| | - Michael M. Tranter
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Farah M. Barakat
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Harween Dogra
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Natalie Bab
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Eva Wozniak
- Genome Centre, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Katie H. Barker
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
| | - Catherine H. Wilson
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
| | - Charles A. Mein
- Genome Centre, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Tim Raine
- Department of Gastroenterology, Addenbrookes Hospital, Cambridge University Teaching Hospitals, Cambridge, United Kingdom
| | - James J. Cox
- Wolfson Institute for Biomedical Research, University College London, London, United Kingdom
| | - John N. Wood
- Wolfson Institute for Biomedical Research, University College London, London, United Kingdom
| | - Nicholas M. Croft
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Paul D. Wright
- LifeArc, SBC Open Innovation Campus, Stevenage, United Kingdom
| | - David C. Bulmer
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
16
|
Zurek NA, Ehsanian R, Goins AE, Adams IM, Petersen T, Goyal S, Shilling M, Westlund KN, Alles SRA. Electrophysiological Analyses of Human Dorsal Root Ganglia and Human Induced Pluripotent Stem Cell-derived Sensory Neurons From Male and Female Donors. THE JOURNAL OF PAIN 2024; 25:104451. [PMID: 38154622 PMCID: PMC11128351 DOI: 10.1016/j.jpain.2023.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/15/2023] [Accepted: 12/16/2023] [Indexed: 12/30/2023]
Abstract
Human induced pluripotent stem cell-derived sensory neurons (hiPSC-SNs) and human dorsal root ganglia neurons (hDRG-N) are popular tools in the field of pain research; however, few groups make use of both approaches. For screening and analgesic validation purposes, important characterizations can be determined of the similarities and differences between hDRG-N and hiPSC-SNs. This study focuses specifically on the electrophysiology properties of hDRG-N in comparison to hiPSC-SNs. We also compared hDRG-N and hiPSC-SNs from both male and female donors to evaluate potential sex differences. We recorded neuronal size, rheobase, resting membrane potential, input resistance, and action potential waveform properties from 83 hiPSCs-SNs (2 donors) and 108 hDRG-N neurons (8 donors). We observed several statistically significant electrophysiological differences between hDRG-N and hiPSC-SNs, such as size, rheobase, input resistance, and several action potential waveform properties. Correlation analysis also revealed many properties that were positively or negatively correlated, some of which were differentially correlated between hDRG-N and hiPSC-SNs. This study shows several differences between hDRG-N and hiPSC-SNs and allows a better understanding of the advantages and disadvantages of both for use in pain research. We hope this study will be a valuable resource for pain researchers considering the use of these human in vitro systems for mechanistic studies and/or drug development projects. PERSPECTIVE: hiPSC-SNs and hDRG-N are popular tools in the field of pain research. This study allows for a better functional understanding of the pros and cons of both tools.
Collapse
Affiliation(s)
- Nesia A Zurek
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Reza Ehsanian
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Aleyah E Goins
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Ian M Adams
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Timothy Petersen
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Sachin Goyal
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Mark Shilling
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Karin N Westlund
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Sascha R A Alles
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico.
| |
Collapse
|
17
|
Allison RL, Welby E, Ehlers V, Burand A, Isaeva O, Nieves Torres D, Highland J, Brandow AM, Stucky CL, Ebert AD. Sickle cell disease iPSC-derived sensory neurons exhibit increased excitability and sensitization to patient plasma. Blood 2024; 143:2037-2052. [PMID: 38427938 PMCID: PMC11143522 DOI: 10.1182/blood.2023022591] [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: 09/18/2023] [Revised: 01/25/2024] [Accepted: 02/14/2024] [Indexed: 03/03/2024] Open
Abstract
ABSTRACT Individuals living with sickle cell disease (SCD) experience severe recurrent acute and chronic pain. Challenges to gaining mechanistic insight into pathogenic SCD pain processes include differential gene expression and function of sensory neurons between humans and mice with SCD, and extremely limited availability of neuronal tissues from patients with SCD. Here, we used induced pluripotent stem cells (iPSCs), derived from patients with SCD, differentiated into sensory neurons (SCD iSNs) to begin to overcome these challenges. We characterize key gene expression and function of SCD iSNs to establish a model to investigate intrinsic and extrinsic factors that may contribute to SCD pain. Despite similarities in receptor gene expression, SCD iSNs show pronounced excitability using patch clamp electrophysiology. Furthermore, we find that plasma taken from patients with SCD during acute pain associated with a vaso-occlusive event increases the calcium responses to the nociceptive stimulus capsaicin in SCD iSNs compared with those treated with paired plasma from patients with SCD at steady state baseline or healthy control plasma samples. We identified high levels of the polyamine spermine in baseline and acute pain states of plasma from patients with SCD, which sensitizes SCD iSNs to subthreshold concentrations of capsaicin. Together, these data identify potential intrinsic mechanisms within SCD iSNs that may extend beyond a blood-based pathology.
Collapse
Affiliation(s)
- Reilly L. Allison
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI
| | - Emily Welby
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI
| | - Vanessa Ehlers
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI
| | - Anthony Burand
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI
| | - Olena Isaeva
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI
| | - Damaris Nieves Torres
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI
| | - Janelle Highland
- Department of Pediatrics, Section of Hematology/Oncology/Bone Marrow Transplantation, Medical College of Wisconsin, Milwaukee, WI
| | - Amanda M. Brandow
- Department of Pediatrics, Section of Hematology/Oncology/Bone Marrow Transplantation, Medical College of Wisconsin, Milwaukee, WI
| | - Cheryl L. Stucky
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI
| | - Allison D. Ebert
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI
| |
Collapse
|
18
|
Rojas-Galvan NS, Ciotu CI, Heber S, Fischer MJ. Correlation of TRPA1 RNAscope and Agonist Responses. J Histochem Cytochem 2024; 72:275-287. [PMID: 38725415 PMCID: PMC11107437 DOI: 10.1369/00221554241251904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 04/09/2024] [Indexed: 05/18/2024] Open
Abstract
The TRPA1 ion channel is a sensitive detector of reactive chemicals, found primarily on sensory neurons. The phenotype exhibited by mice lacking TRPA1 suggests its potential as a target for pharmacological intervention. Antibody-based detection for distribution analysis is a standard technique. In the case of TRPA1, however, there is no antibody with a plausible validation in knockout animals or functional studies, but many that have failed in this regard. To this end we employed the single molecule in situ hybridization technique RNAscope on sensory neurons immediately after detection of calcium responses to the TRPA1 agonist allyl isothiocyanate. There is a clearly positive correlation between TRPA1 calcium imaging and RNAscope detection (R = 0.43), although less than what might have been expected. Thus, the technique of choice should be carefully considered to suit the research question. The marginal correlation between TRPV1 RNAscope and the specific agonist capsaicin indicates that such validation is advisable for every RNAscope target. Given the recent description of a long-awaited TRPA1 reporter mouse, TRPA1 RNAscope detection might still have its use cases, for detection of RNA at particular sites, for example, defined structurally or by other molecular markers.
Collapse
Affiliation(s)
- Natalia S. Rojas-Galvan
- Centre for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria and Randall Centre for Cell & Molecular Biophysics, King’s College London, London, UK
| | - Cosmin I. Ciotu
- Centre for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Stefan Heber
- Centre for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Michael J.M. Fischer
- Centre for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
19
|
Inyang KE, Evans CM, Heussner M, Petroff M, Reimers M, Vermeer PD, Tykocki N, Folger JK, Laumet G. HPV+ head and neck cancer-derived small extracellular vesicles communicate with TRPV1+ neurons to mediate cancer pain. Pain 2024; 165:608-620. [PMID: 37678566 PMCID: PMC10915104 DOI: 10.1097/j.pain.0000000000003045] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 06/15/2023] [Indexed: 09/09/2023]
Abstract
ABSTRACT Severe pain is often experienced by patients with head and neck cancer and is associated with a poor prognosis. Despite its frequency and severity, current treatments fail to adequately control cancer-associated pain because of our lack of mechanistic understanding. Although recent works have shed some light of the biology underlying pain in HPV-negative oral cancers, the mechanisms mediating pain in HPV+ cancers remain unknown. Cancer-derived small extracellular vesicles (cancer-sEVs) are well positioned to function as mediators of communication between cancer cells and neurons. Inhibition of cancer-sEV release attenuated pain in tumor-bearing mice. Injection of purified cancer-sEVs is sufficient to induce pain hypersensitivity in naive mice that is prevented by QX-314 treatment and in Trpv1-/- mice. Cancer-sEVs triggered calcium influx in nociceptors, and inhibition or ablation of nociceptors protects against cancer pain. Interrogation of published sequencing data of human sensory neurons exposed to human cancer-sEVs suggested a stimulation of protein translation in neurons. Induction of translation by cancer-sEVs was validated in our mouse model, and its inhibition alleviated cancer pain in mice. In summary, our work reveals that HPV+ head and neck squamous cell carcinoma-derived sEVs alter TRPV1+ neurons by promoting nascent translation to mediate cancer pain and identified several promising therapeutic targets to interfere with this pathway.
Collapse
Affiliation(s)
| | - Christine M. Evans
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Matthew Heussner
- Department of Physiology, Michigan State University, East Lansing, MI, USA
- College of Osteopathic Medicine, Michigan State University, East Lansing, MI
| | - Margaret Petroff
- Department of Pathology Michigan State University College of Veterinary Medicine, East Lansing, MI
| | - Mark Reimers
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Paola D. Vermeer
- Cancer Biology and Immunotherapies Group, Sanford Research, Sioux Falls, South Dakota
| | - Nathan Tykocki
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI
| | - Joseph K. Folger
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Geoffroy Laumet
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
20
|
Stewart RG, Camacena M, Copits BA, Sack JT. Distinct cellular expression and subcellular localization of Kv2 voltage-gated K + channel subtypes in dorsal root ganglion neurons conserved between mice and humans. J Comp Neurol 2024; 532:e25575. [PMID: 38335058 PMCID: PMC10861167 DOI: 10.1002/cne.25575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 08/07/2023] [Accepted: 10/03/2023] [Indexed: 02/12/2024]
Abstract
The distinct organization of Kv2 voltage-gated potassium channels on and near the cell body of brain neurons enables their regulation of action potentials and specialized membrane contact sites. Somatosensory neurons have a pseudounipolar morphology and transmit action potentials from peripheral nerve endings through axons that bifurcate to the spinal cord and the cell body within ganglia including the dorsal root ganglia (DRG). Kv2 channels regulate action potentials in somatosensory neurons, yet little is known about where Kv2 channels are located. Here, we define the cellular and subcellular localization of the Kv2 paralogs, Kv2.1 and Kv2.2, in DRG somatosensory neurons with a panel of antibodies, cell markers, and genetically modified mice. We find that relative to spinal cord neurons, DRG neurons have similar levels of detectable Kv2.1 and higher levels of Kv2.2. In older mice, detectable Kv2.2 remains similar, while detectable Kv2.1 decreases. Both Kv2 subtypes adopt clustered subcellular patterns that are distinct from central neurons. Most DRG neurons co-express Kv2.1 and Kv2.2, although neuron subpopulations show preferential expression of Kv2.1 or Kv2.2. We find that Kv2 protein expression and subcellular localization are similar between mouse and human DRG neurons. We conclude that the organization of both Kv2 channels is consistent with physiological roles in the somata and stem axons of DRG neurons. The general prevalence of Kv2.2 in DRG as compared to central neurons and the enrichment of Kv2.2 relative to detectable Kv2.1 in older mice, proprioceptors, and axons suggest more widespread roles for Kv2.2 in DRG neurons.
Collapse
Affiliation(s)
- Robert G Stewart
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, California, USA
| | - Miriam Camacena
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, California, USA
| | - Bryan A Copits
- Washington University Pain Center, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Jon T Sack
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, California, USA
- Department of Anesthesiology and Pain Medicine, University of California, Davis, Davis, California, USA
| |
Collapse
|
21
|
Sabbir MG. Cholinergic Receptor Muscarinic 1 Co-Localized with Mitochondria in Cultured Dorsal Root Ganglion Neurons, and Its Deletion Disrupted Mitochondrial Ultrastructure in Peripheral Neurons: Implications in Alzheimer's Disease. J Alzheimers Dis 2024; 98:247-264. [PMID: 38427478 DOI: 10.3233/jad-230883] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2024]
Abstract
Background Loss of Cholinergic Receptor Muscarinic 1 (CHRM1) has been linked to the pathogenesis of Alzheimer's disease (AD). Our recent study found significantly lower CHRM1 protein levels in AD patient cortices, linked to reduced survival. Furthermore, using knockout mice (Chrm1-/-) we demonstrated that deletion of Chrm1 alters cortical mitochondrial structure and function, directly establishing a connection between its loss and mitochondrial dysfunction in the context of AD. While CHRM1's role in the brain has been extensively investigated, its impact on peripheral neurons in AD remains a crucial area of research, especially considering reported declines in peripheral nerve conduction among AD patients. Objective The objective was to characterize Chrm1 localization and mitochondrial deficits in Chrm1-/- dorsal root ganglion (DRG) neurons. Methods Recombinant proteins tagged with Green or Red Fluorescent Protein (GFP/RFP) were transiently expressed to investigate the localization of Chrm1 and mitochondria, as well as mitochondrial movement in the neurites of cultured primary mouse DRG neurons, using confocal time-lapse live cell imaging. Transmission electron microscopy was performed to examine the ultrastructure of mitochondria in both wild-type and Chrm1-/- DRGs. Results Fluorescence imaging revealed colocalization and comigration of N-terminal GFP-tagged Chrm1 and mitochondrial localization signal peptide-tagged RFP-labelled mitochondria in the DRGs neurons. A spectrum of mitochondrial structural abnormalities, including disruption and loss of cristae was observed in 87% neurons in Chrm1-/- DRGs. Conclusions This study suggests that Chrm1 may be localized in the neuronal mitochondria and loss of Chrm1 in peripheral neurons causes sever mitochondrial structural aberrations resembling AD pathology.
Collapse
Affiliation(s)
- Mohammad Golam Sabbir
- Department of Psychology and Neuroscience, Collegeof Psychology, Nova Southeastern University, Fort Lauderdale, FL, USA
- Alzo Biosciences Inc., San Diego, CA, USA
| |
Collapse
|
22
|
Stewart RG, Camacena M, Copits BA, Sack JT. Distinct cellular expression and subcellular localization of Kv2 voltage-gated K + channel subtypes in dorsal root ganglion neurons conserved between mice and humans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.01.530679. [PMID: 38187582 PMCID: PMC10769185 DOI: 10.1101/2023.03.01.530679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
The distinct organization of Kv2 voltage-gated potassium channels on and near the cell body of brain neurons enables their regulation of action potentials and specialized membrane contact sites. Somatosensory neurons have a pseudounipolar morphology and transmit action potentials from peripheral nerve endings through axons that bifurcate to the spinal cord and the cell body within ganglia including the dorsal root ganglia (DRG). Kv2 channels regulate action potentials in somatosensory neurons, yet little is known about where Kv2 channels are located. Here we define the cellular and subcellular localization of the Kv2 paralogs, Kv2.1 and Kv2.2, in DRG somatosensory neurons with a panel of antibodies, cell markers, and genetically modified mice. We find that relative to spinal cord neurons, DRG neurons have similar levels of detectable Kv2.1, and higher levels of Kv2.2. In older mice, detectable Kv2.2 remains similar while detectable Kv2.1 decreases. Both Kv2 subtypes adopt clustered subcellular patterns that are distinct from central neurons. Most DRG neurons co-express Kv2.1 and Kv2.2, although neuron subpopulations show preferential expression of Kv2.1 or Kv2.2. We find that Kv2 protein expression and subcellular localization is similar between mouse and human DRG neurons. We conclude that the organization of both Kv2 channels is consistent with physiological roles in the somata and stem axons of DRG neurons. The general prevalence of Kv2.2 in DRG as compared to central neurons and the enrichment of Kv2.2 relative to detectable Kv2.1, in older mice, proprioceptors, and axons suggest more widespread roles for Kv2.2 in DRG neurons.
Collapse
Affiliation(s)
- Robert G Stewart
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA 95616, USA
| | - Miriam Camacena
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA 95616, USA
| | - Bryan A Copits
- Washington University Pain Center, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jon T Sack
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA 95616, USA
- Department of Anesthesiology and Pain Medicine, University of California Davis, Davis, CA 95616, USA
| |
Collapse
|
23
|
Zurek NA, Ehsanian R, Goins AE, Adams IM, Petersen T, Goyal S, Shilling M, Westlund KN, Alles SRA. Electrophysiological analyses of human dorsal root ganglia and human induced pluripotent stem cell-derived sensory neurons from male and female donors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.03.565343. [PMID: 37961669 PMCID: PMC10635102 DOI: 10.1101/2023.11.03.565343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Human induced pluripotent stem cell-derived sensory neurons (hiPSC-SNs) and human dorsal root ganglia (hDRG) neurons are popular tools in the field of pain research; however, few groups make use of both approaches. For screening and analgesic validation purposes, important characterizations can be determined of the similarities and differences between hDRG and hiPSC-SNs. This study focuses specifically on electrophysiology properties of hDRG in comparison to hiPSC-SNs. We also compared hDRG and hiPSC-SNs from both male and female donors to evaluate potential sex differences. We recorded neuronal size, rheobase, resting membrane potential, input resistance, and action potential waveform properties from 83 hiPSCs-SNs (2 donors) and 108 hDRG neurons (9 donors). We observed several statistically significant electrophysiological differences between hDRG and hiPSC-SNs, such as size, rheobase, input resistance, and several actional potential (AP) waveform properties. Correlation analysis also revealed many properties that were positively or negatively correlated, some of which were differentially correlated between hDRG and hiPSC-SNs. This study shows several differences between hDRG and hiPSC-SNs and allows better understanding of the advantages and disadvantages of both for use in pain research. We hope this study will be a valuable resource for pain researchers considering the use of these human in vitro systems for mechanistic studies and/or drug development projects.
Collapse
|
24
|
Ochandarena NE, Niehaus JK, Tassou A, Scherrer G. Cell-type specific molecular architecture for mu opioid receptor function in pain and addiction circuits. Neuropharmacology 2023; 238:109597. [PMID: 37271281 PMCID: PMC10494323 DOI: 10.1016/j.neuropharm.2023.109597] [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: 04/12/2023] [Accepted: 05/13/2023] [Indexed: 06/06/2023]
Abstract
Opioids are potent analgesics broadly used for pain management; however, they can produce dangerous side effects including addiction and respiratory depression. These harmful effects have led to an epidemic of opioid abuse and overdose deaths, creating an urgent need for the development of both safer pain medications and treatments for opioid use disorders. Both the analgesic and addictive properties of opioids are mediated by the mu opioid receptor (MOR), making resolution of the cell types and neural circuits responsible for each of the effects of opioids a critical research goal. Single-cell RNA sequencing (scRNA-seq) technology is enabling the identification of MOR-expressing cell types throughout the nervous system, creating new opportunities for mapping distinct opioid effects onto newly discovered cell types. Here, we describe molecularly defined MOR-expressing neuronal cell types throughout the peripheral and central nervous systems and their potential contributions to opioid analgesia and addiction.
Collapse
Affiliation(s)
- Nicole E Ochandarena
- Neuroscience Curriculum, Biological and Biomedical Sciences Program, The University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA; Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| | - Jesse K Niehaus
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Adrien Tassou
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Grégory Scherrer
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; New York Stem Cell Foundation - Robertson Investigator, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
25
|
Arendt-Tranholm A, Mwirigi JM, Price TJ. RNA isoform expression landscape of the human dorsal root ganglion (DRG) generated from long read sequencing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.28.564535. [PMID: 37961262 PMCID: PMC10634934 DOI: 10.1101/2023.10.28.564535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Splicing is a post-transcriptional RNA processing mechanism that enhances genomic complexity by creating multiple isoforms from the same gene. Diversity in splicing in the mammalian nervous system is associated with neuronal development, synaptic function and plasticity, and is also associated with diseases of the nervous system ranging from neurodegeneration to chronic pain. We aimed to characterize the isoforms expressed in the human peripheral nervous system, with the goal of creating a resource to identify novel isoforms of functionally relevant genes associated with somatosensation and nociception. We used long read sequencing (LRS) to document isoform expression in the human dorsal root ganglia (hDRG) from 3 organ donors. Isoforms were validated in silico by confirming expression in hDRG short read sequencing (SRS) data from 3 independent organ donors. 19,547 isoforms of protein-coding genes were detected using LRS and validated with SRS and strict expression cutoffs. We identified 763 isoforms with at least one previously undescribed splice-junction. Previously unannotated isoforms of multiple pain-associated genes, including ASIC3, MRGPRX1 and HNRNPK were identified. In the novel isoforms of ASIC3, a region comprising ~35% of the 5'UTR was excised. In contrast, a novel splice-junction was utilized in isoforms of MRGPRX1 to include an additional exon upstream of the start-codon, consequently adding a region to the 5'UTR. Novel isoforms of HNRNPK were identified which utilized previously unannotated splice-sites to both excise exon 14 and include a sequence in the 5' end of exon 13. The insertion and deletion in the coding region was predicted to excise a serine-phosphorylation site favored by cdc2, and replace it with a tyrosine-phosphorylation site potentially phosphorylated by SRC. We also independently confirm a recently reported DRG-specific splicing event in WNK1 that gives insight into how painless peripheral neuropathy occurs when this gene is mutated. Our findings give a clear overview of mRNA isoform diversity in the hDRG obtained using LRS. Using this work as a foundation, an important next step will be to use LRS on hDRG tissues recovered from people with a history of chronic pain. This should enable identification of new drug targets and a better understanding of chronic pain that may involve aberrant splicing events.
Collapse
Affiliation(s)
- Asta Arendt-Tranholm
- School of Behavioral and Brain Sciences, Department of Neuroscience and Center for Advanced Pain Studies, The University of Texas at Dallas, 800 W Campbell Rd, Richardson, Texas, 75080
| | - Juliet M. Mwirigi
- School of Behavioral and Brain Sciences, Department of Neuroscience and Center for Advanced Pain Studies, The University of Texas at Dallas, 800 W Campbell Rd, Richardson, Texas, 75080
| | - Theodore J. Price
- School of Behavioral and Brain Sciences, Department of Neuroscience and Center for Advanced Pain Studies, The University of Texas at Dallas, 800 W Campbell Rd, Richardson, Texas, 75080
| |
Collapse
|
26
|
Haberberger RV, Kuramatilake J, Barry CM, Matusica D. Ultrastructure of dorsal root ganglia. Cell Tissue Res 2023:10.1007/s00441-023-03770-w. [PMID: 37079097 PMCID: PMC10115609 DOI: 10.1007/s00441-023-03770-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 03/28/2023] [Indexed: 04/21/2023]
Abstract
Dorsal root ganglia (DRG) contains thousands of sensory neurons that transmit information about our external and internal environment to the central nervous system. This includes signals related to proprioception, temperature, and nociception. Our understanding of DRG has increased tremendously over the last 50 years and has established the DRG as an active participant in peripheral processes. This includes interactions between neurons and non-neuronal cells such as satellite glia cells and macrophages that contribute to an increasingly complex cellular environment that modulates neuronal function. Early ultrastructural investigations of the DRG have described subtypes of sensory neurons based on differences in the arrangement of organelles such as the Golgi apparatus and the endoplasmic reticulum. The neuron-satellite cell complex and the composition of the axon hillock in DRG have also been investigated, but, apart from basic descriptions of Schwann cells, ultrastructural investigations of other cell types in DRG are limited. Furthermore, detailed descriptions of key components of DRG, such as blood vessels and the capsule that sits at the intersection of the meninges and the connective tissue covering the peripheral nervous system, are lacking to date. With rising interest in DRG as potential therapeutic targets for aberrant signalling associated with chronic pain conditions, gaining further insights into DRG ultrastructure will be fundamental to understanding cell-cell interactions that modulate DRG function. In this review, we aim to provide a synopsis of the current state of knowledge on the ultrastructure of the DRG and its components, as well as to identify areas of interest for future studies.
Collapse
Affiliation(s)
- Rainer Viktor Haberberger
- Division of Anatomy and Pathology, School of Biomedicine, The University of Adelaide, Adelaide, Australia.
| | - Jaliya Kuramatilake
- Division of Anatomy and Pathology, School of Biomedicine, The University of Adelaide, Adelaide, Australia
| | - Christine M Barry
- Anatomy, Histology & Pathology, College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - Dusan Matusica
- Anatomy, Histology & Pathology, College of Medicine and Public Health, Flinders University, Adelaide, Australia
| |
Collapse
|
27
|
Shiers S, Sahn JJ, Price TJ. MNK1 and MNK2 Expression in the Human Dorsal Root and Trigeminal Ganglion. Neuroscience 2023; 515:96-107. [PMID: 36764601 DOI: 10.1016/j.neuroscience.2023.01.039] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 01/28/2023] [Accepted: 01/31/2023] [Indexed: 02/11/2023]
Abstract
Mitogen activated protein kinase interacting kinases (MNK) 1 and 2 are serine/threonine protein kinases that play an important role in translation of mRNAs through their phosphorylation of the RNA 5'-cap binding protein, eukaryotic translation initiation factor (eIF) 4E. These kinases are downstream targets for mitogen activated protein kinases (MAPKs), extracellular activity regulated protein kinase (ERK) and p38. MNKs have been implicated in the sensitization of peripheral nociceptors of the dorsal root and trigeminal ganglion (DRG and TG) using transgenic mouse lines and through the use of specific inhibitors of MNK1 and MNK2. While specific knockout of the Mknk1 gene suggests that it is the key isoform for regulation of nociceptor excitability and nociceptive behaviors in mice, both MKNK1 and MKNK2 genes are expressed in the DRG and TG of mice and humans based on RNA sequencing experiments. Single cell sequencing in mice suggests that Mknk1 and Mknk2 may be expressed in different populations of nociceptors. We sought to characterize mRNA expression in human DRG and TG (N = 3 ganglia for both DRG and TG) for both MNK1 and MNK2. Our results show that both genes are expressed by nearly all neurons in both human ganglia with expression in other cell types as well. Our findings provide evidence that MNK1 and MNK2 are expressed by human nociceptors of males and females and suggest that efforts to pharmacologically target MNKs for pain would likely be translatable due its conserved expression in both species.
Collapse
Affiliation(s)
- Stephanie Shiers
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, USA
| | | | - Theodore J Price
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, USA.
| |
Collapse
|
28
|
Aparicio GI, León A, Gutiérrez Fuster R, Ravenscraft B, Monje PV, Scorticati C. Endogenous Glycoprotein GPM6a Is Involved in Neurite Outgrowth in Rat Dorsal Root Ganglion Neurons. Biomolecules 2023; 13:biom13040594. [PMID: 37189342 DOI: 10.3390/biom13040594] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/09/2023] [Accepted: 03/17/2023] [Indexed: 03/29/2023] Open
Abstract
The peripheral nervous system (PNS) has a unique ability for self-repair. Dorsal root ganglion (DRG) neurons regulate the expression of different molecules, such as neurotrophins and their receptors, to promote axon regeneration after injury. However, the molecular players driving axonal regrowth need to be better defined. The membrane glycoprotein GPM6a has been described to contribute to neuronal development and structural plasticity in central-nervous-system neurons. Recent evidence indicates that GPM6a interacts with molecules from the PNS, although its role in DRG neurons remains unknown. Here, we characterized the expression of GPM6a in embryonic and adult DRGs by combining analysis of public RNA-seq datasets with immunochemical approaches utilizing cultures of rat DRG explants and dissociated neuronal cells. M6a was detected on the cell surfaces of DRG neurons throughout development. Moreover, GPM6a was required for DRG neurite elongation in vitro. In summary, we provide evidence on GPM6a being present in DRG neurons for the first time. Data from our functional experiments support the idea that GPM6a could contribute to axon regeneration in the PNS.
Collapse
|
29
|
Shen BQ, Sankaranarayanan I, Price TJ, Tavares-Ferreira D. Sex-differences in prostaglandin signaling: a semi-systematic review and characterization of PTGDS expression in human sensory neurons. Sci Rep 2023; 13:4670. [PMID: 36949072 PMCID: PMC10033690 DOI: 10.1038/s41598-023-31603-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 03/14/2023] [Indexed: 03/24/2023] Open
Abstract
There is increasing evidence of sex differences in underlying mechanisms causing pain in preclinical models, and in clinical populations. There are also important disconnects between clinical pain populations and the way preclinical pain studies are conducted. For instance, osteoarthritis pain more frequently affects women, but most preclinical studies have been conducted using males in animal models. The most widely used painkillers, nonsteroidal anti-inflammatory drugs (NSAIDs), act on the prostaglandin pathway by inhibiting cyclooxygenase (COX) enzymes. The purpose of this study was to analyze the preclinical and clinical literature on the role of prostaglandins and COX in inflammation and pain. We aimed to specifically identify studies that used both sexes and investigate whether any sex-differences in the action of prostaglandins and COX inhibition had been reported, either in clinical or preclinical studies. We conducted a PubMed search and identified 369 preclinical studies and 100 clinical studies that matched our inclusion/exclusion criteria. Our analysis shows that only 17% of preclinical studies on prostaglandins used both sexes and, out of those, only 19% analyzed or reported data separated by sex. In contrast, 79% of the clinical studies analyzed used both sexes. However, only 6% of those reported data separated by sex. Interestingly, 14 out of 15 preclinical studies and 5 out of 6 clinical studies that analyzed data separated by sex have identified sex-differences. This builds on the increasing evidence of sex-differences in prostaglandin signaling and the importance of sex as a biological variable in data analysis. The preclinical literature identifies a sex difference in prostaglandin D2 synthase (PTGDS) expression where it is higher in female than in male rodents in the nervous system. We experimentally validated that PTGDS expression is higher in female human dorsal root ganglia (DRG) neurons recovered from organ donors. Our semi-systematic literature review reveals a need for continued inclusivity of both male and female animals in prostaglandins studies and data analysis separated by sex in preclinical and clinical studies. Our finding of sex-differences in neuronal PTGDS expression in humans exemplifies the need for a more comprehensive understanding of how the prostaglandin system functions in the DRG in rodents and humans.
Collapse
Affiliation(s)
- Breanna Q Shen
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX, 75080, USA
| | - Ishwarya Sankaranarayanan
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX, 75080, USA
| | - Theodore J Price
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX, 75080, USA.
| | - Diana Tavares-Ferreira
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX, 75080, USA.
| |
Collapse
|
30
|
Walters ET, Crook RJ, Neely GG, Price TJ, Smith ESJ. Persistent nociceptor hyperactivity as a painful evolutionary adaptation. Trends Neurosci 2023; 46:211-227. [PMID: 36610893 PMCID: PMC9974896 DOI: 10.1016/j.tins.2022.12.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 12/05/2022] [Accepted: 12/16/2022] [Indexed: 01/07/2023]
Abstract
Chronic pain caused by injury or disease of the nervous system (neuropathic pain) has been linked to persistent electrical hyperactivity of the sensory neurons (nociceptors) specialized to detect damaging stimuli and/or inflammation. This pain and hyperactivity are considered maladaptive because both can persist long after injured tissues have healed and inflammation has resolved. While the assumption of maladaptiveness is appropriate in many diseases, accumulating evidence from diverse species, including humans, challenges the assumption that neuropathic pain and persistent nociceptor hyperactivity are always maladaptive. We review studies indicating that persistent nociceptor hyperactivity has undergone evolutionary selection in widespread, albeit selected, animal groups as a physiological response that can increase survival long after bodily injury, using both highly conserved and divergent underlying mechanisms.
Collapse
Affiliation(s)
- Edgar T Walters
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA.
| | - Robyn J Crook
- Department of Biology, San Francisco State University, San Francisco, CA 94132, USA
| | - G Gregory Neely
- Charles Perkins Centre and School of Life and Environmental Sciences, The University of Sydney, NSW 2006, Australia
| | - Theodore J Price
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Ewan St John Smith
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, UK
| |
Collapse
|
31
|
Hough A, Criswell C, Faruk A, Cavanaugh JE, Kolber BJ, Tidgewell KJ. Barbamide Displays Affinity for Membrane-Bound Receptors and Impacts Store-Operated Calcium Entry in Mouse Sensory Neurons. Mar Drugs 2023; 21:110. [PMID: 36827151 PMCID: PMC9966578 DOI: 10.3390/md21020110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/25/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
Marine cyanobacteria are a rich source of bio-active metabolites that have been utilized as leads for drug discovery and pharmacological tools for basic science research. Here, we describe the re-isolation of a well-known metabolite, barbamide, from Curaçao on three different occasions and the characterization of barbamide's biological interactions with targets of the mammalian nervous system. Barbamide was originally discovered as a molluscicidal agent from a filamentous marine cyanobacterium. In our hands, we found little evidence of toxicity against mammalian cell cultures. However, barbamide showed several affinities when screened for binding affinity for a panel of 45 receptors and transporters known to be involved in nociception and sensory neuron activity. We found high levels of binding affinity for the dopamine transporter, the kappa opioid receptor, and the sigma receptors (sigma-1 and sigma-2 also known as transmembrane protein 97; TMEM97). We tested barbamide in vitro in isolated sensory neurons from female mice to explore its functional impact on calcium flux in these cells. Barbamide by itself had no observable impact on calcium flux. However, barbamide enhanced the effect of the TRPV1 agonist capsaicin and enhanced store-operated calcium entry (SOCE) responses after depletion of intracellular calcium. Overall, these results demonstrate the biological potential of barbamide at sensory neurons with implications for future drug development projects surrounding this molecule.
Collapse
Affiliation(s)
- Andrea Hough
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA 15282, USA
| | - Connor Criswell
- Center for Advanced Pain Studies, Department of Neuroscience, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Asef Faruk
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA 15282, USA
| | - Jane E. Cavanaugh
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA 15282, USA
| | - Benedict J. Kolber
- Center for Advanced Pain Studies, Department of Neuroscience, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Kevin J. Tidgewell
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA 15282, USA
| |
Collapse
|
32
|
Allison RL, Burand A, Torres DN, Brandow AM, Stucky CL, Ebert AD. Sickle cell disease patient plasma sensitizes iPSC-derived sensory neurons from sickle cell disease patients. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.10.523446. [PMID: 36711992 PMCID: PMC9882050 DOI: 10.1101/2023.01.10.523446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Individuals living with sickle cell disease (SCD) experience severe recurrent acute and chronic pain. In order to develop novel therapies, it is necessary to better understand the neurobiological mechanisms underlying SCD pain. There are many barriers to gaining mechanistic insight into pathogenic SCD pain processes, such as differential gene expression and function of sensory neurons between humans and mice with SCD, as well as the limited availability of patient samples. These can be overcome by utilizing SCD patient-derived induced pluripotent stem cells (iPSCs) differentiated into sensory neurons (SCD iSNs). Here, we characterize the key gene expression and function of SCD iSNs to establish a model for higher-throughput investigation of intrinsic and extrinsic factors that may contribute to increased SCD patient pain. Importantly, identified roles for C-C Motif Chemokine Ligand 2 (CCL2) and endothelin 1 (ET1) in SCD pain can be recapitulated in SCD iSNs. Further, we find that plasma taken from SCD patients during acute pain increases SCD iSN calcium response to the nociceptive stimulus capsaicin compared to those treated with paired SCD patient plasma at baseline or healthy control plasma samples. Together, these data provide the framework necessary to utilize iSNs as a powerful tool to investigate the neurobiology of SCD and identify potential intrinsic mechanisms of SCD pain which may extend beyond a blood-based pathology.
Collapse
Affiliation(s)
- Reilly L. Allison
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI
| | - Anthony Burand
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI
| | - Damaris Nieves Torres
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI
| | - Amanda M. Brandow
- Department of Pediatrics, Section of Hematology/Oncology/Bone Marrow Transplantation, Medical College of Wisconsin, Milwaukee, WI
| | - Cheryl L. Stucky
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI
| | - Allison D. Ebert
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI
| |
Collapse
|
33
|
Shiers S, Sahn JJ, Price TJ. MNK1 and MNK2 expression in the human dorsal root and trigeminal ganglion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.04.522773. [PMID: 36711529 PMCID: PMC9881964 DOI: 10.1101/2023.01.04.522773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Mitogen activated protein kinase interacting kinases (MNK) 1 and 2 are serine/threonine protein kinases that play an important role in translation of mRNAs through their phosphorylation of the RNA 5’-cap binding protein, eukaryotic translation initiation factor (eIF) 4E. These kinases are downstream targets for mitogen activated protein kinases (MAPKs), extracellular activity regulated protein kinase (ERK) and p38. MNKs have been implicated in the sensitization of peripheral nociceptors of the dorsal root and trigeminal ganglion (DRG and TG) using transgenic mouse lines and through the use of specific inhibitors of MNK1 and MNK2. While specific knockout of the Mknk1 gene suggests that it is the key isoform for regulation of nociceptor excitability and nociceptive behaviors in mice, both MKNK1 and MKNK2 genes are expressed in the DRG and TG of mice and humans based on RNA sequencing experiments. Single cell sequencing in mice suggests that Mknk1 and Mknk2 may be expressed in different populations of nociceptors. We sought to characterize mRNA expression in human DRG and TG for both MNK1 and MNK2. Our results show that both genes are expressed by nearly all neurons in both human ganglia with expression in other cell types as well. Our findings provide evidence that MNK1 and MNK2 are expressed by human nociceptors and suggest that efforts to pharmacologically target MNKs for pain would likely be translatable due its conserved expression in both species.
Collapse
Affiliation(s)
- Stephanie Shiers
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas, USA
| | | | - Theodore J. Price
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas, USA
| |
Collapse
|
34
|
Uhelski ML, Johns ME, Horrmann A, Mohamed S, Sohail A, Khasabova IA, Simone DA, Banik RK. Adverse effects of methylene blue in peripheral neurons: An in vitro electrophysiology and cell culture study. Mol Pain 2022; 18:17448069221142523. [PMID: 36408567 PMCID: PMC9730009 DOI: 10.1177/17448069221142523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Methylene blue (MB) is an effective treatment for methemoglobinemia, ifosfamide-induced encephalopathy, cyanide poisoning, and refractory vasoplegia. However, clinical case reports and preclinical studies indicate potentially neurotoxic activity of MB at certain concentrations. The exact mechanisms of MB neurotoxicity are not known, and while the effects of MB on neuronal tissue from different brain regions and myenteric ganglia have been examined, its effects on primary afferent neurons from dorsal root ganglia (DRG) have not been studied. Mouse DRG were exposed to MB (0.3-10 μM) in vitro to assess neurite outgrowth. Increasing concentrations of MB (0.3-10 μM) were associated with neurotoxicity as shown by a substantial loss of cells with neurite formation, particularly at 10 μM. In parallel experiments, cultured rat DRG neurons were treated with MB (100 μM) to examine how MB affects electrical membrane properties of small-diameter sensory neurons. MB decreased peak inward and outward current densities, decreased action potential amplitude, overshoot, afterhyperpolarization, increased action potential rise time, and decreased action potential firing in response to current stimulation. MB induced dose-dependent toxicity in peripheral neurons, in vitro. These findings are consistent with studies in brain and myenteric ganglion neurons showing increased neuronal loss and altered membrane electrical properties after MB application. Further research is needed to parse out the toxicity profile for MB to minimize damage to neuronal structures and reduce side effects in clinical settings.
Collapse
Affiliation(s)
- Megan L Uhelski
- Department of Pain Medicine, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Malcolm E Johns
- Department of Anesthesiology, School of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Alec Horrmann
- Department of Anesthesiology, School of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Sadiq Mohamed
- Department of Anesthesiology, School of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Ayesha Sohail
- Department of Anesthesiology, School of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Iryna A Khasabova
- Department of Diagnostic and Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN, USA
| | - Donald A Simone
- Department of Diagnostic and Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN, USA
| | - Ratan K Banik
- Department of Anesthesiology, School of Medicine, University of Minnesota, Minneapolis, MN, USA,Ratan K Banik, Department of Anesthesiology, University of Minnesota, B515 Mayo Memorial Building, 420 Delaware Street S.E., MMC 294, Minneapolis, MN 55455, USA.
| |
Collapse
|
35
|
Dvorakova M, Wilson S, Corey W, Billingsley J, Zimmowitch A, Tracey J, Straiker A, Mackie K. A Critical Evaluation of Terpenoid Signaling at Cannabinoid CB1 Receptors in a Neuronal Model. Molecules 2022; 27:molecules27175655. [PMID: 36080421 PMCID: PMC9457791 DOI: 10.3390/molecules27175655] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 08/30/2022] [Accepted: 08/31/2022] [Indexed: 11/29/2022] Open
Abstract
In addition to phytocannabinoids, cannabis contains terpenoids that are claimed to have a myriad of effects on the body. We tested a panel of five common cannabis terpenoids, myrcene, linalool, limonene, α-pinene and nerolidol, in two neuronal models, autaptic hippocampal neurons and dorsal root ganglion (DRG) neurons. Autaptic neurons express a form of cannabinoid CB1 receptor-dependent retrograde plasticity while DRGs express a variety of transient receptor potential (TRP) channels. Most terpenoids had little or no effect on neuronal cannabinoid signaling. The exception was nerolidol, which inhibited endocannabinoid signaling. Notably, this is not via inhibition of CB1 receptors but by inhibiting some aspect of 2-arachidonoylglycerol (2-AG) production/delivery; the mechanism does not involve reducing the activity of the 2-AG-synthesizing diacylglycerol lipases (DAGLs). Nerolidol was also the only terpenoid that activated a sustained calcium response in a small (7%) subpopulation of DRGs. In summary, we found that only one of five terpenoids tested had notable effects on cannabinoid signaling in two neuronal models. Our results suggest that a few terpenoids may indeed interact with some components of the cannabinoid signaling system and may therefore offer interesting insights upon further study.
Collapse
|
36
|
Zeidler M, Kummer KK, Kress M. Towards bridging the translational gap by improved modeling of human nociception in health and disease. Pflugers Arch 2022; 474:965-978. [PMID: 35655042 PMCID: PMC9393146 DOI: 10.1007/s00424-022-02707-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 05/18/2022] [Indexed: 11/09/2022]
Abstract
Despite numerous studies which have explored the pathogenesis of pain disorders in preclinical models, there is a pronounced translational gap, which is at least partially caused by differences between the human and rodent nociceptive system. An elegant way to bridge this divide is the exploitation of human-induced pluripotent stem cell (iPSC) reprogramming into human iPSC-derived nociceptors (iDNs). Several protocols were developed and optimized to model nociceptive processes in health and disease. Here we provide an overview of the different approaches and summarize the knowledge obtained from such models on pain pathologies associated with monogenetic sensory disorders so far. In addition, novel perspectives offered by increasing the complexity of the model systems further to better reflect the natural environment of nociceptive neurons by involving other cell types in 3D model systems are described.
Collapse
Affiliation(s)
- Maximilian Zeidler
- Institute of Physiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Kai K Kummer
- Institute of Physiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Michaela Kress
- Institute of Physiology, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
37
|
Chase R, de la Peña JB, Smith PR, Lawson J, Lou TF, Stanowick AD, Black BJ, Campbell ZT. Global analyses of mRNA expression in human sensory neurons reveal eIF5A as a conserved target for inflammatory pain. FASEB J 2022; 36:e22422. [PMID: 35747924 DOI: 10.1096/fj.202101933rr] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 05/28/2022] [Accepted: 06/08/2022] [Indexed: 11/11/2022]
Abstract
Nociceptors are a type of sensory neuron that are integral to most forms of pain. Targeted disruption of nociceptor sensitization affords unique opportunities to prevent pain. An emerging model for nociceptors are sensory neurons derived from human stem cells. Here, we subjected five groups to high-throughput sequencing: human induced pluripotent stem cells (hiPSCs) prior to differentiation, mature hiPSC-derived sensory neurons, mature co-cultures containing hiPSC-derived astrocytes and sensory neurons, mouse dorsal root ganglion (DRG) tissues, and mouse DRG cultures. Co-culture of nociceptors and astrocytes promotes expression of transcripts enriched in DRG tissues. Comparisons of the hiPSC models to tissue samples reveal that many key transcripts linked to pain are present. Markers indicative of a range of neuronal subtypes present in the DRG were detected in mature hiPSCs. Intriguingly, translation factors were maintained at consistently high expression levels across species and culture systems. As a proof of concept for the utility of this resource, we validated expression of eukaryotic initiation factor 5A (eIF5A) in DRG tissues and hiPSC samples. eIF5A is subject to a unique posttranslational hypusine modification required for its activity. Inhibition of hypusine biosynthesis prevented hyperalgesic priming by inflammatory mediators in vivo and diminished hiPSC activity in vitro. Collectively, our results illuminate the transcriptomes of hiPSC sensory neuron models. We provide a demonstration for this resource through our investigation of eIF5A. Our findings reveal hypusine as a potential target for inflammation associated pain in males.
Collapse
Affiliation(s)
- Rebecca Chase
- Department of Biological Sciences, University of Texas at Dallas, Richardson, Texas, USA
| | - June Bryan de la Peña
- Department of Biological Sciences, University of Texas at Dallas, Richardson, Texas, USA
| | - Patrick R Smith
- Department of Biological Sciences, University of Texas at Dallas, Richardson, Texas, USA
| | - Jennifer Lawson
- Department of Biomedical Engineering, University of Massachusetts Lowell, Lowell, Massachusetts, USA
| | - Tzu-Fang Lou
- Department of Biological Sciences, University of Texas at Dallas, Richardson, Texas, USA
| | - Alexander D Stanowick
- Department of Biological Sciences, University of Texas at Dallas, Richardson, Texas, USA
| | - Bryan J Black
- Department of Biomedical Engineering, University of Massachusetts Lowell, Lowell, Massachusetts, USA
| | - Zachary T Campbell
- Department of Biological Sciences, University of Texas at Dallas, Richardson, Texas, USA.,Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, Texas, USA
| |
Collapse
|
38
|
Barker KH, Higham JP, Pattison LA, Taylor TS, Chessell IP, Welsh F, Smith ESJ, Bulmer DC. Sensitisation of colonic nociceptors by TNFα is dependent on TNFR1 expression and p38 MAPK activity. J Physiol 2022; 600:3819-3836. [PMID: 35775903 PMCID: PMC9543404 DOI: 10.1113/jp283170] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 06/28/2022] [Indexed: 11/23/2022] Open
Abstract
Abstract Visceral pain is a leading cause of morbidity in gastrointestinal diseases, which is exacerbated by the gut‐related side‐effects of many analgesics. New treatments are needed and further understanding of the mediators and mechanisms underpinning visceral nociception in disease states is required to facilitate this. The pro‐inflammatory cytokine TNFα is linked to pain in both patients with inflammatory bowel disease and irritable bowel syndrome, and has been shown to sensitize colonic sensory neurons. Somatic, TNFα‐triggered thermal and mechanical hypersensitivity is mediated by TRPV1 signalling and p38 MAPK activity respectively, downstream of TNFR1 receptor activation. We therefore hypothesized that TNFR1‐evoked p38 MAPK activity may also be responsible for TNFα sensitization of colonic afferent responses to the TRPV1 agonist capsaicin, and noxious distension of the bowel. Using Ca2+ imaging of dorsal root ganglion sensory neurons, we observed TNFα‐mediated increases in intracellular [Ca2+] and sensitization of capsaicin responses. The sensitizing effects of TNFα were dependent on TNFR1 expression and attenuated by p38 MAPK inhibition. Consistent with these findings, ex vivo colonic afferent fibre recordings demonstrated an enhanced response to noxious ramp distention of the bowel and bath application of capsaicin following TNFα pre‐treatment. Responses were reversed by p38 MAPK inhibition and absent in tissue from TNFR1 knockout mice. Our findings demonstrate a contribution of TNFR1, p38 MAPK and TRPV1 to TNFα‐induced sensitization of colonic afferents, highlighting the potential utility of these drug targets for the treatment of visceral pain in gastrointestinal disease.
![]() Key points The pro‐inflammatory cytokine TNFα is elevated in gastrointestinal disease and sensitizes colonic afferents via modulation of TRPA1 and NaV1.8 activity. We further develop this understanding by demonstrating a role for p38 MAPK and TRPV1 in TNFα‐mediated colonic afferent sensitization. Specifically, we show that: TNFα sensitizes sensory neurons and colonic afferents to the TRPV1 agonist capsaicin. TNFα‐mediated sensitization of sensory neurons and colonic nociceptors is dependent on TNFR1 expression. TNFα sensitization of sensory neurons and colonic afferents to capsaicin and noxious ramp distension is abolished by inhibition of p38 MAPK. Collectively these data support the utility of targeting TNFα, TNFR1 and their downstream signalling via p38 MAPK for the treatment of visceral pain in gastrointestinal disease.
Collapse
Affiliation(s)
- Katie H Barker
- Department of Pharmacology, University of Cambridge, Cambridge, CB2 1PD, UK
| | - James P Higham
- Department of Pharmacology, University of Cambridge, Cambridge, CB2 1PD, UK
| | - Luke A Pattison
- Department of Pharmacology, University of Cambridge, Cambridge, CB2 1PD, UK
| | - Toni S Taylor
- Department of Pharmacology, University of Cambridge, Cambridge, CB2 1PD, UK
| | - Iain P Chessell
- Neuroscience, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Fraser Welsh
- Neuroscience, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Ewan St J Smith
- Department of Pharmacology, University of Cambridge, Cambridge, CB2 1PD, UK
| | - David C Bulmer
- Department of Pharmacology, University of Cambridge, Cambridge, CB2 1PD, UK
| |
Collapse
|
39
|
Inclan-Rico JM, Rossi HL, Herbert DR. "Every cell is an immune cell; contributions of non-hematopoietic cells to anti-helminth immunity". Mucosal Immunol 2022; 15:1199-1211. [PMID: 35538230 PMCID: PMC9646929 DOI: 10.1038/s41385-022-00518-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/04/2022] [Accepted: 04/19/2022] [Indexed: 02/04/2023]
Abstract
Helminths are remarkably successful parasites that can invade various mammalian hosts and establish chronic infections that can go unnoticed for years despite causing severe tissue damage. To complete their life cycles, helminths migrate through multiple barrier sites that are densely populated by a complex array of hematopoietic and non-hematopoietic cells. While it is clear that type 2 cytokine responses elicited by immune cells promote worm clearance and tissue healing, the actions of non-hematopoietic cells are increasingly recognized as initiators, effectors and regulators of anti-helminth immunity. This review will highlight the collective actions of specialized epithelial cells, stromal niches, stem, muscle and neuroendocrine cells as well as peripheral neurons in the detection and elimination of helminths at mucosal sites. Studies dissecting the interactions between immune and non-hematopoietic cells will truly provide a better understanding of the mechanisms that ensure homeostasis in the context of helminth infections.
Collapse
Affiliation(s)
- Juan M Inclan-Rico
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Heather L Rossi
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - De'Broski R Herbert
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
40
|
Behrendt M, Solinski HJ, Schmelz M, Carr R. Bradykinin-Induced Sensitization of Transient Receptor Potential Channel Melastatin 3 Calcium Responses in Mouse Nociceptive Neurons. Front Cell Neurosci 2022; 16:843225. [PMID: 35496916 PMCID: PMC9043526 DOI: 10.3389/fncel.2022.843225] [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: 12/25/2021] [Accepted: 03/18/2022] [Indexed: 11/13/2022] Open
Abstract
TRPM3 is a calcium-permeable cation channel expressed in a range of sensory neurons that can be activated by heat and the endogenous steroid pregnenolone sulfate (PS). During inflammation, the expression and function of TRPM3 are both augmented in somatosensory nociceptors. However, in isolated dorsal root ganglion (DRG) neurons application of inflammatory mediators like prostaglandins and bradykinin (BK) inhibit TRPM3. Therefore, the aim of this study was to examine the effect of preceding activation of cultured 1 day old mouse DRG neurons by the inflammatory mediator BK on TRPM3-mediated calcium responses. Calcium signals were recorded using the intensity-based dye Fluo-8. We found that TRPM3-mediated calcium responses to PS were enhanced by preceding application of BK in cells that responded to BK with a calcium signal, indicating BK receptor (BKR) expression. The majority of cells that co-expressed TRPM3 and BKRs also expressed TRPV1, however, only a small fraction co-expressed TRPA1, identified by calcium responses to capsaicin and supercinnamaldehyde, respectively. Signaling and trafficking pathways responsible for sensitization of TRPM3 following BK were characterized using inhibitors of second messenger signaling cascades and exocytosis. Pharmacological blockade of protein kinase C, calcium–calmodulin-dependent protein kinase II and diacylglycerol (DAG) lipase did not affect BK-induced sensitization, but inhibition of DAG kinase did. In addition, release of calcium from intracellular stores using thapsigargin also resulted in TRPM3 sensitization. Finally, BK did not sensitize TRPM3 in the presence of exocytosis inhibitors. Collectively, we show that preceding activation of DRG neurons by BK sensitized TRPM3-mediated calcium responses to PS. Our results indicate that BKR-mediated activation of intracellular signaling pathways comprising DAG kinase, calcium and exocytosis may contribute to TRPM3 sensitization during inflammation.
Collapse
|
41
|
Hall BE, Macdonald E, Cassidy M, Yun S, Sapio MR, Ray P, Doty M, Nara P, Burton MD, Shiers S, Ray-Chaudhury A, Mannes AJ, Price TJ, Iadarola MJ, Kulkarni AB. Transcriptomic analysis of human sensory neurons in painful diabetic neuropathy reveals inflammation and neuronal loss. Sci Rep 2022; 12:4729. [PMID: 35304484 PMCID: PMC8933403 DOI: 10.1038/s41598-022-08100-8] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 02/25/2022] [Indexed: 01/02/2023] Open
Abstract
Pathological sensations caused by peripheral painful neuropathy occurring in Type 2 diabetes mellitus (T2DM) are often described as 'sharp' and 'burning' and are commonly spontaneous in origin. Proposed etiologies implicate dysfunction of nociceptive sensory neurons in dorsal root ganglia (DRG) induced by generation of reactive oxygen species, microvascular defects, and ongoing axonal degeneration and regeneration. To investigate the molecular mechanisms contributing to diabetic pain, DRGs were acquired postmortem from patients who had been experiencing painful diabetic peripheral neuropathy (DPN) and subjected to transcriptome analyses to identify genes contributing to pathological processes and neuropathic pain. DPN occurs in distal extremities resulting in the characteristic "glove and stocking" pattern. Accordingly, the L4 and L5 DRGs, which contain the perikarya of primary afferent neurons innervating the foot, were analyzed from five DPN patients and compared with seven controls. Transcriptome analyses identified 844 differentially expressed genes. We observed increases in levels of inflammation-associated transcripts from macrophages in DPN patients that may contribute to pain hypersensitivity and, conversely, there were frequent decreases in neuronally-related genes. The elevated inflammatory gene profile and the accompanying downregulation of multiple neuronal genes provide new insights into intraganglionic pathology and mechanisms causing neuropathic pain in DPN patients with T2DM.
Collapse
Affiliation(s)
- Bradford E Hall
- Functional Genomics Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Room 130, Bethesda, MD, 20892, USA
| | - Emma Macdonald
- Functional Genomics Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Room 130, Bethesda, MD, 20892, USA
- Present Affiliation: NIH Graduate Partnerships Program, Brown University, Providence, RI, 02912, USA
| | - Margaret Cassidy
- Functional Genomics Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Room 130, Bethesda, MD, 20892, USA
| | - Sijung Yun
- Yotta Biomed, LLC, Bethesda, MD, 20814, USA
| | - Matthew R Sapio
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Pradipta Ray
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Megan Doty
- Functional Genomics Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Room 130, Bethesda, MD, 20892, USA
| | - Pranavi Nara
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Michael D Burton
- Neuroimmunology and Behavior Group, School of Behavior and Brain Sciences, 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
| | - Abhik Ray-Chaudhury
- Surgical Neurology Branch, Disorders and Stroke, National Institute of Neurological, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Andrew J Mannes
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Theodore J Price
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Michael J Iadarola
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Ashok B Kulkarni
- Functional Genomics Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Room 130, Bethesda, MD, 20892, USA.
| |
Collapse
|
42
|
Tavares-Ferreira D, Shiers S, Ray PR, Wangzhou A, Jeevakumar V, Sankaranarayanan I, Cervantes AM, Reese JC, Chamessian A, Copits BA, Dougherty PM, Gereau RW, Burton MD, Dussor G, Price TJ. Spatial transcriptomics of dorsal root ganglia identifies molecular signatures of human nociceptors. Sci Transl Med 2022; 14:eabj8186. [PMID: 35171654 PMCID: PMC9272153 DOI: 10.1126/scitranslmed.abj8186] [Citation(s) in RCA: 225] [Impact Index Per Article: 75.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Nociceptors are specialized sensory neurons that detect damaging or potentially damaging stimuli and are found in the dorsal root ganglia (DRG) and trigeminal ganglia. These neurons are critical for the generation of neuronal signals that ultimately create the perception of pain. Nociceptors are also primary targets for treating acute and chronic pain. Single-cell transcriptomics on mouse nociceptors has transformed our understanding of pain mechanisms. We sought to generate equivalent information for human nociceptors with the goal of identifying transcriptomic signatures of nociceptors, identifying species differences and potential drug targets. We used spatial transcriptomics to molecularly characterize transcriptomes of single DRG neurons from eight organ donors. We identified 12 clusters of human sensory neurons, 5 of which are C nociceptors, as well as 1 C low-threshold mechanoreceptors (LTMRs), 1 Aβ nociceptor, 2 Aδ, 2 Aβ, and 1 proprioceptor subtypes. By focusing on expression profiles for ion channels, G protein-coupled receptors (GPCRs), and other pharmacological targets, we provided a rich map of potential drug targets in the human DRG with direct comparison to mouse sensory neuron transcriptomes. We also compared human DRG neuronal subtypes to nonhuman primates showing conserved patterns of gene expression among many cell types but divergence among specific nociceptor subsets. Last, we identified sex differences in human DRG subpopulation transcriptomes, including a marked increase in calcitonin-related polypeptide alpha (CALCA) expression in female pruritogen receptor-enriched nociceptors. This comprehensive spatial characterization of human nociceptors might open the door to development of better treatments for acute and chronic pain disorders.
Collapse
Affiliation(s)
- Diana Tavares-Ferreira
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson TX 75080, USA.,Corresponding author: (T.J.P.); (D.T.-F.)
| | - Stephanie Shiers
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson TX 75080, USA
| | - Pradipta R. Ray
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson TX 75080, USA
| | - Andi Wangzhou
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson TX 75080, USA
| | - Vivekanand Jeevakumar
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson TX 75080, USA
| | - Ishwarya Sankaranarayanan
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson TX 75080, USA
| | | | | | - Alexander Chamessian
- Department of Anesthesiology, Washington University Pain Center, St. Louis, MO 63110, USA
| | - Bryan A. Copits
- Department of Anesthesiology, Washington University Pain Center, St. Louis, MO 63110, USA
| | - Patrick M. Dougherty
- Department of Pain Medicine, Division of Anesthesiology and Critical Care, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Robert W. Gereau
- Department of Anesthesiology, Washington University Pain Center, St. Louis, MO 63110, USA
| | - Michael D. Burton
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson TX 75080, USA
| | - Gregory Dussor
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson TX 75080, USA
| | - Theodore J. Price
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson TX 75080, USA.,Corresponding author: (T.J.P.); (D.T.-F.)
| |
Collapse
|
43
|
Tavares-Ferreira D, Shiers S, Ray PR, Wangzhou A, Jeevakumar V, Sankaranarayanan I, Cervantes AM, Reese JC, Chamessian A, Copits BA, Dougherty PM, Gereau RW, Burton MD, Dussor G, Price TJ. Spatial transcriptomics of dorsal root ganglia identifies molecular signatures of human nociceptors. Sci Transl Med 2022. [DOI: 10.1126/scitranslmed.abj8186\] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Nociceptors are specialized sensory neurons that detect damaging or potentially damaging stimuli and are found in the dorsal root ganglia (DRG) and trigeminal ganglia. These neurons are critical for the generation of neuronal signals that ultimately create the perception of pain. Nociceptors are also primary targets for treating acute and chronic pain. Single-cell transcriptomics on mouse nociceptors has transformed our understanding of pain mechanisms. We sought to generate equivalent information for human nociceptors with the goal of identifying transcriptomic signatures of nociceptors, identifying species differences and potential drug targets. We used spatial transcriptomics to molecularly characterize transcriptomes of single DRG neurons from eight organ donors. We identified 12 clusters of human sensory neurons, 5 of which are C nociceptors, as well as 1 C low-threshold mechanoreceptors (LTMRs), 1 Aβ nociceptor, 2 Aδ, 2 Aβ, and 1 proprioceptor subtypes. By focusing on expression profiles for ion channels, G protein–coupled receptors (GPCRs), and other pharmacological targets, we provided a rich map of potential drug targets in the human DRG with direct comparison to mouse sensory neuron transcriptomes. We also compared human DRG neuronal subtypes to nonhuman primates showing conserved patterns of gene expression among many cell types but divergence among specific nociceptor subsets. Last, we identified sex differences in human DRG subpopulation transcriptomes, including a marked increase in calcitonin-related polypeptide alpha (
CALCA
) expression in female pruritogen receptor–enriched nociceptors. This comprehensive spatial characterization of human nociceptors might open the door to development of better treatments for acute and chronic pain disorders.
Collapse
Affiliation(s)
- Diana Tavares-Ferreira
- 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
| | - Pradipta R. Ray
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson TX 75080, USA
| | - Andi Wangzhou
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson TX 75080, USA
| | - Vivekanand Jeevakumar
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson TX 75080, USA
| | - Ishwarya Sankaranarayanan
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson TX 75080, USA
| | | | | | - Alexander Chamessian
- Department of Anesthesiology , Washington University Pain Center, St. Louis, MO 63110, USA
| | - Bryan A. Copits
- Department of Anesthesiology , Washington University Pain Center, St. Louis, MO 63110, USA
| | - Patrick M. Dougherty
- Department of Pain Medicine, Division of Anesthesiology and Critical Care, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Robert W. Gereau
- Department of Anesthesiology , Washington University Pain Center, St. Louis, MO 63110, USA
| | - Michael D. Burton
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson TX 75080, USA
| | - Gregory Dussor
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson TX 75080, USA
| | - Theodore J. Price
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson TX 75080, USA
| |
Collapse
|
44
|
Slow touch in non-human species: translational research into the C-tactile (CT) afferent system. Curr Opin Behav Sci 2022. [DOI: 10.1016/j.cobeha.2021.10.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
45
|
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
|
46
|
Skin-resident dendritic cells mediate postoperative pain via CCR4 on sensory neurons. Proc Natl Acad Sci U S A 2022; 119:2118238119. [PMID: 35046040 PMCID: PMC8794894 DOI: 10.1073/pnas.2118238119] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/01/2021] [Indexed: 01/08/2023] Open
Abstract
Interactions between the nervous and immune systems control the generation and maintenance of inflammatory pain. However, the immune cells and mediators controlling this response remain poorly characterized. We identified the cytokines CCL22 and CCL17 as secreted mediators that act directly on sensory neurons to mediate postoperative pain via their shared receptor, CCR4. We also show that skin-resident dendritic cells are key contributors to the inflammatory pain response. Blocking the interaction between these dendritic cell–derived ligands and their receptor can abrogate the pain response, highlighting CCR4 antagonists as potentially effective therapies for postoperative pain. Our findings identify functions for these tissue-resident myeloid cells and uncover mechanisms underlying pain pathophysiology. Inflammatory pain, such as hypersensitivity resulting from surgical tissue injury, occurs as a result of interactions between the immune and nervous systems with the orchestrated recruitment and activation of tissue-resident and circulating immune cells to the site of injury. Our previous studies identified a central role for Ly6Clow myeloid cells in the pathogenesis of postoperative pain. We now show that the chemokines CCL17 and CCL22, with their cognate receptor CCR4, are key mediators of this response. Both chemokines are up-regulated early after tissue injury by skin-resident dendritic and Langerhans cells to act on peripheral sensory neurons that express CCR4. CCL22, and to a lesser extent CCL17, elicit acute mechanical and thermal hypersensitivity when administered subcutaneously; this response abrogated by pharmacological blockade or genetic silencing of CCR4. Electrophysiological assessment of dissociated sensory neurons from naïve and postoperative mice showed that CCL22 was able to directly activate neurons and enhance their excitability after injury. These responses were blocked using C 021 and small interfering RNA (siRNA)-targeting CCR4. Finally, our data show that acute postoperative pain is significantly reduced in mice lacking CCR4, wild-type animals treated with CCR4 antagonist/siRNA, as well as transgenic mice depleted of dendritic cells. Together, these results suggest an essential role for the peripheral CCL17/22:CCR4 axis in the genesis of inflammatory pain via direct communication between skin-resident dendritic cells and sensory neurons, opening therapeutic avenues for its control.
Collapse
|
47
|
Yang CC, Hokanson JA, Keast JR. Advancing our understanding of the neural control of the female human urethra. Neurourol Urodyn 2022; 41:35-41. [PMID: 34605569 PMCID: PMC8738110 DOI: 10.1002/nau.24807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 09/17/2021] [Accepted: 09/20/2021] [Indexed: 01/03/2023]
Affiliation(s)
- Claire C Yang
- Department of Urology, University of Washington, Seattle, Washington, USA
| | - James A Hokanson
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Janet R Keast
- Department of Anatomy and Physiology, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
48
|
Tmem160 contributes to the establishment of discrete nerve injury-induced pain behaviors in male mice. Cell Rep 2021; 37:110152. [PMID: 34936870 DOI: 10.1016/j.celrep.2021.110152] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 09/01/2021] [Accepted: 11/30/2021] [Indexed: 12/26/2022] Open
Abstract
Chronic pain is a prevalent medical problem, and its molecular basis remains poorly understood. Here, we demonstrate the significance of the transmembrane protein (Tmem) 160 for nerve injury-induced neuropathic pain. An extensive behavioral assessment suggests a pain modality- and entity-specific phenotype in male Tmem160 global knockout (KO) mice: delayed establishment of tactile hypersensitivity and alterations in self-grooming after nerve injury. In contrast, Tmem160 seems to be dispensable for other nerve injury-induced pain modalities, such as non-evoked and movement-evoked pain, and for other pain entities. Mechanistically, we show that global KO males exhibit dampened neuroimmune signaling and diminished TRPA1-mediated activity in cultured dorsal root ganglia. Neither these changes nor altered pain-related behaviors are observed in global KO female and male peripheral sensory neuron-specific KO mice. Our findings reveal Tmem160 as a sexually dimorphic factor contributing to the establishment, but not maintenance, of discrete nerve injury-induced pain behaviors in male mice.
Collapse
|
49
|
Zeidler M, Kummer KK, Schöpf CL, Kalpachidou T, Kern G, Cader MZ, Kress M. NOCICEPTRA: Gene and microRNA Signatures and Their Trajectories Characterizing Human iPSC-Derived Nociceptor Maturation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2102354. [PMID: 34486248 PMCID: PMC8564443 DOI: 10.1002/advs.202102354] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Indexed: 05/07/2023]
Abstract
Nociceptors are primary afferent neurons serving the reception of acute pain but also the transit into maladaptive pain disorders. Since native human nociceptors are hardly available for mechanistic functional research, and rodent models do not necessarily mirror human pathologies in all aspects, human induced pluripotent stem cell-derived nociceptors (iDN) offer superior advantages as a human model system. Unbiased mRNA::microRNA co-sequencing, immunofluorescence staining, and qPCR validations, reveal expression trajectories as well as miRNA target spaces throughout the transition of pluripotent cells into iDNs. mRNA and miRNA candidates emerge as regulatory hubs for neurite outgrowth, synapse development, and ion channel expression. The exploratory data analysis tool NOCICEPTRA is provided as a containerized platform to retrieve experimentally determined expression trajectories, and to query custom gene sets for pathway and disease enrichments. Querying NOCICEPTRA for marker genes of cortical neurogenesis reveals distinct similarities and differences for cortical and peripheral neurons. The platform provides a public domain neuroresource to exploit the entire data sets and explore miRNA and mRNA as hubs regulating human nociceptor differentiation and function.
Collapse
Affiliation(s)
- Maximilian Zeidler
- Institute of PhysiologyMedical University of InnsbruckInnsbruck6020Austria
| | - Kai K. Kummer
- Institute of PhysiologyMedical University of InnsbruckInnsbruck6020Austria
| | - Clemens L. Schöpf
- Institute of PhysiologyMedical University of InnsbruckInnsbruck6020Austria
| | | | - Georg Kern
- Institute of PhysiologyMedical University of InnsbruckInnsbruck6020Austria
| | - M. Zameel Cader
- Weatherall Institute of Molecular MedicineUniversity of OxfordOxfordOX3 9DSUK
| | - Michaela Kress
- Institute of PhysiologyMedical University of InnsbruckInnsbruck6020Austria
| |
Collapse
|
50
|
In Vitro Model to Investigate Communication between Dorsal Root Ganglion and Spinal Cord Glia. Int J Mol Sci 2021; 22:ijms22189725. [PMID: 34575886 PMCID: PMC8470479 DOI: 10.3390/ijms22189725] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/02/2021] [Accepted: 09/03/2021] [Indexed: 12/30/2022] Open
Abstract
Chronic discogenic back pain is associated with increased inflammatory cytokine levels that can influence the proximal peripheral nervous system, namely the dorsal root ganglion (DRG). However, transition to chronic pain is widely thought to involve glial activation in the spinal cord. In this study, an in vitro model was used to evaluate the communication between DRG and spinal cord glia. Primary neonatal rat DRG cells were treated with/without inflammatory cytokines (TNF-α, IL-1β, and IL-6). The conditioned media were collected at two time points (12 and 24 h) and applied to spinal cord mixed glial culture (MGC) for 24 h. Adult bovine DRG and spinal cord cell cultures were also tested, as an alternative large animal model, and results were compared with the neonatal rat findings. Compared with untreated DRG-conditioned medium, the second cytokine-treated DRG-conditioned medium (following medium change, thus containing solely DRG-derived molecules) elevated CD11b expression and calcium signal in neonatal rat microglia and enhanced Iba1 expression in adult bovine microglia. Cytokine treatment induced a DRG-mediated microgliosis. The described in vitro model allows the use of cells from large species and may represent an alternative to animal pain models (3R principles).
Collapse
|