1
|
Price T, Shiers S, Mazhar K, Wangzhou A, Haberberger R, Lesnak J, Sankaranarayanan I, Tavares-Ferreira D, Cervantes A, Funk G, Horton P, Vines E, Dussor G. Nageotte nodules in human DRG reveal neurodegeneration in painful diabetic neuropathy. RESEARCH SQUARE 2024:rs.3.rs-5006011. [PMID: 39399674 PMCID: PMC11469377 DOI: 10.21203/rs.3.rs-5006011/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Diabetic neuropathy is frequently accompanied by pain and loss of sensation attributed to axonal dieback. We recovered dorsal root ganglia (DRGs) from 90 organ donors, 19 of whom had medical indices for diabetic painful neuropathy (DPN). Nageotte nodules, dead sensory neurons engulfed by non-neuronal cells, were abundant in DPN DRGs and accounted for 25% of all neurons. Peripherin-and Nav1.7-positive dystrophic axons invaded Nageotte nodules, forming small neuroma-like structures. Using histology and spatial sequencing, we demonstrate that Nageotte nodules are mainly composed of satellite glia and non-myelinating Schwann cells that express SPP1 and are intertwined with sprouting sensory axons originating from neighboring neurons. Our findings solve a 100-year mystery of the nature of Nageotte nodules linking these pathological structures to pain and sensory loss in DPN.
Collapse
|
2
|
Shiers SI, Mazhar K, Wangzhou A, Haberberger R, Lesnak JB, Sankaranarayanan I, Tavares-Ferreira D, Cervantes A, Funk G, Horton P, Vines E, Dussor G, Price TJ. Nageotte nodules in human DRG reveal neurodegeneration in painful diabetic neuropathy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.22.609215. [PMID: 39229145 PMCID: PMC11370606 DOI: 10.1101/2024.08.22.609215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Diabetic neuropathy is frequently accompanied by pain and loss of sensation attributed to axonal dieback. We recovered dorsal root ganglia (DRGs) from 90 organ donors, 19 of whom had medical indices for diabetic painful neuropathy (DPN). Nageotte nodules, dead sensory neurons engulfed by non-neuronal cells, were abundant in DPN DRGs and accounted for 25% of all neurons. Peripherin-and Nav1.7-positive dystrophic axons invaded Nageotte nodules, forming small neuroma-like structures. Using histology and spatial sequencing, we demonstrate that Nageotte nodules are mainly composed of satellite glia and non-myelinating Schwann cells that express SPP1 and are intertwined with sprouting sensory axons originating from neighboring neurons. Our findings solve a 100-year mystery of the nature of Nageotte nodules linking these pathological structures to pain and sensory loss in DPN.
Collapse
Affiliation(s)
- Stephanie I Shiers
- Department of Neuroscience, Center for Advanced Pain Studies, The University of Texas at Dallas. Richardson, TX
| | - Khadijah Mazhar
- Department of Neuroscience, Center for Advanced Pain Studies, The University of Texas at Dallas. Richardson, TX
| | - Andi Wangzhou
- Department of Neuroscience, Center for Advanced Pain Studies, The University of Texas at Dallas. Richardson, TX
| | | | - Joseph B Lesnak
- Department of Neuroscience, Center for Advanced Pain Studies, The University of Texas at Dallas. Richardson, TX
| | - Ishwarya Sankaranarayanan
- Department of Neuroscience, Center for Advanced Pain Studies, The University of Texas at Dallas. Richardson, TX
| | - Diana Tavares-Ferreira
- Department of Neuroscience, Center for Advanced Pain Studies, The University of Texas at Dallas. Richardson, TX
| | | | | | | | | | - Gregory Dussor
- Department of Neuroscience, Center for Advanced Pain Studies, The University of Texas at Dallas. Richardson, TX
| | - Theodore J Price
- Department of Neuroscience, Center for Advanced Pain Studies, The University of Texas at Dallas. Richardson, TX
| |
Collapse
|
3
|
Mitchell ME, Torrijos G, Cook LF, Mwirigi JM, He L, Shiers S, Price TJ. Interleukin-6 induces nascent protein synthesis in human dorsal root ganglion nociceptors primarily via MNK-eIF4E signaling. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2024; 16:100159. [PMID: 39156884 PMCID: PMC11327947 DOI: 10.1016/j.ynpai.2024.100159] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 06/26/2024] [Indexed: 08/20/2024]
Abstract
Plasticity of dorsal root ganglion (DRG) nociceptors in the peripheral nervous system requires new protein synthesis. This plasticity is believed to be responsible for the physiological changes seen in DRG nociceptors in animal models of chronic pain. Experiments in human DRG (hDRG) neurons also support this hypothesis, but a direct observation of nascent protein synthesis in response to a pain promoting substance, like interleukin-6 (IL-6), has not been measured in these neurons. To fill this gap in knowledge, we used acutely prepared human DRG explants from organ donors. These explants provide a physiologically relevant microenvironment, closer to in vivo conditions, allowing for the examination of functional alterations in DRG neurons reflective of human neuropathophysiology. Using this newly developed assay, we demonstrate upregulation of the target of the MNK1/2 kinases, phosphorylated eIF4E (p-eIF4E), and nascently synthesized proteins in a substantial subset of hDRG neurons following exposure to IL-6. To pinpoint the specific molecular mechanisms driving this IL-6-driven increase in nascent proteins, we used the specific MNK1/2 inhibitor eFT508. Treatment with eFT508 resulted in the inhibition of IL-6-induced increases in p-eIF4E and nascent proteins. Additionally, using TRPV1 as a marker for nociceptors, we found that these effects occurred in a large number of human nociceptors. Our findings provide clear evidence that IL-6 drives nascent protein synthesis in human TRPV1+ nociceptors primarily via MNK1/2-eIF4E signaling. The work links animal findings to human nociception, creates a framework for additional hDRG signaling experiments, and substantiates the continued development of MNK inhibitors for pain.
Collapse
Affiliation(s)
| | | | - Lauren F. Cook
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Juliet M. Mwirigi
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Lucy He
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Stephanie Shiers
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Theodore J. Price
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080, USA
| |
Collapse
|
4
|
Ferreira DT, Shen BQ, Mwirigi JM, Shiers S, Sankaranarayanan I, Kotamarti M, Inturi NN, Mazhar K, Ubogu EE, Thomas G, Lalli T, Wukich D, Price TJ. Deciphering the molecular landscape of human peripheral nerves: implications for diabetic peripheral neuropathy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.15.599167. [PMID: 38915676 PMCID: PMC11195245 DOI: 10.1101/2024.06.15.599167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Diabetic peripheral neuropathy (DPN) is a prevalent complication of diabetes mellitus that is caused by metabolic toxicity to peripheral axons. We aimed to gain deep mechanistic insight into the disease process using bulk and spatial RNA sequencing on tibial and sural nerves recovered from lower leg amputations in a mostly diabetic population. First, our approach comparing mixed sensory and motor tibial and purely sensory sural nerves shows key pathway differences in affected nerves, with distinct immunological features observed in sural nerves. Second, spatial transcriptomics analysis of sural nerves reveals substantial shifts in endothelial and immune cell types associated with severe axonal loss. We also find clear evidence of neuronal gene transcript changes, like PRPH, in nerves with axonal loss suggesting perturbed RNA transport into distal sensory axons. This motivated further investigation into neuronal mRNA localization in peripheral nerve axons generating clear evidence of robust localization of mRNAs such as SCN9A and TRPV1 in human sensory axons. Our work gives new insight into the altered cellular and transcriptomic profiles in human nerves in DPN and highlights the importance of sensory axon mRNA transport as an unappreciated potential contributor to peripheral nerve degeneration.
Collapse
Affiliation(s)
- Diana Tavares Ferreira
- Department of Neuroscience and Center for Advanced Pain Studies; University of Texas at Dallas, Richardson, TX, USA
| | - Breanna Q Shen
- Department of Neuroscience and Center for Advanced Pain Studies; University of Texas at Dallas, Richardson, TX, USA
| | - Juliet M Mwirigi
- Department of Neuroscience and Center for Advanced Pain Studies; University of Texas at Dallas, Richardson, TX, USA
| | - Stephanie Shiers
- Department of Neuroscience and Center for Advanced Pain Studies; University of Texas at Dallas, Richardson, TX, USA
| | - Ishwarya Sankaranarayanan
- Department of Neuroscience and Center for Advanced Pain Studies; University of Texas at Dallas, Richardson, TX, USA
| | - Miriam Kotamarti
- Department of Neuroscience and Center for Advanced Pain Studies; University of Texas at Dallas, Richardson, TX, USA
| | - Nikhil N Inturi
- Department of Neuroscience and Center for Advanced Pain Studies; University of Texas at Dallas, Richardson, TX, USA
| | - Khadijah Mazhar
- Department of Neuroscience and Center for Advanced Pain Studies; University of Texas at Dallas, Richardson, TX, USA
| | - Eroboghene E Ubogu
- Department of Neurology, Division of Neuromuscular Disease, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Geneva Thomas
- Department of Orthopedic Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Trapper Lalli
- Department of Orthopedic Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Dane Wukich
- Department of Orthopedic Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Theodore J Price
- Department of Neuroscience and Center for Advanced Pain Studies; University of Texas at Dallas, Richardson, TX, USA
| |
Collapse
|
5
|
Kalia AK, Rösseler C, Granja-Vazquez R, Ahmad A, Pancrazio JJ, Neureiter A, Zhang M, Sauter D, Vetter I, Andersson A, Dussor G, Price TJ, Kolber BJ, Truong V, Walsh P, Lampert A. How to differentiate induced pluripotent stem cells into sensory neurons for disease modelling: a functional assessment. Stem Cell Res Ther 2024; 15:99. [PMID: 38581069 PMCID: PMC10998320 DOI: 10.1186/s13287-024-03696-2] [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: 07/07/2023] [Accepted: 03/13/2024] [Indexed: 04/07/2024] Open
Abstract
BACKGROUND Human induced pluripotent stem cell (iPSC)-derived peripheral sensory neurons present a valuable tool to model human diseases and are a source for applications in drug discovery and regenerative medicine. Clinically, peripheral sensory neuropathies can result in maladies ranging from a complete loss of pain to severe painful neuropathic disorders. Sensory neurons are located in the dorsal root ganglion and are comprised of functionally diverse neuronal types. Low efficiency, reproducibility concerns, variations arising due to genetic factors and time needed to generate functionally mature neuronal populations from iPSCs remain key challenges to study human nociception in vitro. Here, we report a detailed functional characterization of iPSC-derived sensory neurons with an accelerated differentiation protocol ("Anatomic" protocol) compared to the most commonly used small molecule approach ("Chambers" protocol). Anatomic's commercially available RealDRG™ were further characterized for both functional and expression phenotyping of key nociceptor markers. METHODS Multiple iPSC clones derived from different reprogramming methods, genetics, age, and somatic cell sources were used to generate sensory neurons. Manual patch clamp was used to functionally characterize both control and patient-derived neurons. High throughput techniques were further used to demonstrate that RealDRGs™ derived from the Anatomic protocol are amenable to high throughput technologies for disease modelling. RESULTS The Anatomic protocol rendered a purer culture without the use of mitomycin C to suppress non-neuronal outgrowth, while Chambers differentiations yielded a mix of cell types. Chambers protocol results in predominantly tonic firing when compared to Anatomic protocol. Patient-derived nociceptors displayed higher frequency firing compared to control subject with both, Chambers and Anatomic differentiation approaches, underlining their potential use for clinical phenotyping as a disease-in-a-dish model. RealDRG™ sensory neurons show heterogeneity of nociceptive markers indicating that the cells may be useful as a humanized model system for translational studies. CONCLUSIONS We validated the efficiency of two differentiation protocols and their potential application for functional assessment and thus understanding the disease mechanisms from patients suffering from pain disorders. We propose that both differentiation methods can be further exploited for understanding mechanisms and development of novel treatments in pain disorders.
Collapse
Affiliation(s)
- Anil Kumar Kalia
- Institute of Neurophysiology, Uniklinik RWTH Aachen University, Pauwelsstr. 30, 52074, Aachen, Germany
- Research Training Group 2416 MultiSenses-MultiScales, RWTH Aachen University, Aachen, Germany
| | - Corinna Rösseler
- Institute of Neurophysiology, Uniklinik RWTH Aachen University, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Rafael Granja-Vazquez
- Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, 75080, USA
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Ayesha Ahmad
- Department of Neuroscience, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Joseph J Pancrazio
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Anika Neureiter
- Institute of Neurophysiology, Uniklinik RWTH Aachen University, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Mei Zhang
- Sophion Bioscience Inc., Bedford, MA, 01730, USA
| | | | - Irina Vetter
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, 4072, Australia
- School of Pharmacy, The University of Queensland, Woolloongabba, QLD, 4102, Australia
| | - Asa Andersson
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Gregory Dussor
- Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, 75080, USA
- Department of Neuroscience, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Theodore J Price
- Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, 75080, USA
- Department of Neuroscience, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Benedict J Kolber
- Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, 75080, USA
- Department of Neuroscience, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Vincent Truong
- Anatomic Incorporated, 2112 Broadway Street NE #135, Minneapolis, MN, 55413, USA
| | - Patrick Walsh
- Anatomic Incorporated, 2112 Broadway Street NE #135, Minneapolis, MN, 55413, USA
| | - Angelika Lampert
- Institute of Neurophysiology, Uniklinik RWTH Aachen University, Pauwelsstr. 30, 52074, Aachen, Germany.
- Research Training Group 2416 MultiSenses-MultiScales, RWTH Aachen University, Aachen, Germany.
- Scientific Center for Neuropathic Pain Aachen - SCN-Aachen, Uniklinik RWTH Aachen University, 52074, Aachen, Germany.
| |
Collapse
|
6
|
Srikanth KD, Elahi H, Chander P, Washburn HR, Hassler S, Mwirigi JM, Kume M, Loucks J, Arjarapu R, Hodge R, Shiers SI, Sankaranarayanan I, Erdjument-Bromage H, Neubert TA, Campbell ZT, Paik R, Price TJ, Dalva MB. VLK drives extracellular phosphorylation of EphB2 to govern the EphB2-NMDAR interaction and injury-induced pain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.18.585314. [PMID: 38562765 PMCID: PMC10983893 DOI: 10.1101/2024.03.18.585314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Phosphorylation of hundreds of protein extracellular domains is mediated by two kinase families, yet the significance of these kinases is underexplored. Here, we find that the presynaptic release of the tyrosine directed-ectokinase, Vertebrate Lonesome Kinase (VLK/Pkdcc), is necessary and sufficient for the direct extracellular interaction between EphB2 and GluN1 at synapses, for phosphorylation of the ectodomain of EphB2, and for injury-induced pain. Pkdcc is an essential gene in the nervous system, and VLK is found in synaptic vesicles, and is released from neurons in a SNARE-dependent fashion. VLK is expressed by nociceptive sensory neurons where presynaptic sensory neuron-specific knockout renders mice impervious to post-surgical pain, without changing proprioception. VLK defines an extracellular mechanism that regulates protein-protein interaction and non-opioid-dependent pain in response to injury.
Collapse
Affiliation(s)
- Kolluru D. Srikanth
- Tulane Brain Institute, Department of Cell and Molecular Biology, Tulane University; New Orleans, LA 70118, USA
- Jefferson Synaptic Biology Center, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107
| | - Hajira Elahi
- Department of Neuroscience, The University of Texas at Dallas; Richardson, TX 75080, USA
- Center for Advanced Pain Studies, University of Texas at Dallas; Richardson, TX 75080, USA
| | - Praveen Chander
- Tulane Brain Institute, Department of Cell and Molecular Biology, Tulane University; New Orleans, LA 70118, USA
- Jefferson Synaptic Biology Center, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107
| | - Halley R. Washburn
- Jefferson Synaptic Biology Center, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107
- Department of Molecular Biology, Princeton University; Princeton, NJ 08544, USA
| | - Shayne Hassler
- Department of Neuroscience, The University of Texas at Dallas; Richardson, TX 75080, USA
- College of Medicine, University of Houston; Houston, TX 77004, USA
| | - Juliet M. Mwirigi
- Department of Neuroscience, The University of Texas at Dallas; Richardson, TX 75080, USA
- Center for Advanced Pain Studies, University of Texas at Dallas; Richardson, TX 75080, USA
| | - Moeno Kume
- Department of Neuroscience, The University of Texas at Dallas; Richardson, TX 75080, USA
- Center for Advanced Pain Studies, University of Texas at Dallas; Richardson, TX 75080, USA
| | - Jessica Loucks
- Department of Neuroscience, The University of Texas at Dallas; Richardson, TX 75080, USA
| | - Rohita Arjarapu
- Department of Neuroscience, The University of Texas at Dallas; Richardson, TX 75080, USA
| | - Rachel Hodge
- Jefferson Synaptic Biology Center, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107
| | - Stephanie I. Shiers
- Department of Neuroscience, The University of Texas at Dallas; Richardson, TX 75080, USA
- Center for Advanced Pain Studies, University of Texas at Dallas; Richardson, TX 75080, USA
| | - Ishwarya Sankaranarayanan
- Department of Neuroscience, The University of Texas at Dallas; Richardson, TX 75080, USA
- Center for Advanced Pain Studies, University of Texas at Dallas; Richardson, TX 75080, USA
| | - Hediye Erdjument-Bromage
- Department of Neuroscience and Physiology and Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, 10016, USA
| | - Thomas A. Neubert
- Department of Neuroscience and Physiology and Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, 10016, USA
| | - Zachary T. Campbell
- Department of Anesthesiology, University of Wisconsin-Madison; Madison, WI 53792, USA
| | - Raehum Paik
- Department of Anesthesiology, University of Wisconsin-Madison; Madison, WI 53792, USA
- Department of Genetics, University of Texas Health Science Center at San Antonio; San Antonio, TX 78229, USA
| | - Theodore J. Price
- Department of Neuroscience, The University of Texas at Dallas; Richardson, TX 75080, USA
- Center for Advanced Pain Studies, University of Texas at Dallas; Richardson, TX 75080, USA
| | - Matthew B. Dalva
- Tulane Brain Institute, Department of Cell and Molecular Biology, Tulane University; New Orleans, LA 70118, USA
- Jefferson Synaptic Biology Center, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107
| |
Collapse
|
7
|
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
|
8
|
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
|
9
|
Parnell J, Martin N, Dedek A, Rudyk C, Landrigan J, Bellavance J, VanDerLoo S, Tsai EC, Hildebrand ME. Cannabinoid CB1 Receptor Expression and Localization in the Dorsal Horn of Male and Female Rat and Human Spinal Cord. Can J Pain 2023; 7:2264895. [PMID: 38170158 PMCID: PMC10761112 DOI: 10.1080/24740527.2023.2264895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 09/26/2023] [Indexed: 01/05/2024]
Abstract
Background Preclinical and clinical evidence suggests that cannabis has potential analgesic properties. However, cannabinoid receptor expression and localization within spinal cord pain processing circuits remain to be characterized across sex and species. Aims We aimed to investigate the differential expression of the cannabinoid type 1 (CB1) receptor across dorsal horn laminae and cell populations in male and female adult rats and humans. Methods To investigate and quantify CB1 receptor expression in the spinal dorsal horn across species, we refined immunohistochemical procedures for successful rat and human fixed tissue staining and confocal imaging. Immunohistochemical results were complemented with analysis of CB1 gene (CNR1) expression within rodent and human dorsal horn using single-cell/nuclei RNA sequencing data sets. Results We found that CB1 was preferentially localized to the neuropil within the superficial dorsal horn of both rats and humans, with CB1 somatic staining across dorsal horn laminae. CB1 receptor immunoreactivity was significantly higher in the superficial dorsal horn compared to the deeper dorsal horn laminae for both rats and humans, which was conserved across sex. Interestingly, we found that CB1 immunoreactivity was not primarily localized to peptidergic afferents in rats and humans and that CNR1 (CB1) but not CNR2 (CB2) was robustly expressed in dorsal horn neuron subpopulations of both rodents and humans. Conclusions The conserved preferential expression of CB1 receptors in the superficial dorsal horn in male and female rodents and humans has significant implications for understanding the roles of this cannabinoid receptor in spinal mechanisms of nociception and analgesia.
Collapse
Affiliation(s)
- Jessica Parnell
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Newton Martin
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Annemarie Dedek
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Christopher Rudyk
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Jeffrey Landrigan
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Justin Bellavance
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Simon VanDerLoo
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Eve C. Tsai
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Brain and Mind Research Institute, University of Ottawa, Ottawa, Ontario, Canada
- Division of Neurosurgery, Department of Surgery, The Ottawa Hospital, Ottawa, Ontario, Canada
| | - Michael E. Hildebrand
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| |
Collapse
|
10
|
Zhu T, Li H, Chen Y, Jia X, Ma X, Liu X, Feng Y, Ke J. ALPK1 Expressed in IB4-Positive Neurons of Mice Trigeminal Ganglions Promotes MIA-Induced TMJ pain. Mol Neurobiol 2023; 60:6264-6274. [PMID: 37442857 DOI: 10.1007/s12035-023-03462-0] [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: 12/25/2022] [Accepted: 06/22/2023] [Indexed: 07/15/2023]
Abstract
Pain is one of the main reasons for patients with temporomandibular joint (TMJ) disorders seeking medical care. However, there is no effective treatment yet as its mechanism remains unclear. Herein, we found that the injection of monoiodoacetate (MIA) into mice TMJs can induce typical joint pain as early as 3 days, accompanied by an increased percentage of calcitonin gene-related peptide positive (CGRP+) neurons and isolectin B4 positive (IB4+) in the trigeminal ganglions (TGs). Our previous study has discovered that alpha-kinase 1 (ALPK1) may be involved in joint pain. Here, we detected the expression of ALPK1 in neurons of TGs in wild-type (WT) mice, and it was upregulated after intra-TMJ injection of MIA. Meanwhile, the increased percentage of neurons in TGs expressing ALPK1 and CGRP or ALPK1 and IB4 was also demonstrated by the immunofluorescent double staining. Furthermore, after the MIA injection, ALPK1-/- mice exhibited attenuated pain behavior, as well as a remarkably decreased percentage of IB4+ neurons and an unchanged percentage of CGRP+ neurons, as compared with WT mice. In vitro assay showed that the value of calcium intensity was weakened in Dil+ neurons from ALPK1-/- mice of TMJ pain induced by the MIA injection, in relation to those from WT mice, while it was significantly enhanced with the incubation of recombinant human ALPK1 (rhA). Taken together, these results suggest that ALPK1 promotes mice TMJ pain induced by MIA through upregulation of the sensitization of IB4+ neurons in TGs. This study will provide a new potential therapeutic target for the treatment of TMJ pain.
Collapse
Affiliation(s)
- Taomin Zhu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Wuhan, 430079, Hubei Province, China
- Department of Oral and Maxillofacial Trauma and Temporomandibular Joint Surgery, Hubei-MOST KLOS & KLOBM, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Huimin Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Wuhan, 430079, Hubei Province, China
- Department of Oral and Maxillofacial Trauma and Temporomandibular Joint Surgery, Hubei-MOST KLOS & KLOBM, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Yuxiang Chen
- GuangDong Women and Children Hospital, Guangdong, 511400, China
| | - Xueke Jia
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Wuhan, 430079, Hubei Province, China
- Department of Oral and Maxillofacial Trauma and Temporomandibular Joint Surgery, Hubei-MOST KLOS & KLOBM, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Xiaohan Ma
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Wuhan, 430079, Hubei Province, China
- Department of Oral and Maxillofacial Trauma and Temporomandibular Joint Surgery, Hubei-MOST KLOS & KLOBM, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Xin Liu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Wuhan, 430079, Hubei Province, China
- Department of Oral and Maxillofacial Trauma and Temporomandibular Joint Surgery, Hubei-MOST KLOS & KLOBM, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Yaping Feng
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Wuhan, 430079, Hubei Province, China
- Department of Oral and Maxillofacial Trauma and Temporomandibular Joint Surgery, Hubei-MOST KLOS & KLOBM, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Jin Ke
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Wuhan, 430079, Hubei Province, China.
- Department of Oral and Maxillofacial Trauma and Temporomandibular Joint Surgery, Hubei-MOST KLOS & KLOBM, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China.
| |
Collapse
|
11
|
Kalia AK, Rösseler C, Granja-Vazquez R, Ahmad A, Pancrazio JJ, Neureiter A, Zhang M, Sauter D, Vetter I, Andersson A, Dussor G, Price TJ, Kolber BJ, Truong V, Walsh P, Lampert A. How to differentiate induced pluripotent stem cells into sensory neurons for disease modelling: a comparison of two protocols. RESEARCH SQUARE 2023:rs.3.rs-3127017. [PMID: 37961300 PMCID: PMC10635298 DOI: 10.21203/rs.3.rs-3127017/v1] [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
Background Human induced pluripotent stem cell (iPSC)-derived peripheral sensory neurons present a valuable tool to model human diseases and are a source for applications in drug discovery and regenerative medicine. Clinically, peripheral sensory neuropathies can result in maladies ranging from a complete loss of pain to severe painful neuropathic symptoms. Sensory neurons are located in the dorsal root ganglion and are comprised of functionally diverse neuronal types. Low efficiency, reproducibility concerns, variations arising due to genetic factors and time needed to generate functionally mature neuronal populations from iPSCs for disease modelling remain key challenges to study human nociception in vitro. Here, we report a detailed characterization of iPSC-derived sensory neurons with an accelerated differentiation protocol ("Anatomic" protocol) compared to the most commonly used small molecule approach ("Chambers" protocol). Methods Multiple iPSC clones derived from different reprogramming methods, genetics, age, and somatic cell sources were used to generate sensory neurons. Expression profiling of sensory neurons was performed with Immunocytochemistry and in situ hybridization techniques. Manual patch clamp and high throughput cellular screening systems (Fluorescence imaging plate reader, automated patch clamp and multi-well microelectrode arrays recordings) were applied to functionally characterize the generated sensory neurons. Results The Anatomic protocol rendered a purer culture without the use of mitomycin C to suppress non-neuronal outgrowth, while Chambers differentiations yielded a mix of cell types. High throughput systems confirmed functional expression of Na+ and K+ ion channels. Multi-well microelectrode recordings display spontaneously active neurons with sensitivity to increased temperature indicating expression of heat sensitive ion channels. Patient-derived nociceptors displayed higher frequency firing compared to control subject with both, Chambers and Anatomic differentiation approaches, underlining their potential use for clinical phenotyping as a disease-in-a-dish model. Conclusions We validated the efficiency of two differentiation protocols and their potential application for understanding the disease mechanisms from patients suffering from pain disorders. We propose that both differentiation methods can be further exploited for understanding mechanisms and development of novel treatments in pain disorders.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Mei Zhang
- Sophion Bioscience A/S: Biolin Scientific AB
| | | | - Irina Vetter
- The University of Queensland Institute for Molecular Bioscience
| | - Asa Andersson
- The University of Queensland Institute for Molecular Bioscience
| | | | | | | | | | | | | |
Collapse
|
12
|
Yousuf MS, Sahn JJ, Yang H, David ET, Shiers S, Moreno MM, Iketem J, Royer DM, Garcia CD, Zhang J, Hong VM, Mian SM, Ahmad A, Kolber BJ, Liebl DJ, Martin SF, Price TJ. Highly specific σ 2R/TMEM97 ligand alleviates neuropathic pain and inhibits the integrated stress response. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.11.536439. [PMID: 37090527 PMCID: PMC10120691 DOI: 10.1101/2023.04.11.536439] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
The Sigma 2 receptor (σ2R) was described pharmacologically more than three decades ago, but its molecular identity remained obscure until recently when it was identified as transmembrane protein 97 (TMEM97). We and others have shown that σ2R/TMEM97 ligands alleviate mechanical hypersensitivity in mouse neuropathic pain models with a time course wherein maximal anti-nociceptive effect is approximately 24 hours following dosing. We sought to understand this unique anti-neuropathic pain effect by addressing two key questions: do these σ2R/TMEM97 compounds act selectively via the receptor, and what is their downstream mechanism on nociceptive neurons? Using male and female conventional knockout (KO) mice for Tmem97, we find that a new σ2R/TMEM97 binding compound, FEM-1689, requires the presence of the gene to produce anti-nociception in the spared nerve injury model in mice. Using primary mouse dorsal root ganglion (DRG) neurons, we demonstrate that FEM-1689 inhibits the integrated stress response (ISR) and promotes neurite outgrowth via a σ2R/TMEM97-specific action. We extend the clinical translational value of these findings by showing that FEM-1689 reduces ISR and p-eIF2α levels in human sensory neurons and that it alleviates the pathogenic engagement of ISR by methylglyoxal. We also demonstrate that σ2R/TMEM97 is expressed in human nociceptors and satellite glial cells. These results validate σ2R/TMEM97 as a promising target for further development for the treatment of neuropathic pain.
Collapse
Affiliation(s)
- Muhammad Saad Yousuf
- Center for Advanced Pain Studies and Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080
- NuvoNuro, Austin, TX 78712
| | - James J. Sahn
- Department of Chemistry, University of Texas at Austin, Austin, TX 78712
- NuvoNuro, Austin, TX 78712
| | - Hongfen Yang
- Department of Chemistry, University of Texas at Austin, Austin, TX 78712
| | - Eric T. David
- Center for Advanced Pain Studies and Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080
| | - Stephanie Shiers
- Center for Advanced Pain Studies and Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080
| | - Marisol Mancilla Moreno
- Center for Advanced Pain Studies and Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080
| | - Jonathan Iketem
- Center for Advanced Pain Studies and Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080
| | - Danielle M. Royer
- Center for Advanced Pain Studies and Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080
| | - Chelsea D. Garcia
- Center for Advanced Pain Studies and Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080
| | - Jennifer Zhang
- Center for Advanced Pain Studies and Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080
| | - Veronica M. Hong
- Center for Advanced Pain Studies and Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080
| | - Subhaan M. Mian
- Center for Advanced Pain Studies and Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080
| | - Ayesha Ahmad
- Center for Advanced Pain Studies and Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080
| | - Benedict J. Kolber
- Center for Advanced Pain Studies and Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080
| | | | - Stephen F. Martin
- Department of Chemistry, University of Texas at Austin, Austin, TX 78712
- NuvoNuro, Austin, TX 78712
| | - Theodore J. Price
- Center for Advanced Pain Studies and Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080
- NuvoNuro, Austin, TX 78712
| |
Collapse
|
13
|
Grlickova-Duzevik E, Reimonn TM, Michael M, Tian T, Owyoung J, McGrath-Conwell A, Neufeld P, Mueth M, Molliver DC, Ward PJ, Harrison BJ. Members of the CUGBP Elav-like family of RNA-binding proteins are expressed in distinct populations of primary sensory neurons. J Comp Neurol 2023; 531:1425-1442. [PMID: 37537886 DOI: 10.1002/cne.25520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 05/16/2023] [Accepted: 06/10/2023] [Indexed: 08/05/2023]
Abstract
Primary sensory dorsal root ganglia (DRG) neurons are diverse, with distinct populations that respond to specific stimuli. Previously, we observed that functionally distinct populations of DRG neurons express mRNA transcript variants with different 3' untranslated regions (3'UTRs). 3'UTRs harbor binding sites for interaction with RNA-binding proteins (RBPs) for transporting mRNAs to subcellular domains, modulating transcript stability, and regulating the rate of translation. In the current study, analysis of publicly available single-cell RNA-sequencing data generated from adult mice revealed that 17 3'UTR-binding RBPs were enriched in specific populations of DRG neurons. This included four members of the CUG triplet repeat (CUGBP) Elav-like family (CELF): CELF2 and CELF4 were enriched in peptidergic, CELF6 in both peptidergic and nonpeptidergic, and CELF3 in tyrosine hydroxylase-expressing neurons. Immunofluorescence studies confirmed that 60% of CELF4+ neurons are small-diameter C fibers and 33% medium-diameter myelinated (likely Aδ) fibers and showed that CELF4 is distributed to peripheral termini. Coexpression analyses using transcriptomic data and immunofluorescence revealed that CELF4 is enriched in nociceptive neurons that express GFRA3, CGRP, and the capsaicin receptor TRPV1. Reanalysis of published transcriptomic data from macaque DRG revealed a highly similar distribution of CELF members, and reanalysis of single-nucleus RNA-sequencing data derived from mouse and rat DRG after sciatic injury revealed differential expression of CELFs in specific populations of sensory neurons. We propose that CELF RBPs may regulate the fate of mRNAs in populations of nociceptors, and may play a role in pain and/or neuronal regeneration following nerve injury.
Collapse
Affiliation(s)
- Eliza Grlickova-Duzevik
- Biomedical Sciences, College of Osteopathic Medicine, University of New England, Biddeford, Maine, USA
- Center for Excellence in the Neurosciences, University of New England, Biddeford, Maine, USA
| | - Thomas M Reimonn
- Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Merilla Michael
- Biomedical Sciences, College of Osteopathic Medicine, University of New England, Biddeford, Maine, USA
- Center for Excellence in the Neurosciences, University of New England, Biddeford, Maine, USA
| | - Tina Tian
- Medical Scientist Training Program, Emory University, Atlanta, Georgia, USA
- Neuroscience Graduate Program, Emory University, Atlanta, Georgia, USA
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Jordan Owyoung
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia, USA
- Genetics and Molecular Biology Graduate Program, Emory University, Atlanta, Georgia, USA
| | - Aidan McGrath-Conwell
- Center for Excellence in the Neurosciences, University of New England, Biddeford, Maine, USA
- College of Arts and Sciences, University of New England, Biddeford, Maine, USA
| | - Peter Neufeld
- Center for Excellence in the Neurosciences, University of New England, Biddeford, Maine, USA
- College of Arts and Sciences, University of New England, Biddeford, Maine, USA
| | - Madison Mueth
- Center for Excellence in the Neurosciences, University of New England, Biddeford, Maine, USA
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, Maine, USA
| | - Derek C Molliver
- Biomedical Sciences, College of Osteopathic Medicine, University of New England, Biddeford, Maine, USA
- Center for Excellence in the Neurosciences, University of New England, Biddeford, Maine, USA
| | - Patricia Jillian Ward
- Neuroscience Graduate Program, Emory University, Atlanta, Georgia, USA
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Benjamin J Harrison
- Biomedical Sciences, College of Osteopathic Medicine, University of New England, Biddeford, Maine, USA
- Center for Excellence in the Neurosciences, University of New England, Biddeford, Maine, USA
| |
Collapse
|
14
|
Berta T, Strong JA, Zhang JM, Ji RR. Targeting dorsal root ganglia and primary sensory neurons for the treatment of chronic pain: an update. Expert Opin Ther Targets 2023; 27:665-678. [PMID: 37574713 PMCID: PMC10530032 DOI: 10.1080/14728222.2023.2247563] [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: 04/04/2023] [Revised: 06/30/2023] [Accepted: 08/09/2023] [Indexed: 08/15/2023]
Abstract
INTRODUCTION Current treatments for chronic pain are inadequate. Here, we provide an update on the new therapeutic strategies that target dorsal root ganglia (DRGs) in the peripheral nervous system for a better and safer treatment of chronic pain. AREAS COVERED Despite the complex nature of chronic pain and its underlying mechanisms, we do know that changes in the plasticity and modality of neurons in DRGs play a pivotal role. DRG neurons are heterogenous and offer potential pain targets for different therapeutic interventions. We discuss the last advancements of these interventions, which include the use of systemic and local administrations, selective nerve drug delivery, and gene therapy. In particular, we provide updates and further details on the molecular characterization of primary sensory neurons, new analgesics entering the market, and future gene therapy approaches. EXPERT OPINION DRGs and primary sensory neurons are promising targets for chronic pain treatment due to their key role in pain signaling, unique anatomical location, and the potential for different targeted therapeutic interventions.
Collapse
Affiliation(s)
- Temugin Berta
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH 45267, USA
| | - Judith A. Strong
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH 45267, USA
| | - Jun-Ming Zhang
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH 45267, USA
| | - Ru-Rong Ji
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
- Departments of Cell Biology and Neurobiology, Duke University Medical Center, Durham, North Carolina 27710
| |
Collapse
|
15
|
Mitchell ME, Cook LC, Shiers S, Tavares-Ferreira D, Akopian AN, Dussor G, Price TJ. Characterization of Fragile X Mental Retardation Protein expression in human nociceptors and their axonal projections to the spinal dorsal horn. J Comp Neurol 2023; 531:814-835. [PMID: 36808110 PMCID: PMC10038933 DOI: 10.1002/cne.25463] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 12/20/2022] [Accepted: 01/27/2023] [Indexed: 02/23/2023]
Abstract
Fragile X Mental Retardation Protein (FMRP) regulates activity-dependent RNA localization and local translation to modulate synaptic plasticity throughout the central nervous system. Mutations in the FMR1 gene that hinder or ablate FMRP function cause Fragile X Syndrome (FXS), a disorder associated with sensory processing dysfunction. FXS premutations are associated with increased FMRP expression and neurological impairments including sex dimorphic presentations of chronic pain. In mice, FMRP ablation causes dysregulated dorsal root ganglion (DRG) neuron excitability and synaptic vesicle exocytosis, spinal circuit activity, and decreased translation-dependent nociceptive sensitization. Activity-dependent, local translation is a key mechanism for enhancing primary nociceptor excitability that promotes pain in animals and humans. These works indicate that FMRP likely regulates nociception and pain at the level of the primary nociceptor or spinal cord. Therefore, we sought to better understand FMRP expression in the human DRG and spinal cord using immunostaining in organ donor tissues. We find that FMRP is highly expressed in DRG and spinal neuron subsets with substantia gelatinosa exhibiting the most abundant immunoreactivity in spinal synaptic fields. Here, it is expressed in nociceptor axons. FMRP puncta colocalized with Nav1.7 and TRPV1 receptor signals suggesting a pool of axoplasmic FMRP localizes to plasma membrane-associated loci in these branches. Interestingly, FMRP puncta exhibited notable colocalization with calcitonin gene-related peptide (CGRP) immunoreactivity selectively in female spinal cord. Our results support a regulatory role for FMRP in human nociceptor axons of the dorsal horn and implicate it in the sex dimorphic actions of CGRP signaling in nociceptive sensitization and chronic pain.
Collapse
Affiliation(s)
- Molly E Mitchell
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas, USA
| | - Lauren C Cook
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas, USA
| | - Stephanie Shiers
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas, USA
| | - Diana Tavares-Ferreira
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas, USA
| | - Armen N Akopian
- Department of Endodontics, UT Health San Antonio, San Antonio, Texas, USA
| | - Gregory Dussor
- 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
|
16
|
Monné Rodríguez JM, Frisk AL, Kreutzer R, Lemarchand T, Lezmi S, Saravanan C, Stierstorfer B, Thuilliez C, Vezzali E, Wieczorek G, Yun SW, Schaudien D. European Society of Toxicologic Pathology (Pathology 2.0 Molecular Pathology Special Interest Group): Review of In Situ Hybridization Techniques for Drug Research and Development. Toxicol Pathol 2023; 51:92-111. [PMID: 37449403 PMCID: PMC10467011 DOI: 10.1177/01926233231178282] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2023]
Abstract
In situ hybridization (ISH) is used for the localization of specific nucleic acid sequences in cells or tissues by complementary binding of a nucleotide probe to a specific target nucleic acid sequence. In the last years, the specificity and sensitivity of ISH assays were improved by innovative techniques like synthetic nucleic acids and tandem oligonucleotide probes combined with signal amplification methods like branched DNA, hybridization chain reaction and tyramide signal amplification. These improvements increased the application spectrum for ISH on formalin-fixed paraffin-embedded tissues. ISH is a powerful tool to investigate DNA, mRNA transcripts, regulatory noncoding RNA, and therapeutic oligonucleotides. ISH can be used to obtain spatial information of a cell type, subcellular localization, or expression levels of targets. Since immunohistochemistry and ISH share similar workflows, their combination can address simultaneous transcriptomics and proteomics questions. The goal of this review paper is to revisit the current state of the scientific approaches in ISH and its application in drug research and development.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Seong-Wook Yun
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany
| | - Dirk Schaudien
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
| |
Collapse
|
17
|
Russo AF, Hay DL. CGRP physiology, pharmacology, and therapeutic targets: migraine and beyond. Physiol Rev 2023; 103:1565-1644. [PMID: 36454715 PMCID: PMC9988538 DOI: 10.1152/physrev.00059.2021] [Citation(s) in RCA: 83] [Impact Index Per Article: 83.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 11/23/2022] [Accepted: 11/27/2022] [Indexed: 12/03/2022] Open
Abstract
Calcitonin gene-related peptide (CGRP) is a neuropeptide with diverse physiological functions. Its two isoforms (α and β) are widely expressed throughout the body in sensory neurons as well as in other cell types, such as motor neurons and neuroendocrine cells. CGRP acts via at least two G protein-coupled receptors that form unusual complexes with receptor activity-modifying proteins. These are the CGRP receptor and the AMY1 receptor; in rodents, additional receptors come into play. Although CGRP is known to produce many effects, the precise molecular identity of the receptor(s) that mediates CGRP effects is seldom clear. Despite the many enigmas still in CGRP biology, therapeutics that target the CGRP axis to treat or prevent migraine are a bench-to-bedside success story. This review provides a contextual background on the regulation and sites of CGRP expression and CGRP receptor pharmacology. The physiological actions of CGRP in the nervous system are discussed, along with updates on CGRP actions in the cardiovascular, pulmonary, gastrointestinal, immune, hematopoietic, and reproductive systems and metabolic effects of CGRP in muscle and adipose tissues. We cover how CGRP in these systems is associated with disease states, most notably migraine. In this context, we discuss how CGRP actions in both the peripheral and central nervous systems provide a basis for therapeutic targeting of CGRP in migraine. Finally, we highlight potentially fertile ground for the development of additional therapeutics and combinatorial strategies that could be designed to modulate CGRP signaling for migraine and other diseases.
Collapse
Affiliation(s)
- Andrew F Russo
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa
- Department of Neurology, University of Iowa, Iowa City, Iowa
- Center for the Prevention and Treatment of Visual Loss, Department of Veterans Affairs Health Center, Iowa City, Iowa
| | - Debbie L Hay
- Department of Pharmacology and Toxicology, University of Otago, Dunedin, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, School of Biological Sciences, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
18
|
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: 0] [Impact Index Per Article: 0] [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
|
19
|
Austin PJ, Karrasch JF, O'Brien JA. A dual role of microbiota type 17 immunity in tissue repair and pain. Immunol Cell Biol 2023; 101:281-284. [PMID: 36789629 DOI: 10.1111/imcb.12630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 02/12/2023] [Indexed: 02/16/2023]
Abstract
In this commentary, we discuss the findings of Enamorado et al. who have, for the first time, demonstrated that immunity to the microbiota enhances repair of cutaneous sensory nerves and epithelial tissues following skin injury. Commensal-specific IL-17 producing CD4+ T helper cells have direct contact with injured sensory neurons, inducing multiple epithelial and neuronal repair genes. We speculate that an altered balance of T cell populations in the skin of people with chronic neuropathic pain may contribute to a reduction in neuronal repair and the consequent decease in intraepidermal nerve fibre density and persistent pain.
Collapse
Affiliation(s)
- Paul J Austin
- Brain and Mind Centre, Laboratory of Neuroimmunology and Behaviour, Neuroscience theme, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia
| | - Jackson F Karrasch
- Brain and Mind Centre, Laboratory of Neuroimmunology and Behaviour, Neuroscience theme, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia
| | - Jayden A O'Brien
- Brain and Mind Centre, Laboratory of Neuroimmunology and Behaviour, Neuroscience theme, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia
| |
Collapse
|
20
|
Shiers S, Funk G, Cervantes A, Horton P, Dussor G, Hennen S, Price TJ. Na V1.7 mRNA and protein expression in putative projection neurons of the human spinal dorsal horn. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.04.527110. [PMID: 36778234 PMCID: PMC9915702 DOI: 10.1101/2023.02.04.527110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
NaV1.7, a membrane-bound voltage-gated sodium channel, is preferentially expressed along primary sensory neurons, including their peripheral & central nerve endings, axons, and soma within the dorsal root ganglia and plays an integral role in amplifying membrane depolarization and pain neurotransmission. Loss- and gain-of-function mutations in the gene encoding NaV1.7, SCN9A, are associated with a complete loss of pain sensation or exacerbated pain in humans, respectively. As an enticing pain target supported by human genetic validation, many compounds have been developed to inhibit NaV1.7 but have disappointed in clinical trials. The underlying reasons are still unclear, but recent reports suggest that inhibiting NaV1.7 in central terminals of nociceptor afferents is critical for achieving pain relief by pharmacological inhibition of NaV1.7. We report for the first time that NaV1.7 mRNA is expressed in putative projection neurons (NK1R+) in the human spinal dorsal horn, predominantly in lamina 1 and 2, as well as in deep dorsal horn neurons and motor neurons in the ventral horn. NaV1.7 protein was found in the central axons of sensory neurons terminating in lamina 1-2, but also was detected in the axon initial segment of resident spinal dorsal horn neurons and in axons entering the anterior commissure. Given that projection neurons are critical for conveying nociceptive information from the dorsal horn to the brain, these data support that dorsal horn NaV1.7 expression may play an unappreciated role in pain phenotypes observed in humans with genetic SCN9A mutations, and in achieving analgesic efficacy in clinical trials.
Collapse
Affiliation(s)
- Stephanie Shiers
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | | | | | | | - Gregory Dussor
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | | | - Theodore J. Price
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| |
Collapse
|
21
|
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
|
22
|
Reissig LF, Carrero-Rojas G, Maierhofer U, Moghaddam AS, Hainfellner A, Gesslbauer B, Haider T, Streicher J, Aszmann OC, Pastor AM, Weninger WJ, Blumer R. Spinal cord from body donors is suitable for multicolor immunofluorescence. Histochem Cell Biol 2023; 159:23-45. [PMID: 36201037 PMCID: PMC9899749 DOI: 10.1007/s00418-022-02154-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/10/2022] [Indexed: 02/07/2023]
Abstract
Immunohistochemistry is a powerful tool for studying neuronal tissue from humans at the molecular level. Obtaining fresh neuronal tissue from human organ donors is difficult and sometimes impossible. In anatomical body donations, neuronal tissue is dedicated to research purposes and because of its easier availability, it may be an alternative source for research. In this study, we harvested spinal cord from a single organ donor 2 h (h) postmortem and spinal cord from body donors 24, 48, and 72 h postmortem and tested how long after death, valid multi-color immunofluorescence or horseradish peroxidase (HRP) immunohistochemistry is possible. We used general and specific neuronal markers and glial markers for immunolabeling experiments. Here we showed that it is possible to visualize molecularly different neuronal elements with high precision in the body donor spinal cord 24 h postmortem and the quality of the image data was comparable to those from the fresh organ donor spinal cord. High-contrast multicolor images of the 24-h spinal cords allowed accurate automated quantification of different neuronal elements in the same sample. Although there was antibody-specific signal reduction over postmortem intervals, the signal quality for most antibodies was acceptable at 48 h but no longer at 72 h postmortem. In conclusion, our study has defined a postmortem time window of more than 24 h during which valid immunohistochemical information can be obtained from the body donor spinal cord. Due to the easier availability, neuronal tissue from body donors is an alternative source for basic and clinical research.
Collapse
Affiliation(s)
- Lukas F. Reissig
- Division of Anatomy, MIC, Medical University Vienna, Vienna, Austria
| | | | - Udo Maierhofer
- Clinical Laboratory for Bionic Extremity Reconstruction, Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Vienna, Austria
| | | | | | - Bernhard Gesslbauer
- Clinical Laboratory for Bionic Extremity Reconstruction, Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Vienna, Austria
| | - Thomas Haider
- Department of Orthopedic and Trauma Surgery, Medical University of Vienna, Vienna, Austria
| | - Johannes Streicher
- Department of Anatomy and Biomechanics, Division of Anatomy and Developmental Biology, Karl Landsteiner University of Health Science, Krems an der Donau, Austria
| | - Oskar C. Aszmann
- Clinical Laboratory for Bionic Extremity Reconstruction, Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Vienna, Austria
| | - Angel M. Pastor
- Departamento de Fisiología, Facultad de Biología, Universidad de Sevilla, Seville, Spain
| | | | - Roland Blumer
- Division of Anatomy, MIC, Medical University Vienna, Vienna, Austria
| |
Collapse
|
23
|
High-fat diet causes mechanical allodynia in the absence of injury or diabetic pathology. Sci Rep 2022; 12:14840. [PMID: 36050326 PMCID: PMC9437006 DOI: 10.1038/s41598-022-18281-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 08/09/2022] [Indexed: 12/04/2022] Open
Abstract
Understanding the interactions between diet, obesity, and diabetes is important to tease out mechanisms in painful pathology. Western diet is rich in fats, producing high amounts of circulating bioactive metabolites. However, no research has assessed how a high-fat diet (HFD) alone may sensitize an individual to non-painful stimuli in the absence of obesity or diabetic pathology. To investigate this, we tested the ability of a HFD to stimulate diet-induced hyperalgesic priming, or diet sensitization in male and female mice. Our results revealed that 8 weeks of HFD did not alter baseline pain sensitivity, but both male and female HFD-fed animals exhibited robust mechanical allodynia when exposed to a subthreshold dose of intraplantar Prostaglandin E2 (PGE2) compared to mice on chow diet. Furthermore, calcium imaging in isolated primary sensory neurons of both sexes revealed HFD induced an increased percentage of capsaicin-responsive neurons compared to their chow counterparts. Immunohistochemistry (IHC) showed a HFD-induced upregulation of ATF3, a neuronal marker of injury, in lumbar dorsal root ganglia (DRG). This suggests that a HFD induces allodynia in the absence of a pre-existing condition or injury via dietary components. With this new understanding of how a HFD can contribute to the onset of pain, we can understand the dissociation behind the comorbidities associated with obesity and diabetes to develop pharmacological interventions to treat them more efficiently.
Collapse
|
24
|
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: 2.0] [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
|
25
|
Carozzi VA, Salio C, Rodriguez-Menendez V, Ciglieri E, Ferrini F. 2D <em>vs</em> 3D morphological analysis of dorsal root ganglia in health and painful neuropathy. Eur J Histochem 2021; 65. [PMID: 34664808 PMCID: PMC8547168 DOI: 10.4081/ejh.2021.3276] [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: 05/28/2021] [Accepted: 08/16/2021] [Indexed: 11/23/2022] Open
Abstract
Dorsal root ganglia (DRGs) are clusters of sensory neurons that transmit the sensory information from the periphery to the central nervous system, and satellite glial cells (SGCs), their supporting trophic cells. Sensory neurons are pseudounipolar neurons with a heterogeneous neurochemistry reflecting their functional features. DRGs, not protected by the blood brain barrier, are vulnerable to stress and damage of different origin (i.e., toxic, mechanical, metabolic, genetic) that can involve sensory neurons, SGCs or, considering their intimate intercommunication, both cell populations. DRG damage, primary or secondary to nerve damage, produces a sensory peripheral neuropathy, characterized by neurophysiological abnormalities, numbness, paraesthesia and dysesthesia, tingling and burning sensations and neuropathic pain. DRG stress can be morphologically detected by light and electron microscope analysis with alterations in cell size (swelling/atrophy) and in different subcellular compartments (i.e., mitochondria, endoplasmic reticulum, and nucleus) of neurons and/or SGCs. In addition, neurochemical changes can be used to portray abnormalities of neurons and SGC. Conventional immunostaining, i.e., immunohistochemical detection of specific molecules in tissue slices, can be employed to detect, localize and quantify particular markers of damage in neurons (i.e., nuclear expression of ATF3) or SGCs (i.e., increased expression of GFAP), markers of apoptosis (i.e., caspases), markers of mitochondrial suffering and oxidative stress (i.e., 8-OHdG), markers of tissue inflammation (i.e., CD68 for macrophage infiltration) etc. However classical (2D) methods of immunostaining disrupt the overall organization of the DRG, thus resulting in the loss of some crucial information. Whole-mount (3D) methods have been recently developed to investigate DRG morphology and neurochemistry without tissue slicing, giving the opportunity to study the intimate relationship between SGCs and sensory neurons in health and disease. Here, we aim to compare classical (2D) vs whole-mount (3D) approaches to highlight “pros” and “cons” of the two methodologies when analysing neuropathy-induced alterations in DRGs.
Collapse
Affiliation(s)
- Valentina Alda Carozzi
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, Monza (MB).
| | - Chiara Salio
- Department of Veterinary Sciences, University of Turin, Grugliasco (TO).
| | | | | | - Francesco Ferrini
- Department of Veterinary Sciences, University of Turin, Grugliasco (TO).
| |
Collapse
|
26
|
Javed H, Rehmathulla S, Tariq S, Ali MA, Emerald BS, Shehab S. Co-localization of nociceptive markers in the lumbar dorsal root ganglion and spinal cord of dromedary camel. J Comp Neurol 2021; 529:3710-3725. [PMID: 34468017 DOI: 10.1002/cne.25240] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/19/2021] [Accepted: 08/26/2021] [Indexed: 11/12/2022]
Abstract
Nociceptive markers in mice have been identified in two distinct peptidergic and nonpeptidergic neurons in the dorsal root ganglion (DRG) and distributed in different laminae of the dorsal horn of the spinal cord. Recently, however, a study in humans showed a significant overlapping in these two populations. In this study, we investigated the distribution of various nociceptive markers in the lumbar DRG and spinal cord of the dromedary camel. Immunohistochemical data showed a remarkable percentage of total neurons in the DRG expressed IB4 binding (54.5%), calcitonin gene-related peptide (CGRP; 49.5%), transient receptor potential vanilloid 1 (TRPV1; 48.2%), and nitric oxide synthase (NOS; 30.6%). The co-localization data showed that 89.6% and 74.0% of CGRP- and TRPV1-labeled neurons, respectively, were IB4 positive. In addition, 61.6% and 84.2% of TRPV1- and NOS-immunoreactive neurons, respectively, were also co-localized with CGRP. The distribution of IB4, CGRP, TRPV1, substance P, and NOS immunoreactivities in the spinal cord were observed in lamina I and outer lamina II (IIo). Quantitative data showed that 82.4% of IB4-positive nerve terminals in laminae I and IIo were co-localized with CGRP, and 86.0% of CGRP-labeled terminals were co-localized with IB4. Similarly, 85.1% of NOS-labeled nerve terminals were co-localized with CGRP. No neuropeptide Y (NPY) or cholecystokinin (CCK) immunoreactivities were detected in the DRG, and no co-localization between IB4, NPY, and CCK were observed in the spinal cord. Our results demonstrate marked convergence of nociceptive markers in the primary afferent neurons in camels, which is similar to humans rather than the mouse. The data also emphasizes the importance of interspecies differences when selecting ideal animal models for studying nociception and treating chronic pain.
Collapse
Affiliation(s)
- Hayate Javed
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, UAE
| | - Sumisha Rehmathulla
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, UAE
| | - Saeed Tariq
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, UAE
| | - Mahmoud A Ali
- Department of Pharmacology, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, UAE
| | - Bright Starling Emerald
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, UAE
| | - Safa Shehab
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, UAE
| |
Collapse
|
27
|
Middleton SJ, Barry AM, Comini M, Li Y, Ray PR, Shiers S, Themistocleous AC, Uhelski ML, Yang X, Dougherty PM, Price TJ, Bennett DL. Studying human nociceptors: from fundamentals to clinic. Brain 2021; 144:1312-1335. [PMID: 34128530 PMCID: PMC8219361 DOI: 10.1093/brain/awab048] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 11/26/2020] [Accepted: 12/08/2020] [Indexed: 12/14/2022] Open
Abstract
Chronic pain affects one in five of the general population and is the third most important cause of disability-adjusted life-years globally. Unfortunately, treatment remains inadequate due to poor efficacy and tolerability. There has been a failure in translating promising preclinical drug targets into clinic use. This reflects challenges across the whole drug development pathway, from preclinical models to trial design. Nociceptors remain an attractive therapeutic target: their sensitization makes an important contribution to many chronic pain states, they are located outside the blood-brain barrier, and they are relatively specific. The past decade has seen significant advances in the techniques available to study human nociceptors, including: the use of corneal confocal microscopy and biopsy samples to observe nociceptor morphology, the culture of human nociceptors (either from surgical or post-mortem tissue or using human induced pluripotent stem cell derived nociceptors), the application of high throughput technologies such as transcriptomics, the in vitro and in vivo electrophysiological characterization through microneurography, and the correlation with pain percepts provided by quantitative sensory testing. Genome editing in human induced pluripotent stem cell-derived nociceptors enables the interrogation of the causal role of genes in the regulation of nociceptor function. Both human and rodent nociceptors are more heterogeneous at a molecular level than previously appreciated, and while we find that there are broad similarities between human and rodent nociceptors there are also important differences involving ion channel function, expression, and cellular excitability. These technological advances have emphasized the maladaptive plastic changes occurring in human nociceptors following injury that contribute to chronic pain. Studying human nociceptors has revealed new therapeutic targets for the suppression of chronic pain and enhanced repair. Cellular models of human nociceptors have enabled the screening of small molecule and gene therapy approaches on nociceptor function, and in some cases have enabled correlation with clinical outcomes. Undoubtedly, challenges remain. Many of these techniques are difficult to implement at scale, current induced pluripotent stem cell differentiation protocols do not generate the full diversity of nociceptor populations, and we still have a relatively poor understanding of inter-individual variation in nociceptors due to factors such as age, sex, or ethnicity. We hope our ability to directly investigate human nociceptors will not only aid our understanding of the fundamental neurobiology underlying acute and chronic pain but also help bridge the translational gap.
Collapse
Affiliation(s)
- Steven J Middleton
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Allison M Barry
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Maddalena Comini
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Yan Li
- Department of Anesthesia and Pain Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Pradipta R Ray
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Stephanie Shiers
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Andreas C Themistocleous
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK.,Brain Function Research Group, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2000, South Africa
| | - Megan L Uhelski
- Department of Anesthesia and Pain Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Xun Yang
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Patrick M Dougherty
- Brain Function Research Group, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2000, South Africa
| | - Theodore J Price
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX 75080, USA
| | - David L Bennett
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| |
Collapse
|