1
|
Tokizane K, Imai SI. Protocol to study inter-tissue communication between the hypothalamus and white adipose tissue and lifespan using a chemogenetic approach in aged mice. STAR Protoc 2025; 6:103551. [PMID: 39798096 PMCID: PMC11969402 DOI: 10.1016/j.xpro.2024.103551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 10/28/2024] [Accepted: 12/12/2024] [Indexed: 01/15/2025] Open
Abstract
Here, we present a protocol for assessing the impact of a chemogenetic manipulation in a subpopulation of the hypothalamic neurons on aging and lifespan control using a mouse model developed specifically for this purpose. We describe steps for stereotaxic viral injection and assess inter-tissue communication between protein phosphatase 1 regulatory subunit 17 (Ppp1r17)-expressing neurons in the dorsomedial hypothalamus and white adipose tissue. We then detail procedures for lifespan measurements following chemogenetic manipulation in aged mice. For complete details on the use and execution of this protocol, please refer to Tokizane et al.1.
Collapse
Affiliation(s)
- Kyohei Tokizane
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Shin-Ichiro Imai
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
2
|
Kang HJ, Krumm BE, Tassou A, Geron M, DiBerto JF, Kapolka NJ, Gumpper RH, Sakamoto K, Dewran Kocak D, Olsen RHJ, Huang XP, Zhang S, Huang KL, Zaidi SA, Nguyen MT, Jo MJ, Katritch V, Fay JF, Scherrer G, Roth BL. Structure-guided design of a peripherally restricted chemogenetic system. Cell 2024; 187:7433-7449.e20. [PMID: 39631393 DOI: 10.1016/j.cell.2024.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 07/30/2024] [Accepted: 11/04/2024] [Indexed: 12/07/2024]
Abstract
Designer receptors exclusively activated by designer drugs (DREADDs) are chemogenetic tools for remotely controlling cellular signaling, neural activity, behavior, and physiology. Using a structure-guided approach, we provide a peripherally restricted Gi-DREADD, hydroxycarboxylic acid receptor DREADD (HCAD), whose native receptor is minimally expressed in the brain, and a chemical actuator that does not cross the blood-brain barrier (BBB). This was accomplished by combined mutagenesis, analoging via an ultra-large make-on-demand library, structural determination of the designed DREADD receptor via cryoelectron microscopy (cryo-EM), and validation of HCAD function. Expression and activation of HCAD in dorsal root ganglion (DRG) neurons inhibit action potential (AP) firing and reduce both acute and tissue-injury-induced inflammatory pain. The HCAD chemogenetic system expands the possibilities for studying numerous peripheral systems with little adverse effects on the central nervous system (CNS). The structure-guided approach used to generate HCAD also has the potential to accelerate the development of emerging chemogenetic tools for basic and translational sciences.
Collapse
Affiliation(s)
- Hye Jin Kang
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| | - Brian E Krumm
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Adrien Tassou
- Department of Cell Biology and Physiology, UNC Neuroscience Center, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Matan Geron
- Department of Cell Biology and Physiology, UNC Neuroscience Center, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jeffrey F DiBerto
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Nicholas J Kapolka
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ryan H Gumpper
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kensuke Sakamoto
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - D Dewran Kocak
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Reid H J Olsen
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Xi-Ping Huang
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; National Institute of Mental Health Psychoactive Drug Screening Program (NIMH-PDSP), School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Shicheng Zhang
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Karen L Huang
- Department of Cell Biology and Physiology, UNC Neuroscience Center, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Saheem A Zaidi
- Department of Quantitative and Computational Biology, Department of Chemistry, Center for New Technologies in Drug Discovery and Development, Bridge Institute, Michelson Center for Convergent Biosciences, University of Southern California, Los Angeles, CA, USA
| | - MyV T Nguyen
- Department of Quantitative and Computational Biology, Department of Chemistry, Center for New Technologies in Drug Discovery and Development, Bridge Institute, Michelson Center for Convergent Biosciences, University of Southern California, Los Angeles, CA, USA
| | - Min Jeong Jo
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| | - Vsevolod Katritch
- Department of Quantitative and Computational Biology, Department of Chemistry, Center for New Technologies in Drug Discovery and Development, Bridge Institute, Michelson Center for Convergent Biosciences, University of Southern California, Los Angeles, CA, USA
| | - Jonathan F Fay
- Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, Baltimore, MD 21201, USA
| | - Grégory Scherrer
- Department of Cell Biology and Physiology, UNC Neuroscience Center, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Bryan L Roth
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
3
|
Ben-Shaanan TL, Knöpper K, Duan L, Liu R, Taglinao H, Xu Y, An J, Plikus MV, Cyster JG. Dermal TRPV1 innervations engage a macrophage- and fibroblast-containing pathway to activate hair growth in mice. Dev Cell 2024; 59:2818-2833.e7. [PMID: 38851191 PMCID: PMC11537826 DOI: 10.1016/j.devcel.2024.05.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 02/25/2024] [Accepted: 05/15/2024] [Indexed: 06/10/2024]
Abstract
Pain, detected by nociceptors, is an integral part of injury, yet whether and how it can impact tissue physiology and recovery remain understudied. Here, we applied chemogenetics in mice to locally activate dermal TRPV1 innervations in naive skin and found that it triggered new regenerative cycling by dormant hair follicles (HFs). This was preceded by rapid apoptosis of dermal macrophages, mediated by the neuropeptide calcitonin gene-related peptide (CGRP). TRPV1 activation also triggered a macrophage-dependent induction of osteopontin (Spp1)-expressing dermal fibroblasts. The neuropeptide CGRP and the extracellular matrix protein Spp1 were required for the nociceptor-triggered hair growth. Finally, we showed that epidermal abrasion injury induced Spp1-expressing dermal fibroblasts and hair growth via a TRPV1 neuron and CGRP-dependent mechanism. Collectively, these data demonstrated a role for TRPV1 nociceptors in orchestrating a macrophage and fibroblast-supported mechanism to promote hair growth and enabling the efficient restoration of this mechano- and thermo-protective barrier after wounding.
Collapse
Affiliation(s)
- Tamar L Ben-Shaanan
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA.
| | - Konrad Knöpper
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Lihui Duan
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Ruiqi Liu
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Hanna Taglinao
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Ying Xu
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jinping An
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Maksim V Plikus
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Jason G Cyster
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
4
|
Kabanova A, Yang M, Logothetis NK, Eschenko O. Partial chemogenetic inhibition of the locus coeruleus due to heterogeneous transduction of noradrenergic neurons preserved auditory salience processing in wild-type rats. Eur J Neurosci 2024; 60:6237-6253. [PMID: 39349382 DOI: 10.1111/ejn.16550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 08/30/2024] [Accepted: 09/09/2024] [Indexed: 10/02/2024]
Abstract
The acoustic startle reflex (ASR) and prepulse inhibition of the ASR (PPI) assess the efficiency of salience processing, a fundamental brain function that is impaired in many psychiatric conditions. Both ASR and PPI depend on noradrenergic transmission, yet the modulatory role of the locus coeruleus (LC) remains controversial. Clonidine (0.05 mg/kg, i.p.), an alpha2-adrenoreceptor agonist, strongly reduced the ASR amplitude. In contrast, chemogenetic LC inhibition only mildly suppressed the ASR and did affect the PPI in virus-transduced rats. The canine adenovirus type 2 (CAV2)-based vector carrying a gene cassette for the expression of inhibitory receptors (hM4Di) and noradrenergic cell-specific promoter (PRSx8) had high cell-type specificity (94.4 ± 3.1%) but resulted in heterogeneous virus transduction of DbH-positive LC neurons (range: 9.2-94.4%). Clozapine-N-oxide (CNO; 1 mg/kg, i.p.), a hM4Di actuator, caused the firing cessation of hM4Di-expressing LC neurons, yet complete inhibition of the entire population of LC neurons was not achieved. Case-based immunohistochemistry revealed that virus injections distal (> 150 μm) to the LC core resulted in partial LC transduction, while proximal (< 50 μm) injections caused neuronal loss due to virus neurotoxicity. Neither the ASR nor PPI differed between the intact and virus-transduced rats. Our results suggest that a residual activity of virus-non-transduced LC neurons might have been sufficient for mediating an unaltered ASR and PPI. Our study highlights the importance of a case-based assessment of the virus efficiency, specificity, and neurotoxicity for targeted cell populations and of considering these factors when interpreting behavioral effects in experiments employing chemogenetic modulation.
Collapse
Affiliation(s)
- Anna Kabanova
- Department of Physiology of Cognitive Processes, Max Planck Institute for Biological Cybernetics, Tübingen, Germany
| | - Mingyu Yang
- Department of Physiology of Cognitive Processes, Max Planck Institute for Biological Cybernetics, Tübingen, Germany
| | - Nikos K Logothetis
- Department of Physiology of Cognitive Processes, Max Planck Institute for Biological Cybernetics, Tübingen, Germany
- International Center for Primate Brain Research, Center for Excellence in Brain Science and Intelligence Technology (CEBSIT), Institute of Neuroscience (ION), Chinese Academy of Sciences, Shanghai, China
- Division of Imaging Science and Biomedical Engineering, University of Manchester, Manchester, UK
| | - Oxana Eschenko
- Department of Physiology of Cognitive Processes, Max Planck Institute for Biological Cybernetics, Tübingen, Germany
| |
Collapse
|
5
|
Miyake T, Tanaka K, Inoue Y, Nagai Y, Nishimura R, Seta T, Nakagawa S, Inoue KI, Hasegawa E, Minamimoto T, Doi M. Size-reduced DREADD derivatives for AAV-assisted multimodal chemogenetic control of neuronal activity and behavior. CELL REPORTS METHODS 2024; 4:100881. [PMID: 39437713 PMCID: PMC11573748 DOI: 10.1016/j.crmeth.2024.100881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 07/24/2024] [Accepted: 09/23/2024] [Indexed: 10/25/2024]
Abstract
Designer receptors exclusively activated by designer drugs (DREADDs) are engineered G-protein-coupled receptors that afford reversible manipulation of neuronal activity in vivo. Here, we introduce size-reduced DREADD derivatives miniDq and miniDi, which inherit the basic receptor properties from the Gq-coupled excitatory receptor hM3Dq and the Gi-coupled inhibitory receptor hM4Di, respectively, while being approximately 30% smaller in size. Taking advantage of the compact size of the receptors, we generated an adeno-associated virus (AAV) vector carrying both miniDq and the other DREADD family receptor (κ-opioid receptor-based inhibitory DREADD [KORD]) within the maximum AAV capacity (4.7 kb), allowing us to modulate neuronal activity and animal behavior in both excitatory and inhibitory directions using a single viral vector. We confirmed that expressing miniDq, but not miniDi, allowed activation of striatum activity in the cynomolgus monkey (Macaca fascicularis). The compact DREADDs may thus widen the opportunity for multiplexed interrogation and/or intervention in neuronal regulation in mice and non-human primates.
Collapse
Affiliation(s)
- Takahito Miyake
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyō-ku, Kyoto 606-8501, Japan.
| | - Kaho Tanaka
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyō-ku, Kyoto 606-8501, Japan
| | - Yutsuki Inoue
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyō-ku, Kyoto 606-8501, Japan
| | - Yuji Nagai
- Advanced Neuroimaging Center, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
| | - Reo Nishimura
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyō-ku, Kyoto 606-8501, Japan
| | - Takehito Seta
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyō-ku, Kyoto 606-8501, Japan
| | - Shumpei Nakagawa
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyō-ku, Kyoto 606-8501, Japan
| | - Ken-Ichi Inoue
- Systems Neuroscience Section, Center for the Evolutionary Origins of Human Behavior, Kyoto University, Inuyama 484-8506, Japan
| | - Emi Hasegawa
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyō-ku, Kyoto 606-8501, Japan
| | - Takafumi Minamimoto
- Advanced Neuroimaging Center, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
| | - Masao Doi
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyō-ku, Kyoto 606-8501, Japan.
| |
Collapse
|
6
|
Li YZ, Ji RR. Gene therapy for chronic pain management. Cell Rep Med 2024; 5:101756. [PMID: 39366385 PMCID: PMC11513853 DOI: 10.1016/j.xcrm.2024.101756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 07/20/2024] [Accepted: 09/09/2024] [Indexed: 10/06/2024]
Abstract
Despite significant advances in identifying molecular targets for chronic pain over the past two decades, many remain difficult to target with traditional methods. Gene therapies such as antisense oligonucleotides (ASOs), RNA interference (RNAi), CRISPR, and virus-based delivery systems have played crucial roles in discovering and validating new pain targets. While there has been a surge in gene therapy-based clinical trials, those focusing on pain as the primary outcome remain uncommon. This review examines various gene therapy strategies, including ASOs, small interfering RNA (siRNAs), optogenetics, chemogenetics, and CRISPR, and their delivery methods targeting primary sensory neurons and non-neuronal cells, including glia and chondrocytes. We also explore emerging gene therapy tools and highlight gene therapy's clinical potential in pain management, including trials targeting pain-related diseases. Advances in single-cell analysis of sensory neurons and non-neuronal cells, along with the development of new delivery tools, are poised to accelerate the application of gene therapy in pain medicine.
Collapse
Affiliation(s)
- Yi-Ze Li
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Ru-Rong Ji
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA; Departments of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA; Departments of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
7
|
Saloman JL, Epouhe AY, Ruff CF, Albers KM. PDX1, a transcription factor essential for organ differentiation, regulates SERCA-dependent Ca 2+ homeostasis in sensory neurons. Cell Calcium 2024; 120:102884. [PMID: 38574509 PMCID: PMC11188734 DOI: 10.1016/j.ceca.2024.102884] [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: 01/05/2024] [Revised: 03/13/2024] [Accepted: 03/31/2024] [Indexed: 04/06/2024]
Abstract
Pancreatic and duodenal homeobox 1 (PDX1) is a transcription factor required for the development and differentiation of the pancreas. Previous studies indicated that PDX1 expression was restricted to the gastrointestinal tract. Using a cre-dependent reporter, we observed PDX1-dependent expression of tdtomato (PDX1-tom) in a subpopulation of sensory nerves. Many of these PDX1-tom afferents expressed the neurofilament 200 protein and projected to the skin. Tdtomato-labeled terminals were associated with hair follicles in the form of longitudinal and circumferential lanceolate endings suggesting a role in tactile and proprioceptive perception. To begin to examine the functional significance of PDX1 in afferents, we used Fura-2 imaging to examine calcium (Ca2+) handling under naïve and nerve injury conditions. Neuropathic injury is associated with increased intracellular Ca2+ signaling that in part results from dysregulation of the sarco/endoplasmic reticulum calcium transport ATPase (SERCA). Here we demonstrate that under naïve conditions, PDX1 regulates expression of the SERCA2B isoform in sensory neurons. In response to infraorbital nerve injury, a significant reduction of PDX1 and SERCA2B expression and dysregulation of Ca2+ handling occurs in PDX1-tom trigeminal ganglia neurons. The identification of PDX1 expression in the somatosensory system and its regulation of SERCA2B and Ca2+ handling provide a new mechanism to explain pathological changes in primary afferents that may contribute to pain associated with nerve injury.
Collapse
Affiliation(s)
- Jami L Saloman
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Neurobiology, Center for Neuroscience and Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Ariel Y Epouhe
- Department of Neurobiology, Center for Neuroscience and Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Catherine F Ruff
- Department of Neurobiology, Center for Neuroscience and Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Kathryn M Albers
- Department of Neurobiology, Center for Neuroscience and Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
8
|
Kumar PA, Stallman J, Kharbat Y, Hoppe J, Leonards A, Kerim E, Nguyen B, Adkins RL, Baltazar A, Milligan S, Letchuman S, Hook MA, Dulin JN. Chemogenetic Attenuation of Acute Nociceptive Signaling Enhances Functional Outcomes Following Spinal Cord Injury. J Neurotrauma 2024; 41:1060-1076. [PMID: 37905504 PMCID: PMC11564839 DOI: 10.1089/neu.2023.0141] [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] [Indexed: 11/02/2023] Open
Abstract
Identifying novel therapeutic approaches to promote recovery of neurological functions following spinal cord injury (SCI) remains a great unmet need. Nociceptive signaling in the acute phase of SCI has been shown to inhibit recovery of locomotor function and promote the development of chronic neuropathic pain. We therefore hypothesized that inhibition of nociceptive signaling in the acute phase of SCI might improve long-term functional outcomes in the chronic phase of injury. To test this hypothesis, we took advantage of a selective strategy utilizing AAV6 to deliver inhibitory (hM4Di) Designer Receptors Exclusively Activated by Designer Drugs (DREADDs) to nociceptors of the L4-L6 dorsal root ganglia to evaluate the effects of transient nociceptor silencing on long-term sensory and motor functional outcomes in a rat thoracic contusion SCI model. Following hM4Di-mediated nociceptor inhibition from 0-14 days post-SCI, we conducted behavioral assessments until 70 days post-SCI, then performed histological assessments of lesion severity and axon plasticity. Our results show highly selective expression of hM4Di within small diameter nociceptors including calcitonin gene-related peptide (CGRP)+ and IB4-binding neurons. Expression of hM4Di in less than 25% of nociceptors was sufficient to increase hindlimb thermal withdrawal latency in naïve rats. Compared with subjects who received AAV-yellow fluorescent protein (YFP; control), subjects who received AAV-hM4Di exhibited attenuated thermal hyperalgesia, greater coordination, and improved hindlimb locomotor function. However, treatment did not impact the development of cold allodynia or mechanical hyperalgesia. Histological assessments of spinal cord tissue suggested trends toward reduced lesion volume, increased neuronal sparing and increased CGRP+ axon sprouting in hM4Di-treated animals. Together, these findings suggest that nociceptor silencing early after SCI may promote beneficial plasticity in the acute phase of injury that can impact long-term functional outcomes, and support previous work highlighting primary nociceptors as possible therapeutic targets for pain management after SCI.
Collapse
Affiliation(s)
| | - Jacob Stallman
- Department of Biology, Texas A&M University, College Station, Texas, USA
| | - Yahya Kharbat
- Department of Biology, Texas A&M University, College Station, Texas, USA
| | - Joseph Hoppe
- Department of Biology, Texas A&M University, College Station, Texas, USA
| | - Amy Leonards
- Department of Biology, Texas A&M University, College Station, Texas, USA
| | - Ethan Kerim
- Department of Biology, Texas A&M University, College Station, Texas, USA
| | - Britney Nguyen
- Department of Biology, Texas A&M University, College Station, Texas, USA
| | - Robert L. Adkins
- Department of Biology, Texas A&M University, College Station, Texas, USA
| | - Angelina Baltazar
- Department of Biology, Texas A&M University, College Station, Texas, USA
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
| | - Sara Milligan
- Department of Biology, Texas A&M University, College Station, Texas, USA
| | - Sunjay Letchuman
- Mays Business School, Texas A&M University, College Station, Texas, USA
| | - Michelle A. Hook
- Texas A&M Institute for Neuroscience, Texas A&M University, College Station, Texas, USA
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M Health Science Center, Bryan, Texas, USA
| | - Jennifer N. Dulin
- Department of Biology, Texas A&M University, College Station, Texas, USA
- Texas A&M Institute for Neuroscience, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
9
|
Defaye M, Bradaia A, Abdullah NS, Agosti F, Iftinca M, Delanne-Cuménal M, Soubeyre V, Svendsen K, Gill G, Ozmaeian A, Gheziel N, Martin J, Poulen G, Lonjon N, Vachiery-Lahaye F, Bauchet L, Basso L, Bourinet E, Chiu IM, Altier C. Induction of antiviral interferon-stimulated genes by neuronal STING promotes the resolution of pain in mice. J Clin Invest 2024; 134:e176474. [PMID: 38690737 PMCID: PMC11060736 DOI: 10.1172/jci176474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 03/12/2024] [Indexed: 05/03/2024] Open
Abstract
Inflammation and pain are intertwined responses to injury, infection, or chronic diseases. While acute inflammation is essential in determining pain resolution and opioid analgesia, maladaptive processes occurring during resolution can lead to the transition to chronic pain. Here we found that inflammation activates the cytosolic DNA-sensing protein stimulator of IFN genes (STING) in dorsal root ganglion nociceptors. Neuronal activation of STING promotes signaling through TANK-binding kinase 1 (TBK1) and triggers an IFN-β response that mediates pain resolution. Notably, we found that mice expressing a nociceptor-specific gain-of-function mutation in STING exhibited an IFN gene signature that reduced nociceptor excitability and inflammatory hyperalgesia through a KChIP1-Kv4.3 regulation. Our findings reveal a role of IFN-regulated genes and KChIP1 downstream of STING in the resolution of inflammatory pain.
Collapse
Affiliation(s)
- Manon Defaye
- Department of Physiology and Pharmacology, Cumming School of Medicine
- Inflammation Research Network–Snyder Institute for Chronic Diseases, Cumming School of Medicine, and
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Amyaouch Bradaia
- Department of Physiology and Pharmacology, Cumming School of Medicine
- Inflammation Research Network–Snyder Institute for Chronic Diseases, Cumming School of Medicine, and
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Nasser S. Abdullah
- Department of Physiology and Pharmacology, Cumming School of Medicine
- Inflammation Research Network–Snyder Institute for Chronic Diseases, Cumming School of Medicine, and
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Francina Agosti
- Department of Physiology and Pharmacology, Cumming School of Medicine
- Inflammation Research Network–Snyder Institute for Chronic Diseases, Cumming School of Medicine, and
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Mircea Iftinca
- Department of Physiology and Pharmacology, Cumming School of Medicine
- Inflammation Research Network–Snyder Institute for Chronic Diseases, Cumming School of Medicine, and
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Mélissa Delanne-Cuménal
- Department of Physiology and Pharmacology, Cumming School of Medicine
- Inflammation Research Network–Snyder Institute for Chronic Diseases, Cumming School of Medicine, and
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Vanessa Soubeyre
- Department of Neurosurgery, Gui de Chauliac Hospital, Donation and Transplantation Coordination Unit, Montpellier University Medical Center, Montpellier, France
| | - Kristofer Svendsen
- Department of Physiology and Pharmacology, Cumming School of Medicine
- Inflammation Research Network–Snyder Institute for Chronic Diseases, Cumming School of Medicine, and
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Gurveer Gill
- Department of Physiology and Pharmacology, Cumming School of Medicine
- Inflammation Research Network–Snyder Institute for Chronic Diseases, Cumming School of Medicine, and
| | - Aye Ozmaeian
- Department of Physiology and Pharmacology, Cumming School of Medicine
- Inflammation Research Network–Snyder Institute for Chronic Diseases, Cumming School of Medicine, and
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Nadine Gheziel
- Toulouse Institute for Infectious and Inflammatory Diseases (INFINITy), INSERM UMR1291, University of Toulouse III, Toulouse, France
| | - Jérémy Martin
- Toulouse Institute for Infectious and Inflammatory Diseases (INFINITy), INSERM UMR1291, University of Toulouse III, Toulouse, France
| | - Gaetan Poulen
- Department of Neurosurgery, Gui de Chauliac Hospital, Donation and Transplantation Coordination Unit, Montpellier University Medical Center, Montpellier, France
| | - Nicolas Lonjon
- Department of Neurosurgery, Gui de Chauliac Hospital, Donation and Transplantation Coordination Unit, Montpellier University Medical Center, Montpellier, France
| | - Florence Vachiery-Lahaye
- Department of Neurosurgery, Gui de Chauliac Hospital, Donation and Transplantation Coordination Unit, Montpellier University Medical Center, Montpellier, France
| | - Luc Bauchet
- Department of Neurosurgery, Gui de Chauliac Hospital, Donation and Transplantation Coordination Unit, Montpellier University Medical Center, Montpellier, France
- Institute of Functional Genomics, Montpellier University, CNRS, INSERM, Montpellier, France
| | - Lilian Basso
- Toulouse Institute for Infectious and Inflammatory Diseases (INFINITy), INSERM UMR1291, University of Toulouse III, Toulouse, France
| | - Emmanuel Bourinet
- Institute of Functional Genomics, Montpellier University, CNRS, INSERM, Montpellier, France
| | - Isaac M. Chiu
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Christophe Altier
- Department of Physiology and Pharmacology, Cumming School of Medicine
- Inflammation Research Network–Snyder Institute for Chronic Diseases, Cumming School of Medicine, and
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
10
|
Hsiao IH, Yen CM, Hsu HC, Liao HY, Lin YW. Chemogenetics Modulation of Electroacupuncture Analgesia in Mice Spared Nerve Injury-Induced Neuropathic Pain through TRPV1 Signaling Pathway. Int J Mol Sci 2024; 25:1771. [PMID: 38339048 PMCID: PMC10855068 DOI: 10.3390/ijms25031771] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 01/18/2024] [Accepted: 01/26/2024] [Indexed: 02/12/2024] Open
Abstract
Neuropathic pain, which is initiated by a malfunction of the somatosensory cortex system, elicits inflammation and simultaneously activates glial cells that initiate neuroinflammation. Electroacupuncture (EA) has been shown to have therapeutic effects for neuropathic pain, although with uncertain mechanisms. We suggest that EA can reliably cure neuropathic disease through anti-inflammation and transient receptor potential V1 (TRPV1) signaling pathways from the peripheral to the central nervous system. To explore this, we used EA to treat the mice spared nerve injury (SNI) model and explore the underlying molecular mechanisms through novel chemogenetics techniques. Both mechanical and thermal pain were found in SNI mice at four weeks (mechanical: 3.23 ± 0.29 g; thermal: 4.9 ± 0.14 s). Mechanical hyperalgesia was partially attenuated by 2 Hz EA (mechanical: 4.05 ± 0.19 g), and thermal hyperalgesia was fully reduced (thermal: 6.22 ± 0.26 s) but not with sham EA (mechanical: 3.13 ± 0.23 g; thermal: 4.58 ± 0.37 s), suggesting EA's specificity. In addition, animals with Trpv1 deletion showed partial mechanical hyperalgesia and no significant induction of thermal hyperalgesia in neuropathic pain mice (mechanical: 4.43 ± 0.26 g; thermal: 6.24 ± 0.09 s). Moreover, we found increased levels of inflammatory factors such as interleukin-1 beta (IL1-β), IL-3, IL-6, IL-12, IL-17, tumor necrosis factor alpha, and interferon gamma after SNI modeling, which decreased in the EA and Trpv1-/- groups rather than the sham group. Western blot and immunofluorescence analysis showed similar tendencies in the dorsal root ganglion, spinal cord dorsal horn, somatosensory cortex (SSC), and anterior cingulate cortex (ACC). In addition, a novel chemogenetics method was used to precisely inhibit SSC to ACC activity, which showed an analgesic effect through the TRPV1 pathway. In summary, our findings indicate a novel mechanism underlying neuropathic pain as a beneficial target for neuropathic pain.
Collapse
Affiliation(s)
- I-Han Hsiao
- School of Medicine, College of Medicine, China Medical University, Taichung 40402, Taiwan;
- Department of Neurosurgery, China Medical University Hospital, Taichung 40402, Taiwan
| | - Chia-Ming Yen
- Department of Anesthesiology, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung 42743, Taiwan;
- School of Post-Baccalaureate Chinese Medicine, Tzu Chi University, Hualien 97004, Taiwan
| | - Hsin-Cheng Hsu
- School of Post-Baccalaureate Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung 40402, Taiwan;
- Department of Traditional Chinese Medicine, China Medical University Hsinchu Hospital, Hsinchu 302, Taiwan
| | - Hsien-Yin Liao
- School of Post-Baccalaureate Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung 40402, Taiwan;
| | - Yi-Wen Lin
- Graduate Institute of Acupuncture Science, College of Chinese Medicine, China Medical University, Taichung 40402, Taiwan
- Chinese Medicine Research Center, China Medical University, Taichung 40402, Taiwan
| |
Collapse
|
11
|
Dheer A, Bosco DB, Zheng J, Wang L, Zhao S, Haruwaka K, Yi MH, Barath A, Tian DS, Wu LJ. Chemogenetic approaches reveal dual functions of microglia in seizures. Brain Behav Immun 2024; 115:406-418. [PMID: 37926132 PMCID: PMC10841657 DOI: 10.1016/j.bbi.2023.11.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 10/14/2023] [Accepted: 11/02/2023] [Indexed: 11/07/2023] Open
Abstract
Microglia are key players in maintaining brain homeostasis and exhibit phenotypic alterations in response to epileptic stimuli. However, it is still relatively unknown if these alterations are pro- or anti-epileptic. To unravel this dilemma, we employed chemogenetic manipulation of microglia using the artificial Gi-Dreadd receptor within a kainic acid (KA) induced murine seizure model. Our results indicate that acute Gi-Dreadd activation with Clozapine-N-Oxide can reduce seizure severity. Additionally, we observed increased interaction between microglia and neuronal soma, which correlated with reduced neuronal hyperactivity. Interestingly, prolonged activation of microglial Gi-Dreadds by repeated doses of CNO over 3 days, arrested microglia in a less active, homeostatic-like state, which associated with increased neuronal loss after KA induced seizures. RNAseq analysis revealed that prolonged activation of Gi-Dreadd interferes with interferon β signaling and microglia proliferation. Thus, our findings highlight the importance of microglial Gi signaling not only during status epilepticus (SE) but also within later seizure induced pathology.
Collapse
Affiliation(s)
- Aastha Dheer
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Dale B Bosco
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Jiaying Zheng
- Department of Neurology, Mayo Clinic, Rochester, MN, USA; Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
| | - Lingxiao Wang
- Department of Neurology, Mayo Clinic, Rochester, MN, USA; Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
| | - Shunyi Zhao
- Department of Neurology, Mayo Clinic, Rochester, MN, USA; Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
| | | | - Min-Hee Yi
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Abhijeet Barath
- Department of Neurology, Mayo Clinic, Rochester, MN, USA; Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
| | - Dai-Shi Tian
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN, USA; Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA; Department of Immunology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
12
|
Middleton SJ, Hu H, Perez-Sanchez J, Zuberi S, McGrath Williams J, Weir GA, Bennett DL. GluCl.Cre ON enables selective inhibition of molecularly defined pain circuits. Pain 2023; 164:2780-2791. [PMID: 37366588 PMCID: PMC10652717 DOI: 10.1097/j.pain.0000000000002976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 02/17/2023] [Accepted: 05/08/2023] [Indexed: 06/28/2023]
Abstract
ABSTRACT Insight into nociceptive circuits will ultimately build our understanding of pain processing and aid the development of analgesic strategies. Neural circuit analysis has been advanced greatly by the development of optogenetic and chemogenetic tools, which have allowed function to be ascribed to discrete neuronal populations. Neurons of the dorsal root ganglion, which include nociceptors, have proved challenging targets for chemogenetic manipulation given specific confounds with commonly used DREADD technology. We have developed a cre/lox dependant version of the engineered glutamate-gated chloride channel (GluCl) to restrict and direct its expression to molecularly defined neuronal populations. We have generated GluCl.Cre ON that selectively renders neurons expressing cre-recombinase susceptible to agonist-induced silencing. We have functionally validated our tool in multiple systems in vitro, and subsequently generated viral vectors and tested its applicability in vivo. Using Nav1.8 Cre mice to restrict AAV-GluCl.Cre ON to nociceptors, we demonstrate effective silencing of electrical activity in vivo and concomitant hyposensitivity to noxious thermal and noxious mechanical pain, whereas light touch and motor function remained intact. We also demonstrated that our strategy can effectively silence inflammatory-like pain in a chemical pain model. Collectively, we have generated a novel tool that can be used to selectively silence defined neuronal circuits in vitro and in vivo. We believe that this addition to the chemogenetic tool box will facilitate further understanding of pain circuits and guide future therapeutic development.
Collapse
Affiliation(s)
- Steven J. Middleton
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Huimin Hu
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Jimena Perez-Sanchez
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Sana Zuberi
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | | | - Greg A. Weir
- School of Psychology and Neuroscience, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - David L. Bennett
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
13
|
Perez-Sanchez J, Middleton SJ, Pattison LA, Hilton H, Awadelkareem MA, Zuberi SR, Renke MB, Hu H, Yang X, Clark AJ, Smith ESJ, Bennett DL. A humanized chemogenetic system inhibits murine pain-related behavior and hyperactivity in human sensory neurons. Sci Transl Med 2023; 15:eadh3839. [PMID: 37792955 PMCID: PMC7615191 DOI: 10.1126/scitranslmed.adh3839] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 09/15/2023] [Indexed: 10/06/2023]
Abstract
Hyperexcitability in sensory neurons is known to underlie many of the maladaptive changes associated with persistent pain. Chemogenetics has shown promise as a means to suppress such excitability, yet chemogenetic approaches suitable for human applications are needed. PSAM4-GlyR is a modular system based on the human α7 nicotinic acetylcholine and glycine receptors, which responds to inert chemical ligands and the clinically approved drug varenicline. Here, we demonstrated the efficacy of this channel in silencing both mouse and human sensory neurons by the activation of large shunting conductances after agonist administration. Virally mediated expression of PSAM4-GlyR in mouse sensory neurons produced behavioral hyposensitivity upon agonist administration, which was recovered upon agonist washout. Stable expression of the channel led to similar reversible suppression of pain-related behavior even after 10 months of viral delivery. Mechanical and spontaneous pain readouts were also ameliorated by PSAM4-GlyR activation in acute and joint pain inflammation mouse models. Furthermore, suppression of mechanical hypersensitivity generated by a spared nerve injury model of neuropathic pain was also observed upon activation of the channel. Effective silencing of behavioral hypersensitivity was reproduced in a human model of hyperexcitability and clinical pain: PSAM4-GlyR activation decreased the excitability of human-induced pluripotent stem cell-derived sensory neurons and spontaneous activity due to a gain-of-function NaV1.7 mutation causing inherited erythromelalgia. Our results demonstrate the contribution of sensory neuron hyperexcitability to neuropathic pain and the translational potential of an effective, stable, and reversible humanized chemogenetic system for the treatment of pain.
Collapse
Affiliation(s)
- Jimena Perez-Sanchez
- Nuffield Department of Clinical Neurosciences, University of Oxford; Oxford OX3 9DU, UK
| | - Steven J. Middleton
- Nuffield Department of Clinical Neurosciences, University of Oxford; Oxford OX3 9DU, UK
| | - Luke A. Pattison
- Department of Pharmacology, University of Cambridge; Cambridge CB2 1PD, UK
| | - Helen Hilton
- Department of Pharmacology, University of Cambridge; Cambridge CB2 1PD, UK
| | | | - Sana R. Zuberi
- Nuffield Department of Clinical Neurosciences, University of Oxford; Oxford OX3 9DU, UK
| | - Maria B. Renke
- Nuffield Department of Clinical Neurosciences, University of Oxford; Oxford OX3 9DU, UK
| | - Huimin Hu
- Nuffield Department of Clinical Neurosciences, University of Oxford; Oxford OX3 9DU, UK
| | - Xun Yang
- Nuffield Department of Clinical Neurosciences, University of Oxford; Oxford OX3 9DU, UK
| | - Alex J. Clark
- Blizard Institute, Barts and the London School of Medicine and Dentistry; London E1 2AT, UK
| | | | - David L. Bennett
- Nuffield Department of Clinical Neurosciences, University of Oxford; Oxford OX3 9DU, UK
| |
Collapse
|
14
|
Gore R, Esmail T, Pflepsen K, Marron Fernandez de Velasco E, Kitto KF, Riedl MS, Karlen A, McIvor RS, Honda CN, Fairbanks CA, Vulchanova L. AAV-mediated gene transfer to colon-innervating primary afferent neurons. FRONTIERS IN PAIN RESEARCH 2023; 4:1225246. [PMID: 37599864 PMCID: PMC10436501 DOI: 10.3389/fpain.2023.1225246] [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: 05/19/2023] [Accepted: 07/25/2023] [Indexed: 08/22/2023] Open
Abstract
Investigation of neural circuits underlying visceral pain is hampered by the difficulty in achieving selective manipulations of individual circuit components. In this study, we adapted a dual AAV approach, used for projection-specific transgene expression in the CNS, to explore the potential for targeted delivery of transgenes to primary afferent neurons innervating visceral organs. Focusing on the extrinsic sensory innervation of the mouse colon, we first characterized the extent of dual transduction following intrathecal delivery of one AAV9 vector and intracolonic delivery of a second AAV9 vector. We found that if the two AAV9 vectors were delivered one week apart, dorsal root ganglion (DRG) neuron transduction by the second vector was greatly diminished. Following delivery of the two viruses on the same day, we observed colocalization of the transgenes in DRG neurons, indicating dual transduction. Next, we delivered intrathecally an AAV9 vector encoding the inhibitory chemogenetic actuator hM4D(Gi) in a Cre-recombinase dependent manner, and on the same day injected an AAV9 vector carrying Cre-recombinase in the colon. DRG expression of hM4D(Gi) was demonstrated at the mRNA and protein level. However, we were unable to demonstrate selective inhibition of visceral nociception following hM4D(Gi) activation. Taken together, these results establish a foundation for development of strategies for targeted transduction of primary afferent neurons for neuromodulation of peripheral neural circuits.
Collapse
Affiliation(s)
- Reshma Gore
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Tina Esmail
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Kelsey Pflepsen
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN, United States
| | | | - Kelley F. Kitto
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Maureen S. Riedl
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Andrea Karlen
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN, United States
| | - R. Scott McIvor
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN, United States
| | - Christopher N. Honda
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Carolyn A. Fairbanks
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN, United States
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, United States
| | - Lucy Vulchanova
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
15
|
Obeidat AM, Wood MJ, Adamczyk NS, Ishihara S, Li J, Wang L, Ren D, Bennett DA, Miller RJ, Malfait AM, Miller RE. Piezo2 expressing nociceptors mediate mechanical sensitization in experimental osteoarthritis. Nat Commun 2023; 14:2479. [PMID: 37120427 PMCID: PMC10148822 DOI: 10.1038/s41467-023-38241-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 04/17/2023] [Indexed: 05/01/2023] Open
Abstract
Non-opioid targets are needed for addressing osteoarthritis pain, which is mechanical in nature and associated with daily activities such as walking and climbing stairs. Piezo2 has been implicated in the development of mechanical pain, but the mechanisms by which this occurs remain poorly understood, including the role of nociceptors. Here we show that nociceptor-specific Piezo2 conditional knock-out mice were protected from mechanical sensitization associated with inflammatory joint pain in female mice, joint pain associated with osteoarthritis in male mice, as well as both knee swelling and joint pain associated with repeated intra-articular injection of nerve growth factor in male mice. Single cell RNA sequencing of mouse lumbar dorsal root ganglia and in situ hybridization of mouse and human lumbar dorsal root ganglia revealed that a subset of nociceptors co-express Piezo2 and Ntrk1 (the gene that encodes the nerve growth factor receptor TrkA). These results suggest that nerve growth factor-mediated sensitization of joint nociceptors, which is critical for osteoarthritic pain, is also dependent on Piezo2, and targeting Piezo2 may represent a therapeutic option for osteoarthritis pain control.
Collapse
Affiliation(s)
- Alia M Obeidat
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, USA
| | - Matthew J Wood
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, USA
| | - Natalie S Adamczyk
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, USA
| | - Shingo Ishihara
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, USA
| | - Jun Li
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, USA
| | - Lai Wang
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, USA
| | - Dongjun Ren
- Department of Pharmacology, Northwestern University, Chicago, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center and Department of Neurological Sciences, Rush University Medical Center, Chicago, USA
| | - Richard J Miller
- Department of Pharmacology, Northwestern University, Chicago, USA
| | - Anne-Marie Malfait
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, USA
| | - Rachel E Miller
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, USA.
| |
Collapse
|
16
|
Zuk KE, Cansler HL, Wang J, Meeks JP. Arc-Expressing Accessory Olfactory Bulb Interneurons Support Chemosensory Social Behavioral Plasticity. J Neurosci 2023; 43:1178-1190. [PMID: 36623874 PMCID: PMC9962775 DOI: 10.1523/jneurosci.0847-22.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 11/18/2022] [Accepted: 11/22/2022] [Indexed: 01/11/2023] Open
Abstract
The accessory olfactory system (AOS) is critical for the development and expression of social behavior. The first dedicated circuit in the AOS, the accessory olfactory bulb (AOB), exhibits cellular and network plasticity in male and female mice after social experience. In the AOB, interneurons called internal granule cells (IGCs) express the plasticity-associated immediate-early gene Arc following intermale aggression or mating. Here, we sought to better understand how Arc-expressing IGCs shape AOB information processing and social behavior in the context of territorial aggression. We used "ArcTRAP" (Arc-CreERT2) transgenic mice to selectively and permanently label Arc-expressing IGCs following male-male resident-intruder interactions. Using whole-cell patch-clamp electrophysiology, we found that Arc-expressing IGCs display increased intrinsic excitability for several days after a single resident-intruder interaction. Further, we found that Arc-expressing IGCs maintain this increased excitability across repeated resident-intruder interactions, during which resident mice increase or "ramp" their aggression. We tested the hypothesis that Arc-expressing IGCs participate in ramping aggression. Using a combination of ArcTRAP mice and chemogenetics (Cre-dependent hM4D(Gi)-mCherry AAV injections), we found that disruption of Arc-expressing IGC activity during repeated resident-intruder interactions abolishes the ramping aggression exhibited by resident male mice. This work shows that Arc-expressing AOB IGC ensembles are activated by specific chemosensory environments, and play an integral role in the establishment and expression of sex-typical social behavior. These studies identify a population of plastic interneurons in an early chemosensory circuit that display physiological features consistent with simple memory formation, increasing our understanding of central chemosensory processing and mammalian social behavior.SIGNIFICANCE STATEMENT The accessory olfactory system plays a vital role in rodent chemosensory social behavior. We studied experience-dependent plasticity in the accessory olfactory bulb and found that internal granule cells expressing the immediate-early gene Arc after the resident-intruder paradigm increase their excitability for several days. We investigated the roles of these Arc-expressing internal granule cells on chemosensory social behavior by chemogenetically manipulating their excitability during repeated social interactions. We found that inhibiting these cells eliminated intermale aggressive ramping behavior. These studies identify a population of plastic interneurons in an early chemosensory circuit that display physiological features consistent with simple memory formation, increasing our understanding of central chemosensory processing and mammalian social behavior.
Collapse
Affiliation(s)
- Kelsey E Zuk
- Neuroscience Graduate Program, University of Texas Southwestern Medical Center, Dallas, Texas 75390
- Department of Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| | - Hillary L Cansler
- Department of Pharmacology, University of Florida College of Medicine, Gainesville, Florida 32603
| | - Jinxin Wang
- Department of Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| | - Julian P Meeks
- Department of Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
- Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| |
Collapse
|
17
|
Patra PH, Tench B, Hitrec T, Holmes F, Drake R, Cerritelli S, Spanswick D, Pickering AE. Pro-Opiomelanocortin (POMC) neurons in the nucleus of the solitary tract mediate endorphinergic endogenous analgesia in mice. Pain 2022; 164:1051-1066. [PMID: 36448978 DOI: 10.1097/j.pain.0000000000002802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 09/27/2022] [Indexed: 12/02/2022]
Abstract
ABSTRACT The nucleus of the solitary tract (NTS) contains pro-opiomelanocortin (POMC) neurons which are one of the two major sources of β-endorphin in the brain. The functional role of these NTS POMC neurons in nociceptive and cardiorespiratory function is debated. We have shown that NTS POMC optogenetic activation produces bradycardia and transient apnoea in a working heart brainstem preparation and chemogenetic activation with an engineered ion channel (PSAM) produced opioidergic analgesia in vivo . To better define the role of the NTS POMC neurons in behaving animals, we adopted in vivo optogenetics (ChrimsonR) and excitatory/inhibitory chemogenetic DREADD (hM3Dq/hM4Di) strategies in POMC-Cre mice. We show that optogenetic activation of NTS POMC neurons produces time-locked, graded, transient bradycardia and bradypnoea in anaesthetised mice which is naloxone sensitive (1 mg/kg, i.p) suggesting a role of β-endorphin. Both optogenetic and chemogenetic activation of NTS POMC neurons produces sustained thermal analgesia in behaving mice which can be blocked by naloxone. It also produced analgesia in inflammatory pain (carrageenan) but not in a neuropathic pain model (tibial nerve transection). Inhibiting NTS POMC neurons does not produce any effect on basal nociception but inhibits stress-induced analgesia (unlike inhibition of arcuate POMC neurons). Activation of NTS POMC neuronal populations in conscious mice did not cause respiratory depression, anxiety or locomotor deficit (in open field) nor affective preference. These findings indicate that NTS POMC neurons play a key role in the generation of endorphinergic endogenous analgesia and can also regulate cardiorespiratory function.
Collapse
Affiliation(s)
- Pabitra Hriday Patra
- Anaesthesia, Pain & Critical Care Research, School of Physiology, Pharmacology and Neuroscience, University of Bristol, BS8 1TD, UK
| | - Becks Tench
- Anaesthesia, Pain & Critical Care Research, School of Physiology, Pharmacology and Neuroscience, University of Bristol, BS8 1TD, UK
| | - Timna Hitrec
- Anaesthesia, Pain & Critical Care Research, School of Physiology, Pharmacology and Neuroscience, University of Bristol, BS8 1TD, UK
| | - Fiona Holmes
- Anaesthesia, Pain & Critical Care Research, School of Physiology, Pharmacology and Neuroscience, University of Bristol, BS8 1TD, UK
| | - Robert Drake
- Anaesthesia, Pain & Critical Care Research, School of Physiology, Pharmacology and Neuroscience, University of Bristol, BS8 1TD, UK
| | - Serena Cerritelli
- Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - David Spanswick
- Neurosolutions, University of Warwick, Gibbet Hill Road, Coventry, West Midlands, CV4 7AL, UK
| | - Anthony Edward Pickering
- Anaesthesia, Pain & Critical Care Research, School of Physiology, Pharmacology and Neuroscience, University of Bristol, BS8 1TD, UK
| |
Collapse
|
18
|
Abstract
The fundamental commonality across pharmacotherapies for the epilepsies
is the modulation of neuronal excitability. This poses a clear
challenge—patterned neuronal excitation is essential to normal
function, thus disrupting this activity leads to side effects.
Moreover, the efficacy of current pharmacotherapy remains incomplete
despite decades of drug development. Approaches that allow for the
selective targeting of critical populations of cells and particular
pathways in the brain have the potential to both avoid side effects
and improve efficacy. Chemogenetic methods, which combine the
selective expression of designer receptors with designer drugs, have
rapidly grown in use in the neurosciences, including in epilepsy. This
review will briefly highlight the history of chemogenetics, their
applications to date in epilepsy, and the potential (and potential
hurdles to overcome) for future translation.
Collapse
Affiliation(s)
- Patrick A. Forcelli
- Department of Pharmacology & Physiology, Georgetown University, Washington, DC, USA
- Department of Neuroscience, Georgetown University, Washington, DC, USA
- Interdisciplinary Program in Neuroscience, Georgetown University, Washington, DC, USA
| |
Collapse
|
19
|
Huzard D, Martin M, Maingret F, Chemin J, Jeanneteau F, Mery PF, Fossat P, Bourinet E, François A. The impact of C-tactile low-threshold mechanoreceptors on affective touch and social interactions in mice. SCIENCE ADVANCES 2022; 8:eabo7566. [PMID: 35767616 PMCID: PMC9242590 DOI: 10.1126/sciadv.abo7566] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Affective touch is necessary for proper neurodevelopment and sociability. However, it remains unclear how the neurons innervating the skin detect affective and social behaviors. The C low-threshold mechanoreceptors (C-LTMRs), a specific population of somatosensory neurons in mice, appear particularly well suited, physiologically and anatomically, to perceive affective and social touch. However, their contribution to sociability has not been resolved yet. Our observations revealed that C-LTMR functional deficiency induced social isolation and reduced tactile interactions in adulthood. Conversely, transient increase in C-LTMR excitability in adults, using chemogenetics, was rewarding, promoted touch-seeking behaviors, and had prosocial influences on group dynamics. This work provides the first empirical evidence that specific peripheral inputs alone can drive complex social behaviors. It demonstrates the existence of a specialized neuronal circuit, originating in the skin, wired to promote interactions with other individuals.
Collapse
Affiliation(s)
- Damien Huzard
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Miquel Martin
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - François Maingret
- Institut des Maladies Neurodégénératives, Université de Bordeaux, CNRS, Bordeaux, France
| | - Jean Chemin
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Freddy Jeanneteau
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Pierre-François Mery
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Pascal Fossat
- Institut des Maladies Neurodégénératives, Université de Bordeaux, CNRS, Bordeaux, France
| | - Emmanuel Bourinet
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Amaury François
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, Montpellier, France
- Corresponding author.
| |
Collapse
|
20
|
Claes M, De Groef L, Moons L. The DREADDful Hurdles and Opportunities of the Chronic Chemogenetic Toolbox. Cells 2022; 11:1110. [PMID: 35406674 PMCID: PMC8998042 DOI: 10.3390/cells11071110] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/10/2022] [Accepted: 03/23/2022] [Indexed: 12/22/2022] Open
Abstract
The chronic character of chemogenetics has been put forward as one of the assets of the technique, particularly in comparison to optogenetics. Yet, the vast majority of chemogenetic studies have focused on acute applications, while repeated, long-term neuromodulation has only been booming in the past few years. Unfortunately, together with the rising number of studies, various hurdles have also been uncovered, especially in relation to its chronic application. It becomes increasingly clear that chronic neuromodulation warrants caution and that the effects of acute neuromodulation cannot be extrapolated towards chronic experiments. Deciphering the underlying cellular and molecular causes of these discrepancies could truly unlock the chronic chemogenetic toolbox and possibly even pave the way for chemogenetics towards clinical application. Indeed, we are only scratching the surface of what is possible with chemogenetic research. For example, most investigations are concentrated on behavioral read-outs, whereas dissecting the underlying molecular signature after (chronic) neuromodulation could reveal novel insights in terms of basic neuroscience and deregulated neural circuits. In this review, we highlight the hurdles associated with the use of chemogenetic experiments, as well as the unexplored research questions for which chemogenetics offers the ideal research platform, with a particular focus on its long-term application.
Collapse
Affiliation(s)
- Marie Claes
- Laboratory of Neural Circuit Development and Regeneration, Department of Biology, KU Leuven, 3000 Leuven, Belgium;
- Leuven Brain Institute, KU Leuven, 3000 Leuven, Belgium;
| | - Lies De Groef
- Leuven Brain Institute, KU Leuven, 3000 Leuven, Belgium;
- Laboratory of Cellular Communication and Neurodegeneration, Department of Biology, KU Leuven, 3000 Leuven, Belgium
| | - Lieve Moons
- Laboratory of Neural Circuit Development and Regeneration, Department of Biology, KU Leuven, 3000 Leuven, Belgium;
- Leuven Brain Institute, KU Leuven, 3000 Leuven, Belgium;
| |
Collapse
|
21
|
Dai H, Lou S, Zhang Y, Thanawala M, Huang K, Ji L, Carden S, Liao T, Abbassi M, Shu CJ, Lantermann A, Sadaghiani M, Blom D, Wagner J, Huang P. Transcriptional neural-like signaling contributes to an immune-suppressive tumor microenvironment. FASEB Bioadv 2022; 4:76-89. [PMID: 35024574 PMCID: PMC8728105 DOI: 10.1096/fba.2021-00076] [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] [Received: 06/29/2021] [Revised: 08/31/2021] [Accepted: 09/07/2021] [Indexed: 11/17/2022] Open
Abstract
Tumor innervation has recently been documented and characterized in various settings and tumor types. However, the role that nerves innervating tumors play in the pathogenesis of cancer has not been clarified. In this study, we searched for neural signaling from bulk RNA sequencing from The Cancer Genome Atlas (TCGA) dataset and looked for patterns of interactions between different cell types within the tumor environment. Using a presynapse signature (PSS) as a probe, we showed that multiple stromal cell types crosstalk and/or contribute to neural signals. Based on the correlation and linear regression, we hypothesized that neural signals contribute to an immune-suppressive tumor microenvironment (TME). To test this hypothesis, we performed in vitro dorsal root ganglion (DRG)/macrophage coculture experiments. Compared to the M2 macrophage monoculture, the DRG/M2 macrophage coculture prevented anti-inflammatory M2 to pro-inflammatory M1 polarization by LPS stimulation. Finally, a survey of different TCGA tumor types indicated that higher RNA neural signature is predictive of poor patient outcomes in multiple tumor types.
Collapse
Affiliation(s)
- Hongyue Dai
- Cygnal TherapeuticsCambridgeMassachusettsUSA
| | - Shan Lou
- Cygnal TherapeuticsCambridgeMassachusettsUSA
| | - Yanbo Zhang
- Cygnal TherapeuticsCambridgeMassachusettsUSA
| | | | | | - Lexiang Ji
- Cygnal TherapeuticsCambridgeMassachusettsUSA
| | | | | | | | | | | | | | - Daniel Blom
- Cygnal TherapeuticsCambridgeMassachusettsUSA
| | - John Wagner
- Cygnal TherapeuticsCambridgeMassachusettsUSA
| | - Pearl Huang
- Cygnal TherapeuticsCambridgeMassachusettsUSA
| |
Collapse
|
22
|
Defaye M, Abdullah NS, Iftinca M, Hassan A, Agosti F, Zhang Z, Cumenal M, Zamponi GW, Altier C. Gut-innervating TRPV1+ Neurons Drive Chronic Visceral Pain via Microglial P2Y12 Receptor. Cell Mol Gastroenterol Hepatol 2021; 13:977-999. [PMID: 34954381 PMCID: PMC8867057 DOI: 10.1016/j.jcmgh.2021.12.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 12/15/2021] [Accepted: 12/15/2021] [Indexed: 12/30/2022]
Abstract
BACKGROUND & AIMS Chronic abdominal pain is a common symptom of inflammatory bowel diseases (IBDs). Peripheral and central mechanisms contribute to the transition from acute to chronic pain during active disease and clinical remission. Lower mechanical threshold and hyperexcitability of visceral afferents induce gliosis in central pain circuits, leading to persistent visceral hypersensitivity (VHS). In the spinal cord, microglia, the immune sentinels of the central nervous system, undergo activation in multiple models of VHS. Here, we investigated the mechanisms of microglia activation to identify centrally acting analgesics for chronic IBD pain. METHODS Using Designer Receptors Exclusively Activated by Designer Drugs (DREADD) expressed in transient receptor potential vanilloid member 1-expressing visceral neurons that sense colonic inflammation, we tested whether neuronal activity was indispensable to control microglia activation and VHS. We then investigated the neuron-microglia signaling system involved in visceral pain chronification. RESULTS We found that chemogenetic inhibition of transient receptor potential vanilloid member 1+ visceral afferents prevents microglial activation in the spinal cord and subsequent VHS in colitis mice. In contrast, chemogenetic activation, in the absence of colitis, enhanced microglial activation associated with VHS. We identified a purinergic signaling mechanism mediated by neuronal adenosine triphosphate (ATP) and microglial P2Y12 receptor, triggering VHS in colitis. Inhibition of P2RY12 prevented microglial reactivity and chronic VHS post-colitis. CONCLUSIONS Overall, these data provide novel insights into the central mechanisms of chronic visceral pain and suggest that targeting microglial P2RY12 signaling could be harnessed to relieve pain in patients with IBD who are in remission.
Collapse
Affiliation(s)
- Manon Defaye
- Department of Physiology and Pharmacology, Calgary, Alberta, Canada,Inflammation Research Network-Snyder Institute for Chronic Diseases, Calgary, Alberta, Canada,Alberta Children's Hospital Research Institute, Cumming School of Medicine, Calgary, Alberta, Canada
| | - Nasser S. Abdullah
- Department of Physiology and Pharmacology, Calgary, Alberta, Canada,Inflammation Research Network-Snyder Institute for Chronic Diseases, Calgary, Alberta, Canada,Alberta Children's Hospital Research Institute, Cumming School of Medicine, Calgary, Alberta, Canada
| | - Mircea Iftinca
- Department of Physiology and Pharmacology, Calgary, Alberta, Canada,Inflammation Research Network-Snyder Institute for Chronic Diseases, Calgary, Alberta, Canada,Alberta Children's Hospital Research Institute, Cumming School of Medicine, Calgary, Alberta, Canada
| | - Ahmed Hassan
- Department of Physiology and Pharmacology, Calgary, Alberta, Canada,Inflammation Research Network-Snyder Institute for Chronic Diseases, Calgary, Alberta, Canada,Alberta Children's Hospital Research Institute, Cumming School of Medicine, Calgary, Alberta, Canada
| | - Francina Agosti
- Department of Physiology and Pharmacology, Calgary, Alberta, Canada,Inflammation Research Network-Snyder Institute for Chronic Diseases, Calgary, Alberta, Canada,Alberta Children's Hospital Research Institute, Cumming School of Medicine, Calgary, Alberta, Canada
| | - Zizhen Zhang
- Department of Physiology and Pharmacology, Calgary, Alberta, Canada,Alberta Children's Hospital Research Institute, Cumming School of Medicine, Calgary, Alberta, Canada,Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Melissa Cumenal
- Department of Physiology and Pharmacology, Calgary, Alberta, Canada,Inflammation Research Network-Snyder Institute for Chronic Diseases, Calgary, Alberta, Canada,Alberta Children's Hospital Research Institute, Cumming School of Medicine, Calgary, Alberta, Canada
| | - Gerald W. Zamponi
- Department of Physiology and Pharmacology, Calgary, Alberta, Canada,Alberta Children's Hospital Research Institute, Cumming School of Medicine, Calgary, Alberta, Canada,Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Christophe Altier
- Department of Physiology and Pharmacology, Calgary, Alberta, Canada,Inflammation Research Network-Snyder Institute for Chronic Diseases, Calgary, Alberta, Canada,Alberta Children's Hospital Research Institute, Cumming School of Medicine, Calgary, Alberta, Canada,Correspondence Address correspondence to: Christophe Altier, PhD, Associate Professor, Canada Research Chair in Inflammatory Pain, Department of Physiology & Pharmacology, Inflammation Research Network, Snyder Institute for Chronic Diseases, University of Calgary, HS 1665, 3330 Hospital Dr NW, Calgary, AB, T2N4N1 Canada. tel: (403) 220-7549.
| |
Collapse
|
23
|
Won J, Pankratov Y, Jang MW, Kim S, Ju YH, Lee S, Lee SE, Kim A, Park S, Lee CJ, Heo WD. Opto-vTrap, an optogenetic trap for reversible inhibition of vesicular release, synaptic transmission, and behavior. Neuron 2021; 110:423-435.e4. [PMID: 34852235 DOI: 10.1016/j.neuron.2021.11.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 09/01/2021] [Accepted: 11/03/2021] [Indexed: 01/13/2023]
Abstract
Spatiotemporal control of brain activity by optogenetics has emerged as an essential tool to study brain function. For silencing brain activity, optogenetic probes, such as halorhodopsin and archaerhodopsin, inhibit transmitter release indirectly by hyperpolarizing membrane potentials. However, these probes cause an undesirable ionic imbalance and rebound spikes. Moreover, they are not applicable to use in non-excitable glial cells. Here we engineered Opto-vTrap, a light-inducible and reversible inhibition system to temporarily trap the transmitter-containing vesicles from exocytotic release. Light activation of Opto-vTrap caused full vesicle clusterization and complete inhibition of exocytosis within 1 min, which recovered within 30 min after light off. We found a significant reduction in synaptic and gliotransmission upon activation of Opto-vTrap in acute brain slices. Opto-vTrap significantly inhibited hippocampus-dependent memory retrieval with full recovery within an hour. We propose Opto-vTrap as a next-generation optogenetic silencer to control brain activity and behavior with minimal confounding effects.
Collapse
Affiliation(s)
- Joungha Won
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon 34126, Republic of Korea; Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Yuriy Pankratov
- School of Life Sciences, University of Warwick, Coventry, CV4 7AL, UK
| | - Minwoo Wendy Jang
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon 34126, Republic of Korea; KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Sunpil Kim
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon 34126, Republic of Korea; KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Yeon Ha Ju
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon 34126, Republic of Korea
| | - Sangkyu Lee
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon 34126, Republic of Korea
| | - Seung Eun Lee
- Virus Facility, Research Animal Resource Center, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Arie Kim
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon 34126, Republic of Korea
| | - Soowon Park
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon 34126, Republic of Korea
| | - C Justin Lee
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon 34126, Republic of Korea; KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea.
| | - Won Do Heo
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon 34126, Republic of Korea; Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea; KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea.
| |
Collapse
|
24
|
Learning-induced plasticity in the barrel cortex is disrupted by inhibition of layer 4 somatostatin-containing interneurons. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1869:119146. [PMID: 34599984 DOI: 10.1016/j.bbamcr.2021.119146] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/29/2021] [Accepted: 09/13/2021] [Indexed: 12/26/2022]
Abstract
Gaba-ergic neurons are a diverse cell class with extensive influence over cortical processing, but their role in experience-dependent plasticity is not completely understood. Here we addressed the role of cortical somatostatin- (SOM-INs) and vasoactive intestinal polypeptide- (VIP-INs) containing interneurons in a Pavlovian conditioning where stimulation of the vibrissae is used as a conditioned stimulus and tail shock as unconditioned one. This procedure induces a plastic change observed as an enlargement of the cortical functional representation of vibrissae activated during conditioning. Using layer-targeted, cell-selective DREADD transductions, we examined the involvement of SOM-INs and VIP-INs activity in learning-related plastic changes. Under optical recordings, we injected DREADD-expressing vectors into layer IV (L4) barrels or layer II/III (L2/3) areas corresponding to the activated vibrissae. The activity of the interneurons was modulated during all conditioning sessions, and functional 2-deoxyglucose (2DG) maps were obtained 24 h after the last session. In mice with L4 but not L2/3 SOM-INs suppressed during conditioning, the plastic change of whisker representation was absent. The behavioral effect of conditioning was disturbed. Both L4 SOM-INs excitation and L2/3 VIP-INs inhibition during conditioning did not affect the plasticity or the conditioned response. We found the activity of L4 SOM-INs is indispensable in the formation of learning-induced plastic change. We propose that L4 SOM-INs may provide disinhibition by blocking L4 parvalbumin interneurons, allowing a flow of information into upper cortical layers during learning.
Collapse
|
25
|
Bony AR, McArthur JR, Finol-Urdaneta RK, Adams DJ. Analgesic α-conotoxins modulate native and recombinant GIRK1/2 channels via activation of GABA B receptors and reduce neuroexcitability. Br J Pharmacol 2021; 179:179-198. [PMID: 34599513 DOI: 10.1111/bph.15690] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 08/25/2021] [Accepted: 08/30/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND AND PURPOSE Activation of GIRK channels via G protein-coupled GABAB receptors has been shown to attenuate nociceptive transmission. The analgesic α-conotoxin Vc1.1 activates GABAB receptors resulting in inhibition of Cav 2.2 and Cav 2.3 channels in mammalian primary afferent neurons. Here, we investigated the effects of analgesic α-conotoxins on recombinant and native GIRK-mediated K+ currents and on neuronal excitability. EXPERIMENTAL APPROACH The effects of analgesic α-conotoxins, Vc1.1, RgIA, and PeIA, were investigated on inwardly-rectifying K+ currents in HEK293T cells recombinantly co-expressing either heteromeric human GIRK1/2 or homomeric GIRK2 subunits, with GABAB receptors. The effects of α-conotoxin Vc1.1 and baclofen were studied on GIRK-mediated K+ currents and the passive and active electrical properties of adult mouse dorsal root ganglion neurons. KEY RESULTS Analgesic α-conotoxins Vc1.1, RgIA, and PeIA potentiate inwardly-rectifying K+ currents in HEK293T cells recombinantly expressing human GIRK1/2 channels and GABAB receptors. GABAB receptor-dependent GIRK channel potentiation by Vc1.1 and baclofen occurs via a pertussis toxin-sensitive G protein and is inhibited by the selective GABAB receptor antagonist CGP 55845. In adult mouse dorsal root ganglion neurons, GABAB receptor-dependent GIRK channel potentiation by Vc1.1 and baclofen hyperpolarizes the cell membrane potential and reduces excitability. CONCLUSIONS AND IMPLICATIONS This is the first report of GIRK channel potentiation via allosteric α-conotoxin Vc1.1-GABAB receptor agonism, leading to decreased neuronal excitability. Such action potentially contributes to the analgesic effects of Vc1.1 and baclofen observed in vivo.
Collapse
Affiliation(s)
- Anuja R Bony
- Illawarra Health and Medical Research Institute (IHMRI), University of Wollongong, Wollongong, NSW, Australia
| | - Jeffrey R McArthur
- Illawarra Health and Medical Research Institute (IHMRI), University of Wollongong, Wollongong, NSW, Australia
| | - Rocio K Finol-Urdaneta
- Illawarra Health and Medical Research Institute (IHMRI), University of Wollongong, Wollongong, NSW, Australia
| | - David J Adams
- Illawarra Health and Medical Research Institute (IHMRI), University of Wollongong, Wollongong, NSW, Australia
| |
Collapse
|
26
|
Sternson SM, Bleakman D. Chemogenetics: drug-controlled gene therapies for neural circuit disorders. ACTA ACUST UNITED AC 2021; 6:1079-1094. [PMID: 34422319 PMCID: PMC8376173 DOI: 10.18609/cgti.2020.112] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Many patients with nervous system disorders have considerable unmet clinical needs or suffer debilitating drug side effects. A major limitation of exiting treatment approaches is that traditional small molecule pharmacotherapy lacks sufficient specificity to effectively treat many neurological diseases. Chemogenetics is a new gene therapy technology that targets an engineered receptor to cell types involved in nervous system dysfunction, enabling highly selective drug-controlled neuromodulation. Here, we discuss chemogenetic platforms and considerations for their potential application as human nervous system therapies.
Collapse
Affiliation(s)
- Scott M Sternson
- Janelia Research Campus, HHMI, 19700 Helix Dr. Ashburn, VA 20147, USA
| | - David Bleakman
- Redpin Therapeutics, 1329, Madison Avenue, Suite 125, New York, NY 10029, USA
| |
Collapse
|
27
|
Deemyad T, Puig S, Papale AE, Qi H, LaRocca GM, Aravind D, LaNoce E, Urban NN. Lateralized Decrease of Parvalbumin+ Cells in the Somatosensory Cortex of ASD Models Is Correlated with Unilateral Tactile Hypersensitivity. Cereb Cortex 2021; 32:554-568. [PMID: 34347040 DOI: 10.1093/cercor/bhab233] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 05/31/2021] [Accepted: 06/21/2021] [Indexed: 12/27/2022] Open
Abstract
Inhibitory control of excitatory networks contributes to cortical functions. Increasing evidence indicates that parvalbumin (PV+)-expressing basket cells (BCs) are a major player in maintaining the balance between excitation (E) and inhibition (I). Disruption of E/I balance in cortical networks is believed to be a hallmark of autism spectrum disorder (ASD). Here, we report a lateralized decrease in the number of PV+ BCs in L2/3 of the somatosensory cortex in the dominant hemisphere of Shank3-/- and Cntnap2-/- mouse models of ASD. The dominant hemisphere was identified during a reaching task to establish each animal's dominant forepaw. Double labeling with anti-PV antibody and a biotinylated lectin (Vicia villosa lectin [VVA]) showed that the number of BCs was not different but rather, some BCs did not express PV (PV-), resulting in an elevated number of PV- VVA+ BCs. Finally, we showed that dominant hindpaws had higher mechanical sensitivity when compared with the other hindpaws. This mechanical hypersensitivity in the dominant paw strongly correlated with the decrease in the number of PV+ interneurons and reduced PV expression in the corresponding cortex. Together, these results suggest that the hypersensitivity in ASD patients could be due to decreased inhibitory inputs to the dominant somatosensory cortex.
Collapse
Affiliation(s)
- Tara Deemyad
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA 15213, USA.,Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Stephanie Puig
- Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Andrew E Papale
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Hang Qi
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Gregory M LaRocca
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Deepthi Aravind
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Emma LaNoce
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Nathaniel N Urban
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA 15213, USA.,Center for Neuroscience at the University of Pittsburgh, University of Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
28
|
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: 75] [Impact Index Per Article: 18.8] [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
|
29
|
Abstract
Preclinical evidence has highlighted the importance of the μ-opioid peptide (MOP) receptor on primary afferents for both the analgesic actions of MOP receptor agonists, as well as the development of tolerance, if not opioid-induced hyperalgesia. There is also growing interest in targeting other opioid peptide receptor subtypes (δ-opioid peptide [DOP], κ-opioid peptide [KOP], and nociceptin/orphanin-FQ opioid peptide [NOP]) on primary afferents, as alternatives to MOP receptors, which may not be associated with as many deleterious side effects. Nevertheless, results from several recent studies of human sensory neurons indicate that although there are many similarities between rodent and human sensory neurons, there may also be important differences. Thus, the purpose of this study was to assess the distribution of opioid receptor subtypes among human sensory neurons. A combination of pharmacology, patch-clamp electrophysiology, Ca imaging, and single-cell semiquantitative polymerase chain reaction was used. Our results suggest that functional MOP-like receptors are present in approximately 50% of human dorsal root ganglion neurons. δ-opioid peptide-like receptors were detected in a subpopulation largely overlapping that with MOP-like receptors. Furthermore, KOP-like and NOP-like receptors are detected in a large proportion (44% and 40%, respectively) of human dorsal root ganglion neurons with KOP receptors also overlapping with MOP receptors at a high rate (83%). Our data confirm that all 4 opioid receptor subtypes are present and functional in human sensory neurons, where the overlap of DOP, KOP, and NOP receptors with MOP receptors suggests that activation of these other opioid receptor subtypes may also have analgesic efficacy.
Collapse
|
30
|
Boehm MA, Bonaventura J, Gomez JL, Solís O, Stein EA, Bradberry CW, Michaelides M. Translational PET applications for brain circuit mapping with transgenic neuromodulation tools. Pharmacol Biochem Behav 2021; 204:173147. [PMID: 33549570 PMCID: PMC8297666 DOI: 10.1016/j.pbb.2021.173147] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 01/29/2021] [Accepted: 02/01/2021] [Indexed: 02/08/2023]
Abstract
Transgenic neuromodulation tools have transformed the field of neuroscience over the past two decades by enabling targeted manipulation of neuronal populations and circuits with unprecedented specificity. Chemogenetic and optogenetic neuromodulation systems are among the most widely used and allow targeted control of neuronal activity through the administration of a selective compound or light, respectively. Innovative genetic targeting strategies are utilized to transduce specific cells to express transgenic receptors and opsins capable of manipulating neuronal activity. These allow mapping of neuroanatomical projection sites and link cellular manipulations with brain circuit functions and behavior. As these tools continue to expand knowledge of the nervous system in preclinical models, developing translational applications for human therapies is becoming increasingly possible. However, new strategies for implementing and monitoring transgenic tools are needed for safe and effective use in translational research and potential clinical applications. A major challenge for such applications is the need to track the location and function of chemogenetic receptors and opsins in vivo, and new developments in positron emission tomography (PET) imaging techniques offer promising solutions. The goal of this review is to summarize current research combining transgenic tools with PET for in vivo mapping and manipulation of brain circuits and to propose future directions for translational applications.
Collapse
Affiliation(s)
- Matthew A Boehm
- National Institute on Drug Abuse Intramural Research Program, 251 Bayview Blvd, Baltimore, MD 21224, United States; Department of Neuroscience, Brown University, Providence, RI 02906, United States.
| | - Jordi Bonaventura
- National Institute on Drug Abuse Intramural Research Program, 251 Bayview Blvd, Baltimore, MD 21224, United States.
| | - Juan L Gomez
- National Institute on Drug Abuse Intramural Research Program, 251 Bayview Blvd, Baltimore, MD 21224, United States.
| | - Oscar Solís
- National Institute on Drug Abuse Intramural Research Program, 251 Bayview Blvd, Baltimore, MD 21224, United States.
| | - Elliot A Stein
- National Institute on Drug Abuse Intramural Research Program, 251 Bayview Blvd, Baltimore, MD 21224, United States.
| | - Charles W Bradberry
- National Institute on Drug Abuse Intramural Research Program, 251 Bayview Blvd, Baltimore, MD 21224, United States.
| | - Michael Michaelides
- National Institute on Drug Abuse Intramural Research Program, 251 Bayview Blvd, Baltimore, MD 21224, United States; Department of Psychiatry & Behavioral Sciences, Johns Hopkins Medicine, Baltimore, MD, 21205, United States.
| |
Collapse
|
31
|
Poth KM, Texakalidis P, Boulis NM. Chemogenetics: Beyond Lesions and Electrodes. Neurosurgery 2021; 89:185-195. [PMID: 33913505 PMCID: PMC8279839 DOI: 10.1093/neuros/nyab147] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 02/26/2021] [Indexed: 01/14/2023] Open
Abstract
The field of chemogenetics has rapidly expanded over the last decade, and engineered receptors are currently utilized in the lab to better understand molecular interactions in the nervous system. We propose that chemogenetic receptors can be used for far more than investigational purposes. The potential benefit of adding chemogenetic neuromodulation to the current neurosurgical toolkit is substantial. There are several conditions currently treated surgically, electrically, and pharmacologically in clinic, and this review highlights how chemogenetic neuromodulation could improve patient outcomes over current neurosurgical techniques. We aim to emphasize the need to take these techniques from bench to bedside.
Collapse
Affiliation(s)
- Kelly M Poth
- Department of Neurosurgery, Emory University, Atlanta, Georgia, USA
| | | | | |
Collapse
|
32
|
Gantz SC, Ortiz MM, Belilos AJ, Moussawi K. Excitation of medium spiny neurons by 'inhibitory' ultrapotent chemogenetics via shifts in chloride reversal potential. eLife 2021; 10:64241. [PMID: 33822716 PMCID: PMC8024007 DOI: 10.7554/elife.64241] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 03/28/2021] [Indexed: 11/13/2022] Open
Abstract
Ultrapotent chemogenetics, including the chloride-permeable inhibitory PSAM4-GlyR receptor, were recently proposed as a powerful strategy to selectively control neuronal activity in awake, behaving animals. We aimed to validate the inhibitory function of PSAM4-GlyR in dopamine D1 receptor-expressing medium spiny neurons (D1-MSNs) in the ventral striatum. Activation of PSAM4-GlyR with the uPSEM792 ligand enhanced rather than suppressed the activity of D1-MSNs in vivo as indicated by increased c-fos expression in D1-MSNs and in vitro as indicated by cell-attached recordings from D1-MSNs in mouse brain slices. Whole-cell recordings showed that activation of PSAM4-GlyR depolarized D1-MSNs, attenuated GABAergic inhibition, and shifted the reversal potential of PSAM4-GlyR current to more depolarized potentials, perpetuating the depolarizing effect of receptor activation. These data show that 'inhibitory' PSAM4-GlyR chemogenetics may activate certain cell types and highlight the pitfalls of utilizing chloride conductances to inhibit neurons.
Collapse
Affiliation(s)
- Stephanie C Gantz
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, United States
| | - Maria M Ortiz
- Biological and Biomedical Neuroscience Program, University of North Carolina, Chapel Hill, United States
| | | | - Khaled Moussawi
- National Institute on Drug Abuse, Baltimore, United States.,Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, United States
| |
Collapse
|
33
|
Meister J, Wang L, Pydi SP, Wess J. Chemogenetic approaches to identify metabolically important GPCR signaling pathways: Therapeutic implications. J Neurochem 2021; 158:603-620. [PMID: 33540469 DOI: 10.1111/jnc.15314] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 01/21/2021] [Accepted: 01/29/2021] [Indexed: 12/16/2022]
Abstract
DREADDs (Designer Receptors Exclusively Activated by a Designer Drug) are designer G protein-coupled receptors (GPCRs) that are widely used in the neuroscience field to modulate neuronal activity. In this review, we will focus on DREADD studies carried out with genetically engineered mice aimed at elucidating signaling pathways important for maintaining proper glucose and energy homeostasis. The availability of muscarinic receptor-based DREADDs endowed with selectivity for one of the four major classes of heterotrimeric G proteins (Gs , Gi , Gq , and G12 ) has been instrumental in dissecting the physiological and pathophysiological roles of distinct G protein signaling pathways in metabolically important cell types. The novel insights gained from this work should inform the development of novel classes of drugs useful for the treatment of several metabolic disorders including type 2 diabetes and obesity.
Collapse
Affiliation(s)
- Jaroslawna Meister
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA
| | - Lei Wang
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA
| | - Sai P Pydi
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA
| | - Jürgen Wess
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA
| |
Collapse
|
34
|
Saleh M, Sharma K, Kalsi R, Fusco J, Sehrawat A, Saloman JL, Guo P, Zhang T, Mohamed N, Wang Y, Prasadan K, Gittes GK. Chemical pancreatectomy treats chronic pancreatitis while preserving endocrine function in preclinical models. J Clin Invest 2021; 131:143301. [PMID: 33351784 PMCID: PMC7843231 DOI: 10.1172/jci143301] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 12/01/2020] [Indexed: 12/20/2022] Open
Abstract
Chronic pancreatitis affects over 250,000 people in the US and millions worldwide. It is associated with chronic debilitating pain, pancreatic exocrine failure, and high risk of pancreatic cancer and usually progresses to diabetes. Treatment options are limited and ineffective. We developed a new potential therapy, wherein a pancreatic ductal infusion of 1%-2% acetic acid in mice and nonhuman primates resulted in a nonregenerative, near-complete ablation of the exocrine pancreas, with complete preservation of the islets. Pancreatic ductal infusion of acetic acid in a mouse model of chronic pancreatitis led to resolution of chronic inflammation and pancreatitis-associated pain. Furthermore, acetic acid-treated animals showed improved glucose tolerance and insulin secretion. The loss of exocrine tissue in this procedure would not typically require further management in patients with chronic pancreatitis because they usually have pancreatic exocrine failure requiring dietary enzyme supplements. Thus, this procedure, which should be readily translatable to humans through an endoscopic retrograde cholangiopancreatography (ERCP), may offer a potential innovative nonsurgical therapy for chronic pancreatitis that relieves pain and prevents the progression of pancreatic diabetes.
Collapse
Affiliation(s)
- Mohamed Saleh
- Division of Pediatric Surgery
- Division of Pediatric Endocrinology, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | | | | | | | - Jami L. Saloman
- Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, Department of Neurobiology, Pittsburgh Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Ping Guo
- Department of Clinical Science, Colorado State University, Fort Collins, Colorado, USA
| | | | | | | | | | | |
Collapse
|
35
|
Yi MH, Liu YU, Liu K, Chen T, Bosco DB, Zheng J, Xie M, Zhou L, Qu W, Wu LJ. Chemogenetic manipulation of microglia inhibits neuroinflammation and neuropathic pain in mice. Brain Behav Immun 2021; 92:78-89. [PMID: 33221486 PMCID: PMC7897256 DOI: 10.1016/j.bbi.2020.11.030] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 11/18/2020] [Accepted: 11/18/2020] [Indexed: 12/21/2022] Open
Abstract
Microglia play an important role in the central sensitization and chronic pain. However, a direct connection between microglial function and pain development in vivo remains incompletely understood. To address this issue, we applied chemogenetic approach by using CX3CR1creER/+:R26LSL-hM4Di/+ transgenic mice to enable expression of inhibitory Designer Receptors Exclusively Activated by Designer Drugs (Gi DREADD) in microglia. We found that microglial Gi DREADD activation inhibited spinal nerve transection (SNT)-induced microglial reactivity as well as chronic pain in both male and female mice. Gi DREADD activation downregulated the transcription factor interferon regulatory factor 8 (IRF8) and its downstream target pro-inflammatory cytokine interleukin 1 beta (IL-1β). Using in vivo spinal cord recording, we found that activation of microglial Gi DREADD attenuated synaptic transmission following SNT. Our results demonstrate that microglial Gi DREADD reduces neuroinflammation, synaptic function and neuropathic pain after SNT. Thus, chemogenetic approaches provide a potential opportunity for interrogating microglial function and neuropathic pain treatment.
Collapse
Affiliation(s)
- Min-Hee Yi
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Yong U. Liu
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Kevin Liu
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA,Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, 08854 USA
| | - Tingjun Chen
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Dale B. Bosco
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Jiaying Zheng
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Manling Xie
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Lijun Zhou
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Wenchun Qu
- Department of Pain Medicine, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA; Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA; Departments of Immunology, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
36
|
Sheahan TD, Warwick CA, Fanien LG, Ross SE. The Neurokinin-1 Receptor is Expressed with Gastrin-Releasing Peptide Receptor in Spinal Interneurons and Modulates Itch. J Neurosci 2020; 40:8816-8830. [PMID: 33051347 PMCID: PMC7659450 DOI: 10.1523/jneurosci.1832-20.2020] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/25/2020] [Accepted: 09/21/2020] [Indexed: 12/22/2022] Open
Abstract
The neurokinin-1 receptor (NK1R; encoded by Tacr1) is expressed in spinal dorsal horn neurons and has been suggested to mediate itch in rodents. However, previous studies relied heavily on neurotoxic ablation of NK1R spinal neurons, which limited further dissection of their function in spinal itch circuitry. To address this limitation, we leveraged a newly developed Tacr1CreER mouse line to characterize the role of NK1R spinal neurons in itch. We show that pharmacological activation of spinal NK1R and chemogenetic activation of Tacr1CreER spinal neurons increases itch behavior in male and female mice, whereas pharmacological inhibition of spinal NK1R suppresses itch behavior. We use fluorescence in situ hybridization (FISH) to characterize the endogenous expression of Tacr1 throughout the superficial and deeper dorsal horn (DDH), as well as the lateral spinal nucleus (LSN), of mouse and human spinal cord. Retrograde labeling studies in mice from the parabrachial nucleus (PBN) show that less than 20% of superficial Tacr1CreER dorsal horn neurons are spinal projection neurons, and thus the majority of Tacr1CreER are local interneurons. We then use a combination of in situ hybridization and ex vivo two-photon Ca2+ imaging of the mouse spinal cord to establish that NK1R and the gastrin-releasing peptide receptor (GRPR) are coexpressed within a subpopulation of excitatory superficial dorsal horn (SDH) neurons. These findings are the first to suggest a role for NK1R interneurons in itch and extend our understanding of the complexities of spinal itch circuitry.SIGNIFICANCE STATEMENT The spinal cord is a critical hub for processing somatosensory input, yet which spinal neurons process itch input and how itch signals are encoded within the spinal cord is not fully understood. We demonstrate neurokinin-1 receptor (NK1R) spinal neurons mediate itch behavior in mice and that the majority of NK1R spinal neurons are local interneurons. These NK1R neurons comprise a subset of gastrin-releasing peptide receptor (GRPR) interneurons and are thus positioned at the center of spinal itch transmission. We show NK1R mRNA expression in human spinal cord, underscoring the translational relevance of our findings in mice. This work is the first to suggest a role for NK1R interneurons in itch and extends our understanding of the complexities of spinal itch circuitry.
Collapse
Affiliation(s)
- Tayler D Sheahan
- Pittsburgh Center for Pain Research and Department of Neurobiology, University of Pittsburgh, Pittsburgh 15213, Pennsylvania
| | - Charles A Warwick
- Pittsburgh Center for Pain Research and Department of Neurobiology, University of Pittsburgh, Pittsburgh 15213, Pennsylvania
| | - Louis G Fanien
- Pittsburgh Center for Pain Research and Department of Neurobiology, University of Pittsburgh, Pittsburgh 15213, Pennsylvania
| | - Sarah E Ross
- Pittsburgh Center for Pain Research and Department of Neurobiology, University of Pittsburgh, Pittsburgh 15213, Pennsylvania
| |
Collapse
|
37
|
Echagarruga CT, Gheres KW, Norwood JN, Drew PJ. nNOS-expressing interneurons control basal and behaviorally evoked arterial dilation in somatosensory cortex of mice. eLife 2020; 9:e60533. [PMID: 33016877 PMCID: PMC7556878 DOI: 10.7554/elife.60533] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 10/02/2020] [Indexed: 12/19/2022] Open
Abstract
Cortical neural activity is coupled to local arterial diameter and blood flow. However, which neurons control the dynamics of cerebral arteries is not well understood. We dissected the cellular mechanisms controlling the basal diameter and evoked dilation in cortical arteries in awake, head-fixed mice. Locomotion drove robust arterial dilation, increases in gamma band power in the local field potential (LFP), and increases calcium signals in pyramidal and neuronal nitric oxide synthase (nNOS)-expressing neurons. Chemogenetic or pharmocological modulation of overall neural activity up or down caused corresponding increases or decreases in basal arterial diameter. Modulation of pyramidal neuron activity alone had little effect on basal or evoked arterial dilation, despite pronounced changes in the LFP. Modulation of the activity of nNOS-expressing neurons drove changes in the basal and evoked arterial diameter without corresponding changes in population neural activity.
Collapse
Affiliation(s)
| | - Kyle W Gheres
- Molecular, Cellular, and Integrative Biology Graduate Program, Pennsylvania State UniversityUniversity ParkUnited States
| | - Jordan N Norwood
- Cell and Developmental Biology Graduate Program, Pennsylvania State UniversityUniversity ParkUnited States
| | - Patrick J Drew
- Bioengineering Graduate Program, Pennsylvania State UniversityUniversity ParkUnited States
- Molecular, Cellular, and Integrative Biology Graduate Program, Pennsylvania State UniversityUniversity ParkUnited States
- Cell and Developmental Biology Graduate Program, Pennsylvania State UniversityUniversity ParkUnited States
- Departments of Engineering Science and Mechanics, Biomedical Engineering, and Neurosurgery, Pennsylvania State UniversityUniversity ParkUnited States
| |
Collapse
|
38
|
Hessmann E, Buchholz SM, Demir IE, Singh SK, Gress TM, Ellenrieder V, Neesse A. Microenvironmental Determinants of Pancreatic Cancer. Physiol Rev 2020; 100:1707-1751. [DOI: 10.1152/physrev.00042.2019] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) belongs to the most lethal solid tumors in humans. A histological hallmark feature of PDAC is the pronounced tumor microenvironment (TME) that dynamically evolves during tumor progression. The TME consists of different non-neoplastic cells such as cancer-associated fibroblasts, immune cells, endothelial cells, and neurons. Furthermore, abundant extracellular matrix components such as collagen and hyaluronic acid as well as matricellular proteins create a highly dynamic and hypovascular TME with multiple biochemical and physical interactions among the various cellular and acellular components that promote tumor progression and therapeutic resistance. In recent years, intensive research efforts have resulted in a significantly improved understanding of the biology and pathophysiology of the TME in PDAC, and novel stroma-targeted approaches are emerging that may help to improve the devastating prognosis of PDAC patients. However, none of anti-stromal therapies has been approved in patients so far, and there is still a large discrepancy between multiple successful preclinical results and subsequent failure in clinical trials. Furthermore, recent findings suggest that parts of the TME may also possess tumor-restraining properties rendering tailored therapies even more challenging.
Collapse
Affiliation(s)
- Elisabeth Hessmann
- Department of Gastroenterology, Gastrointestinal Oncology, and Endocrinology, University Medical Centre Goettingen, Georg August University, Goettingen, Germany; Department of Surgery, Klinikum rechts der Isar, Technische Universität München, School of Medicine Munich, Munich, Germany; Sonderforschungsbereich/Collaborative Research Centre 1321 Modeling and Targeting Pancreatic Cancer, Munich, Germany; Deutsches Konsortium für Translationale Krebsforschung (DKTK) Munich Site, Munich, Germany; and
| | - Soeren M. Buchholz
- Department of Gastroenterology, Gastrointestinal Oncology, and Endocrinology, University Medical Centre Goettingen, Georg August University, Goettingen, Germany; Department of Surgery, Klinikum rechts der Isar, Technische Universität München, School of Medicine Munich, Munich, Germany; Sonderforschungsbereich/Collaborative Research Centre 1321 Modeling and Targeting Pancreatic Cancer, Munich, Germany; Deutsches Konsortium für Translationale Krebsforschung (DKTK) Munich Site, Munich, Germany; and
| | - Ihsan Ekin Demir
- Department of Gastroenterology, Gastrointestinal Oncology, and Endocrinology, University Medical Centre Goettingen, Georg August University, Goettingen, Germany; Department of Surgery, Klinikum rechts der Isar, Technische Universität München, School of Medicine Munich, Munich, Germany; Sonderforschungsbereich/Collaborative Research Centre 1321 Modeling and Targeting Pancreatic Cancer, Munich, Germany; Deutsches Konsortium für Translationale Krebsforschung (DKTK) Munich Site, Munich, Germany; and
| | - Shiv K. Singh
- Department of Gastroenterology, Gastrointestinal Oncology, and Endocrinology, University Medical Centre Goettingen, Georg August University, Goettingen, Germany; Department of Surgery, Klinikum rechts der Isar, Technische Universität München, School of Medicine Munich, Munich, Germany; Sonderforschungsbereich/Collaborative Research Centre 1321 Modeling and Targeting Pancreatic Cancer, Munich, Germany; Deutsches Konsortium für Translationale Krebsforschung (DKTK) Munich Site, Munich, Germany; and
| | - Thomas M. Gress
- Department of Gastroenterology, Gastrointestinal Oncology, and Endocrinology, University Medical Centre Goettingen, Georg August University, Goettingen, Germany; Department of Surgery, Klinikum rechts der Isar, Technische Universität München, School of Medicine Munich, Munich, Germany; Sonderforschungsbereich/Collaborative Research Centre 1321 Modeling and Targeting Pancreatic Cancer, Munich, Germany; Deutsches Konsortium für Translationale Krebsforschung (DKTK) Munich Site, Munich, Germany; and
| | - Volker Ellenrieder
- Department of Gastroenterology, Gastrointestinal Oncology, and Endocrinology, University Medical Centre Goettingen, Georg August University, Goettingen, Germany; Department of Surgery, Klinikum rechts der Isar, Technische Universität München, School of Medicine Munich, Munich, Germany; Sonderforschungsbereich/Collaborative Research Centre 1321 Modeling and Targeting Pancreatic Cancer, Munich, Germany; Deutsches Konsortium für Translationale Krebsforschung (DKTK) Munich Site, Munich, Germany; and
| | - Albrecht Neesse
- Department of Gastroenterology, Gastrointestinal Oncology, and Endocrinology, University Medical Centre Goettingen, Georg August University, Goettingen, Germany; Department of Surgery, Klinikum rechts der Isar, Technische Universität München, School of Medicine Munich, Munich, Germany; Sonderforschungsbereich/Collaborative Research Centre 1321 Modeling and Targeting Pancreatic Cancer, Munich, Germany; Deutsches Konsortium für Translationale Krebsforschung (DKTK) Munich Site, Munich, Germany; and
| |
Collapse
|
39
|
Goutaudier R, Coizet V, Carcenac C, Carnicella S. Compound 21, a two-edged sword with both DREADD-selective and off-target outcomes in rats. PLoS One 2020; 15:e0238156. [PMID: 32946510 PMCID: PMC7500623 DOI: 10.1371/journal.pone.0238156] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 08/10/2020] [Indexed: 01/05/2023] Open
Abstract
Designer Receptors Exclusively Activated by Designer Drugs (DREADDs) represent a technical revolution in integrative neuroscience. However, the first used ligands exhibited dose-dependent selectivity for their molecular target, leading to potential unspecific effects. Compound 21 (C21) was recently proposed as an alternative, but in vivo characterization of its properties is not sufficient yet. Here, we evaluated its potency to selectively modulate the activity of nigral dopaminergic (DA) neurons through the canonical DREADD receptor hM4Di using TH-Cre rats. In males, 1 mg.kg-1 of C21 strongly increased nigral neurons activity in control animals, indicative of a significant off-target effect. Reducing the dose to 0.5 mg.kg-1 circumvented this unspecific effect, while activated the inhibitory DREADDs and selectively reduced nigral neurons firing. In females, 0.5 mg.kg-1 of C21 induced a transient and residual off-target effect that may mitigated the inhibitory DREADDs-mediated effect. This study raises up the necessity to test selectivity and efficacy of chosen ligands for each new experimental condition.
Collapse
Affiliation(s)
- Raphaël Goutaudier
- Institut national de la santé et de la recherche médicale, Grenoble Institut des Neurosciences, U1216, Université, Grenoble Alpes, Grenoble, France
| | - Véronique Coizet
- Institut national de la santé et de la recherche médicale, Grenoble Institut des Neurosciences, U1216, Université, Grenoble Alpes, Grenoble, France
| | - Carole Carcenac
- Institut national de la santé et de la recherche médicale, Grenoble Institut des Neurosciences, U1216, Université, Grenoble Alpes, Grenoble, France
| | - Sebastien Carnicella
- Institut national de la santé et de la recherche médicale, Grenoble Institut des Neurosciences, U1216, Université, Grenoble Alpes, Grenoble, France
| |
Collapse
|
40
|
Chakrabarti S, Pattison LA, Doleschall B, Rickman RH, Blake H, Callejo G, Heppenstall PA, Smith ESJ. Intraarticular Adeno-Associated Virus Serotype AAV-PHP.S-Mediated Chemogenetic Targeting of Knee-Innervating Dorsal Root Ganglion Neurons Alleviates Inflammatory Pain in Mice. Arthritis Rheumatol 2020; 72:1749-1758. [PMID: 32418284 DOI: 10.1002/art.41314] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 05/12/2020] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Joint pain is the major clinical symptom of arthritis that affects millions of people. Controlling the excitability of knee-innervating dorsal root ganglion (DRG) neurons (knee neurons) could potentially provide pain relief. We undertook this study to evaluate whether the newly engineered adeno-associated virus (AAV) serotype, AAV-PHP.S, can deliver functional artificial receptors to control knee neuron excitability following intraarticular knee injection. METHODS The AAV-PHP.S virus, packaged with dTomato fluorescent protein and either excitatory (Gq ) or inhibitory (Gi ) designer receptors exclusively activated by designer drugs (DREADDs), was injected into the knee joints of adult mice. Labeling of DRG neurons with AAV-PHP.S from the knee was evaluated using immunohistochemistry. The functionality of Gq - and Gi -DREADDs was evaluated using whole-cell patch clamp electrophysiology on acutely cultured DRG neurons. Pain behavior in mice was assessed using a digging assay, dynamic weight bearing, and rotarod performance, before and after intraperitoneal administration of the DREADD activator, Compound 21. RESULTS We showed that AAV-PHP.S can deliver functional genes into ~7% of lumbar DRG neurons when injected into the knee joint in a similar manner to the well-established retrograde tracer, fast blue. Short-term activation of AAV-PHP.S-delivered Gq -DREADD increased excitability of knee neurons in vitro (P = 0.02 by unpaired t-test), without inducing overt pain in mice when activated in vivo. By contrast, in vivo Gi -DREADD activation alleviated digging deficits induced by Freund's complete adjuvant-mediated knee inflammation (P = 0.0002 by repeated-measures analysis of variance [ANOVA] followed by Holm-Sidak multiple comparisons test). A concomitant decrease in knee neuron excitability was observed in vitro (P = 0.005 by ANOVA followed by Holm-Sidak multiple comparisons test). CONCLUSION We describe an AAV-mediated chemogenetic approach to specifically control joint pain, which may be utilized in translational arthritic pain research.
Collapse
|
41
|
Chemogenetic Suppression of GnRH Neurons during Pubertal Development Can Alter Adult GnRH Neuron Firing Rate and Reproductive Parameters in Female Mice. eNeuro 2020; 7:ENEURO.0223-20.2020. [PMID: 32513661 PMCID: PMC7363480 DOI: 10.1523/eneuro.0223-20.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 05/28/2020] [Indexed: 12/02/2022] Open
Abstract
Gonadotropin-releasing hormone (GnRH) neurons control anterior pituitary, and thereby gonadal, function. GnRH neurons are active before outward indicators of puberty appear. Prenatal androgen (PNA) exposure mimics reproductive dysfunction of the common fertility disorder polycystic ovary syndrome (PCOS) and reduces prepubertal GnRH neuron activity. Early neuron activity can play a critical role in establishing circuitry and adult function. We tested the hypothesis that changing prepubertal GnRH neuron activity programs adult GnRH neuron activity and reproduction independent of androgen exposure in female mice. Activating (3Dq) or inhibitory (4Di) designer receptors exclusively activated by designer drugs (DREADDs) were targeted to GnRH neurons using Cre-lox technology. In control studies, the DREADD ligand clozapine n-oxide (CNO) produced the expected changes in GnRH neuron activity in vitro and luteinizing hormone (LH) release in vivo. CNO was administered to control or PNA mice between two and three weeks of age, when GnRH neuron firing rate is reduced in PNA mice. In controls, reducing prepubertal GnRH neuron activity with 4Di increased adult GnRH neuron firing rate and days in diestrus but did not change puberty onset or GABA transmission to these cells. In contrast, activating GnRH neurons had no effect on reproductive parameters or firing rate and did not rescue reproductive phenotypes in PNA mice. These studies support the hypothesis that prepubertal neuronal activity sculpts elements of the adult reproductive neuroendocrine axis and cyclicity but indicate that other PNA-induced programming actions are required for full reproductive phenotypes and/or that compensatory mechanisms overcome activity-mediated changes to mitigate reproductive changes in adults.
Collapse
|
42
|
Kuner R, Kuner T. Cellular Circuits in the Brain and Their Modulation in Acute and Chronic Pain. Physiol Rev 2020; 101:213-258. [PMID: 32525759 DOI: 10.1152/physrev.00040.2019] [Citation(s) in RCA: 172] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Chronic, pathological pain remains a global health problem and a challenge to basic and clinical sciences. A major obstacle to preventing, treating, or reverting chronic pain has been that the nature of neural circuits underlying the diverse components of the complex, multidimensional experience of pain is not well understood. Moreover, chronic pain involves diverse maladaptive plasticity processes, which have not been decoded mechanistically in terms of involvement of specific circuits and cause-effect relationships. This review aims to discuss recent advances in our understanding of circuit connectivity in the mammalian brain at the level of regional contributions and specific cell types in acute and chronic pain. A major focus is placed on functional dissection of sub-neocortical brain circuits using optogenetics, chemogenetics, and imaging technological tools in rodent models with a view towards decoding sensory, affective, and motivational-cognitive dimensions of pain. The review summarizes recent breakthroughs and insights on structure-function properties in nociceptive circuits and higher order sub-neocortical modulatory circuits involved in aversion, learning, reward, and mood and their modulation by endogenous GABAergic inhibition, noradrenergic, cholinergic, dopaminergic, serotonergic, and peptidergic pathways. The knowledge of neural circuits and their dynamic regulation via functional and structural plasticity will be beneficial towards designing and improving targeted therapies.
Collapse
Affiliation(s)
- Rohini Kuner
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany; and Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Thomas Kuner
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany; and Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
43
|
Kelly MP, Heckman PRA, Havekes R. Genetic manipulation of cyclic nucleotide signaling during hippocampal neuroplasticity and memory formation. Prog Neurobiol 2020; 190:101799. [PMID: 32360536 DOI: 10.1016/j.pneurobio.2020.101799] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/14/2020] [Accepted: 03/26/2020] [Indexed: 12/12/2022]
Abstract
Decades of research have underscored the importance of cyclic nucleotide signaling in memory formation and synaptic plasticity. In recent years, several new genetic techniques have expanded the neuroscience toolbox, allowing researchers to measure and modulate cyclic nucleotide gradients with high spatiotemporal resolution. Here, we will provide an overview of studies using genetic approaches to interrogate the role cyclic nucleotide signaling plays in hippocampus-dependent memory processes and synaptic plasticity. Particular attention is given to genetic techniques that measure real-time changes in cyclic nucleotide levels as well as newly-developed genetic strategies to transiently manipulate cyclic nucleotide signaling in a subcellular compartment-specific manner with high temporal resolution.
Collapse
Affiliation(s)
- Michy P Kelly
- Department of Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, 6439 Garners Ferry Rd, VA Bldg1, 3(rd) Fl, D-12, Columbia, 29209, SC, USA.
| | - Pim R A Heckman
- Neurobiology Expertise Group, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Nijenborgh 7, 9747 AG Groningen, the Netherlands.
| | - Robbert Havekes
- Neurobiology Expertise Group, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Nijenborgh 7, 9747 AG Groningen, the Netherlands.
| |
Collapse
|
44
|
DREADDs: The Power of the Lock, the Weakness of the Key. Favoring the Pursuit of Specific Conditions Rather than Specific Ligands. eNeuro 2019; 6:ENEURO.0171-19.2019. [PMID: 31562177 PMCID: PMC6791830 DOI: 10.1523/eneuro.0171-19.2019] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 08/06/2019] [Accepted: 08/09/2019] [Indexed: 11/21/2022] Open
|
45
|
Walker MC, Kullmann DM. Optogenetic and chemogenetic therapies for epilepsy. Neuropharmacology 2019; 168:107751. [PMID: 31494141 DOI: 10.1016/j.neuropharm.2019.107751] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 08/28/2019] [Accepted: 08/29/2019] [Indexed: 12/15/2022]
Abstract
Drug-resistant epilepsy remains a significant health-care burden. The most effective treatment is surgery, but this is suitable for very few patients because of the unacceptable consequences of removing brain tissue. In contrast, gene therapy can regulate neuronal excitability in the epileptic focus whilst preserving function. Optogenetics and chemogenetics have the advantage that they are titratable therapies. Optogenetics uses light to control the excitability of specific neuronal populations. Optogenetics can be used in a closed-loop paradigm in which the light source is activated only when seizures are detected. However, expression of foreign proteins raises concerns about immunogenicity. Chemogenetics relies on the modification of an endogenous receptor or the production of a modified chimeric receptor that responds to an exogenous ligand. The main chemogenetic approach applied to epilepsy is to use designer receptors exclusively activated by designer drugs (DREADDs), which have been mainly modified muscarinic receptors or kappa-opioid receptors. Genetically modified human muscarinic receptor DREADDs are activated not by acetylcholine but by specific drugs such as clozapine-n-oxide or olanzepine. The dose of the drugs can be titrated in order to suppress seizures without adverse effects. Lastly, there is a chemogenetic approach that is activated by an endogenous ligand, glutamate. This takes advantage of invertebrate glutamate receptors that are chloride permeable. These bind glutamate released during seizure activity, and the resultant chloride current inhibits neuronal activity. The exogenous ligand, ivermectin, can also be given to reduce neuronal activity either chronically or as a rescue medication. The translation of this technology is hampered by the expression of a foreign protein. This article is part of the special issue entitled 'New Epilepsy Therapies for the 21st Century - From Antiseizure Drugs to Prevention, Modification and Cure of Epilepsy'.
Collapse
Affiliation(s)
- Matthew C Walker
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK.
| | - Dimitri M Kullmann
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| |
Collapse
|
46
|
Overexpression of µ-Opioid Receptors in Peripheral Afferents, but Not in Combination with Enkephalin, Decreases Neuropathic Pain Behavior and Enhances Opioid Analgesia in Mouse. Anesthesiology 2019; 128:967-983. [PMID: 29334500 DOI: 10.1097/aln.0000000000002063] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND The current study used recombinant herpes simplex virus type I to increase expression of µ-opiate receptors and the opioid ligand preproenkephalin in peripheral nerve fibers in a mouse model of neuropathic pain. It was predicted that viral vector delivery of a combination of genes encoding the µ-opioid receptor and preproenkephalin would attenuate neuropathic pain and enhance opioid analgesia. The behavioral effects would be paralleled by changes in response properties of primary afferent neurons. METHODS Recombinant herpes simplex virus type 1 containing cDNA sequences of the µ-opioid receptor, human preproenkephalin, a combination, or Escherichia coli lacZ gene marker (as a control) was used to investigate the role of peripheral opioids in neuropathic pain behaviors. RESULTS Inoculation with the µ-opioid receptor viral vector (n = 13) reversed mechanical allodynia and thermal hyperalgesia and produced leftward shifts in loperamide (ED50 = 0.6 ± 0.2 mg/kg vs. ED50 = 0.9 ± 0.2 mg/kg for control group, n = 8, means ± SD) and morphine dose-response curves (ED50 = 0.3 ± 0.5 mg/kg vs. ED50 = 1.1 ± 0.1 mg/kg for control group). In µ-opioid receptor viral vector inoculated C-fibers, heat-evoked responses (n = 12) and ongoing spontaneous activity (n = 18) were decreased after morphine application. Inoculation with both µ-opioid receptor and preproenkephalin viral vectors did not alter mechanical and thermal responses. CONCLUSIONS Increasing primary afferent expression of opioid receptors can decrease neuropathic pain-associated behaviors and increase systemic opioid analgesia through inhibition of peripheral afferent fiber activity.
Collapse
|
47
|
Mondoloni S, Durand-de Cuttoli R, Mourot A. Cell-Specific Neuropharmacology. Trends Pharmacol Sci 2019; 40:696-710. [PMID: 31400823 DOI: 10.1016/j.tips.2019.07.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 06/04/2019] [Accepted: 07/11/2019] [Indexed: 01/12/2023]
Abstract
Neuronal communication involves a multitude of neurotransmitters and an outstanding diversity of receptors and ion channels. Linking the activity of cell surface receptors and ion channels in defined neural circuits to brain states and behaviors has been a key challenge in neuroscience, since cell targeting is not possible with traditional neuropharmacology. We review here recent technologies that enable the effect of drugs to be restricted to specific cell types, thereby allowing acute manipulation of the brain's own proteins with circuit specificity. We highlight the importance of developing cell-specific neuropharmacology strategies for decoding the nervous system with molecular and circuit precision, and for developing future therapeutics with reduced side effects.
Collapse
Affiliation(s)
- Sarah Mondoloni
- Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), CNRS, INSERM, Sorbonne Université, Paris, France
| | - Romain Durand-de Cuttoli
- Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), CNRS, INSERM, Sorbonne Université, Paris, France; Nash Family Department of Neuroscience, Center for Affective Neuroscience, and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alexandre Mourot
- Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), CNRS, INSERM, Sorbonne Université, Paris, France.
| |
Collapse
|
48
|
Price TJ, Gold MS. From Mechanism to Cure: Renewing the Goal to Eliminate the Disease of Pain. PAIN MEDICINE 2019; 19:1525-1549. [PMID: 29077871 DOI: 10.1093/pm/pnx108] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Objective Persistent pain causes untold misery worldwide and is a leading cause of disability. Despite its astonishing prevalence, pain is undertreated, at least in part because existing therapeutics are ineffective or cause intolerable side effects. In this review, we cover new findings about the neurobiology of pain and argue that all but the most transient forms of pain needed to avoid tissue damage should be approached as a disease where a cure can be the goal of all treatment plans, even if attaining this goal is not yet always possible. Design We reviewed the literature to highlight recent advances in the area of the neurobiology of pain. Results We discuss barriers that are currently hindering the achievement of this goal, as well as the development of new therapeutic strategies. We also discuss innovations in the field that are creating new opportunities to treat and even reverse persistent pain, some of which are in late-phase clinical trials. Conclusion We conclude that the confluence of new basic science discoveries and development of new technologies are creating a path toward pain therapeutics that should offer significant hope of a cure for patients and practitioners alike. Classification of Evidence. Our review points to new areas of inquiry for the pain field to advance the goal of developing new therapeutics to treat chronic pain.
Collapse
Affiliation(s)
- Theodore J Price
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Dallas, Texas
| | - Michael S Gold
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
49
|
In Vitro Nociceptor Neuroplasticity Associated with In Vivo Opioid-Induced Hyperalgesia. J Neurosci 2019; 39:7061-7073. [PMID: 31300521 DOI: 10.1523/jneurosci.1191-19.2019] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 07/02/2019] [Accepted: 07/04/2019] [Indexed: 11/21/2022] Open
Abstract
Opioid-induced hyperalgesia (OIH) is a serious adverse event produced by opioid analgesics. Lack of an in vitro model has hindered study of its underlying mechanisms. Recent evidence has implicated a role of nociceptors in OIH. To investigate the cellular and molecular mechanisms of OIH in nociceptors, in vitro, subcutaneous administration of an analgesic dose of fentanyl (30 μg/kg, s.c.) was performed in vivo in male rats. Two days later, when fentanyl was administered intradermally (1 μg, i.d.), in the vicinity of peripheral nociceptor terminals, it produced mechanical hyperalgesia (OIH). Additionally, 2 d after systemic fentanyl, rats had also developed hyperalgesic priming (opioid-primed rats), long-lasting nociceptor neuroplasticity manifested as prolongation of prostaglandin E2 (PGE2) hyperalgesia. OIH was reversed, in vivo, by intrathecal administration of cordycepin, a protein translation inhibitor that reverses priming. When fentanyl (0.5 nm) was applied to dorsal root ganglion (DRG) neurons, cultured from opioid-primed rats, it induced a μ-opioid receptor (MOR)-dependent increase in [Ca2+]i in 26% of small-diameter neurons and significantly sensitized (decreased action potential rheobase) weakly IB4+ and IB4- neurons. This sensitizing effect of fentanyl was reversed in weakly IB4+ DRG neurons cultured from opioid-primed rats after in vivo treatment with cordycepin, to reverse of OIH. Thus, in vivo administration of fentanyl induces nociceptor neuroplasticity, which persists in culture, providing evidence for the role of nociceptor MOR-mediated calcium signaling and peripheral protein translation, in the weakly IB4-binding population of nociceptors, in OIH.SIGNIFICANCE STATEMENT Clinically used μ-opioid receptor agonists such as fentanyl can produce hyperalgesia and hyperalgesic priming. We report on an in vitro model of nociceptor neuroplasticity mediating this opioid-induced hyperalgesia (OIH) and priming induced by fentanyl. Using this model, we have found qualitative and quantitative differences between cultured nociceptors from opioid-naive and opioid-primed animals, and provide evidence for the important role of nociceptor μ-opioid receptor-mediated calcium signaling and peripheral protein translation in the weakly IB4-binding population of nociceptors in OIH. These findings provide information useful for the design of therapeutic strategies to alleviate OIH, a serious adverse event of opioid analgesics.
Collapse
|
50
|
|