1
|
Pham TL, Sharma R, Neupane C, Gao F, Cha GH, Kim H, Nam MH, Lee SE, Yang S, Sim H, Lee S, Hur GM, Kim HW, Park JB. Neuronal STING-GAT1 signaling maintains paclitaxel-induced neuropathic pain in the spinal cord. Pain 2025:00006396-990000000-00886. [PMID: 40310867 DOI: 10.1097/j.pain.0000000000003593] [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: 03/30/2024] [Accepted: 02/10/2025] [Indexed: 05/03/2025]
Abstract
ABSTRACT Stimulator of interferon genes (STING), a pivotal immune regulator, has emerged as a contributor to nociception, yet its role in chronic pains remains still unknown. Here, we demonstrate that STING plays a dual role in normal and neuropathic pain in mature male rodents. Stimulator of interferon genes maintains type I interferon (IFN-I) level restraining pain sensitivity in normal and sham control, while activated STING/interferon regulatory factor 3 (IRF3) signaling increases the expression of gamma-aminobutyric acid (GABA) transporter 1 (GAT1) in the spinal cord (SC), thus, generating paclitaxel (PTX)-induced peripheral neuropathy. Genetic interference of STING (STING-/- mice) attenuated PTX-induced mechanical hypersensitivity with attenuated PTX-induced GAT1 increase, preventing PTX-induced increase in tonic GABAA inhibition of the spinal dorsal horn neurons. Stimulator of interferon genes regulates GAT expression through a TANK-binding kinase 1 (TBK1)-IRF3 signaling pathway, with IRF3 as a crucial transcription factor. Silencing neuronal STING, as opposed to its astrocytic counterpart, effectively restrained the PTX-induced mechanical hypersensitivity and GAT1 increase in the SC. Pharmacological inhibition of STING (H-151) efficiently diminished the TBK1/IRF3/GAT1 signaling pathway to alleviate PTX-induced mechanical hypersensitivity. Our findings show that STING-IRF3 serves a dual role: suppressing physiological nociception through IFN-I and acting as a transcriptional regulator of GAT1, contributing to chemotherapy-induced neuropathic pain.
Collapse
Affiliation(s)
- Thuy Linh Pham
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, Korea
- Department of Physiology and Medical Science, College of Medicine and Brain Research Institute, Chungnam National University, Daejeon, South Korea
- Department of Obstetrics and Gynecology, Viet Tiep Friendship Hospital, Hai Phong, Vietnam
| | - Ramesh Sharma
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, Korea
- Department of Physiology and Medical Science, College of Medicine and Brain Research Institute, Chungnam National University, Daejeon, South Korea
| | - Chiranjivi Neupane
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, Korea
- Department of Physiology and Medical Science, College of Medicine and Brain Research Institute, Chungnam National University, Daejeon, South Korea
| | - Feifei Gao
- Department of Infectious Biology, Chungnam National University, Daejeon, South Korea
| | - Guang-Ho Cha
- Department of Infectious Biology, Chungnam National University, Daejeon, South Korea
| | - Hyunjin Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Min-Ho Nam
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Seung Eun Lee
- Research Animal Resource Center, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Sunjung Yang
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, Korea
| | - Hunju Sim
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, Korea
| | - Sanghoon Lee
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, Korea
| | - Gang Min Hur
- Pharmacology and Medical Science, Chungnam National University, Daejeon, South Korea
| | - Hyun-Woo Kim
- Department of Physiology and Medical Science, College of Medicine and Brain Research Institute, Chungnam National University, Daejeon, South Korea
| | - Jin Bong Park
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, Korea
| |
Collapse
|
2
|
Bekhbat M, Block AM, Dickinson SY, Tharp GK, Bosinger SE, Felger JC. Neurotransmitter and metabolic effects of interferon-alpha in association with decreased striatal dopamine in a non-human primate model of cytokine-Induced depression. Brain Behav Immun 2025; 125:308-318. [PMID: 39826580 PMCID: PMC11903159 DOI: 10.1016/j.bbi.2025.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 12/13/2024] [Accepted: 01/13/2025] [Indexed: 01/22/2025] Open
Abstract
Inflammatory stimuli administered to humans and laboratory animals affect mesolimbic and nigrostriatal dopaminergic pathways in association with impaired motivation and motor activity. Alterations in dopaminergic corticostriatal reward and motor circuits have also been observed in depressed patients with increased peripheral inflammatory markers. The effects of peripheral inflammation on dopaminergic pathways and associated neurobiologic mechanisms and consequences have been difficult to measure in patients. Postmortem tissue (n = 11) from an established, translationally-relevant non-human primate model of cytokine-induced depressive behavior involving chronic interferon-alpha (IFN-a) administration was utilized herein to explore the molecular mechanisms of peripheral cytokine effects on striatal dopamine. Dopamine (but not serotonin or norepinephrine) was decreased in the nucleus accumbens (NAcc) and putamen of IFN-a-treated animals (p < 0.05). IFN-a had no effect on number of striatal neurons or dopamine terminal density, suggesting no overt neurodegenerative changes. RNA sequencing examined in the caudate, putamen, substantia nigra, and prefrontal cortical subregions revealed that while IFN-a nominally up-regulated limited numbers of genes enriching inflammatory signaling pathways in all regions, robust, whole genome-significant effects of IFN-a were observed specifically in putamen. Genes upregulated in the putamen primarily enriched synaptic signaling, glutamate receptor signaling, and inflammatory/metabolic pathways downstream of IFN-a, including MAPK and PI3K/AKT cascades. Conversely, gene transcripts reduced by IFN-a enriched oxidative phosphorylation (OXPHOS), protein translation, and pathways regulated by dopamine receptors. Unsupervised clustering identified a gene co-expression module in the putamen that was associated with both IFN-a treatment and low dopamine levels, which enriched similar inflammatory, metabolic, and synaptic signaling pathways. IFN-a-induced reductions in dopamine further correlated with genes related to excitotoxic glutamate, kynurenine, and altered dopamine receptor signaling (r = 0.78-97, p < 0.05). These findings provide insight into the immunologic mechanisms and neurobiological consequences of peripheral inflammation effects on dopamine, which may inform novel treatment strategies targeting inflammatory, metabolic or neurotransmitter systems in depressed patients with high inflammation.
Collapse
Affiliation(s)
- Mandakh Bekhbat
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA.
| | - Andrew M Block
- Department of Orthopaedic Surgery, University of Connecticut, Farmington, CT 06030, USA
| | - Sarah Y Dickinson
- Neuroscience and Behavior Program, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Gregory K Tharp
- Emory Nonhuman Primate Genomics Core, Division of Microbiology and Immunology, Emory National Primate Research Center (EPC), Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Steven E Bosinger
- Emory Nonhuman Primate Genomics Core, Division of Microbiology and Immunology, Emory National Primate Research Center (EPC), Emory University School of Medicine, Atlanta, GA 30322, USA; Department of Pathology and Laboratory Medicine, Emory School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Jennifer C Felger
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA; Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA.
| |
Collapse
|
3
|
Buonfiglioli A, Kübler R, Missall R, De Jong R, Chan S, Haage V, Wendt S, Lin AJ, Mattei D, Graziani M, Latour B, Gigase F, Chiu R, Zhang Y, Nygaard HB, De Jager PL, De Witte LD. A microglia-containing cerebral organoid model to study early life immune challenges. Brain Behav Immun 2025; 123:1127-1146. [PMID: 39500415 PMCID: PMC11753195 DOI: 10.1016/j.bbi.2024.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 10/10/2024] [Accepted: 11/02/2024] [Indexed: 11/13/2024] Open
Abstract
Prenatal infections and activation of the maternal immune system have been proposed to contribute to causing neurodevelopmental disorders (NDDs), chronic conditions often linked to brain abnormalities. Microglia are the resident immune cells of the brain and play a key role in neurodevelopment. Disruption of microglial functions can lead to brain abnormalities and increase the risk of developing NDDs. How the maternal as well as the fetal immune system affect human neurodevelopment and contribute to NDDs remains unclear. An important reason for this knowledge gap is the fact that the impact of exposure to prenatal risk factors has been challenging to study in the human context. Here, we characterized a model of cerebral organoids (CO) with integrated microglia (COiMg). These organoids express typical microglial markers and respond to inflammatory stimuli. The presence of microglia influences cerebral organoid development, including cell density and neural differentiation, and regulates the expression of several ciliated and mesenchymal cell markers. Moreover, COiMg and organoids without microglia show similar but also distinct responses to inflammatory stimuli. Additionally, IFN-γ induced significant transcriptional and structural changes in the cerebral organoids, that appear to be regulated by the presence of microglia. Specifically, interferon-gamma (IFN-γ) was found to alter the expression of genes linked to autism. This model provides a valuable tool to study how inflammatory perturbations and microglial presence affect neurodevelopmental processes.
Collapse
Affiliation(s)
- Alice Buonfiglioli
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Raphael Kübler
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Human Genetics, Radboud UMC, Nijmegen, Netherlands (the)
| | - Roy Missall
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Renske De Jong
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Stephanie Chan
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Verena Haage
- Center for Translational & Computational Neuroimmunology, Department of Neurology and the Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Stefan Wendt
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver V6T 1Z3, Canada
| | - Ada J Lin
- Division of Neurology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver V6T 1Z3, Canada
| | - Daniele Mattei
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mara Graziani
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Human Genetics, Radboud UMC, Nijmegen, Netherlands (the); Donders Institute for Brain, Cognition and Behaviour, 6500 HB, Nijmegen, Netherlands (the)
| | - Brooke Latour
- Department of Human Genetics, Radboud UMC, Nijmegen, Netherlands (the); Donders Institute for Brain, Cognition and Behaviour, 6500 HB, Nijmegen, Netherlands (the)
| | - Frederieke Gigase
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Rebecca Chiu
- Center for Translational & Computational Neuroimmunology, Department of Neurology and the Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Ya Zhang
- Center for Translational & Computational Neuroimmunology, Department of Neurology and the Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Haakon B Nygaard
- Division of Neurology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver V6T 1Z3, Canada
| | - Philip L De Jager
- Center for Translational & Computational Neuroimmunology, Department of Neurology and the Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Lot D De Witte
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Human Genetics, Radboud UMC, Nijmegen, Netherlands (the); Donders Institute for Brain, Cognition and Behaviour, 6500 HB, Nijmegen, Netherlands (the); Department of Psychiatry, Radboud UMC, Nijmegen, Netherlands (the)
| |
Collapse
|
4
|
Zhao W, Liu SL, Lin SS, Zhang Y, Yu C. Astrocytic P2X7 receptor in retrosplenial cortex drives electroacupuncture analgesia. Purinergic Signal 2024:10.1007/s11302-024-10043-w. [PMID: 39222236 DOI: 10.1007/s11302-024-10043-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 08/15/2024] [Indexed: 09/04/2024] Open
Abstract
P2X7 receptor (P2X7R) has been found to contribute to the peripheral mechanism of acupuncture analgesia (AA). However, whether it plays an important role in central mechanism remains unknown. In this study, we aimed to reveal the role of astrocytic P2X7R in retrosplenial cortex (RSC) in AA and provide new evidence for underlying the central mechanism of AA. We applied the chemogenetic receptors hM3Dq to stimulate or hM4Di to inhibit astrocytes ligand clozapine-N-oxide (CNO) following injection of adeno-associated virus (AAV) into the bilateral RSC, or pharmacologically intervened in the activity of the purinergic receptor P2X7R. Current data indicated that chemogenetic inhibition of astrocytes or injection of P2X7R agonist Bz-ATP in the bilateral RSC significantly reverses the analgesic effect of electroacupuncture (EA) in formalin tests while the bilateral injection of the P2X7R antagonist A438079 alleviated formalin-induced nociceptive behavior. Additionally, chemogenetic suppression of astrocytic P2X7R by injection of AAV in the bilateral RSC decreased hind paw flinches induced by formalin in the mice. These findings indicate the participation of both astrocytes and P2X7R in the RSC in EA analgesic. Moreover, P2X7R on astrocytes in the RSC appears to play a critical role in the ability of EA to attenuate formalin-induced pain responses in mice.
Collapse
Affiliation(s)
- Wei Zhao
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Si-Le Liu
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Si-Si Lin
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ying Zhang
- School of Health and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Chang Yu
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
5
|
Rodriguez-Palma EJ, Allen HN, Khanna R. STINGing away the pain: the role of interferon-stimulated genes. J Clin Invest 2024; 134:e180497. [PMID: 38690736 PMCID: PMC11060727 DOI: 10.1172/jci180497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2024] Open
Abstract
Pain and inflammation are biologically intertwined responses that warn the body of potential danger. In this issue of the JCI, Defaye, Bradaia, and colleagues identified a functional link between inflammation and pain, demonstrating that inflammation-induced activation of stimulator of IFN genes (STING) in dorsal root ganglia nociceptors reduced pain-like behaviors in a rodent model of inflammatory pain. Utilizing mice with a gain-of-function STING mutation, Defaye, Bradaia, and colleagues identified type I IFN regulation of voltage-gated potassium channels as the mechanism of this pain relief. Further investigation into mechanisms by which proinflammatory pathways can reduce pain may reveal druggable targets and insights into new approaches for treating persistent pain.
Collapse
|
6
|
Li H, Wang C, Gong Z, Nie L, Xu J, Wang M. Transient Receptor Potential Ankyrin 1-dependent Activation of Extracellular Signal-regulated Kinase 2 in the Cerebral Cortices Contributes to Cortical Spreading Depolarization. Neuroscience 2024; 543:90-100. [PMID: 38417540 DOI: 10.1016/j.neuroscience.2024.02.009] [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: 11/20/2023] [Revised: 02/07/2024] [Accepted: 02/12/2024] [Indexed: 03/01/2024]
Abstract
Extracellular signal-regulated kinase (ERK) are serine/threonine-selective proteins and ERK1/2 can be phosphorylated in peripheral and central brain regions after cortical spreading depolarization (CSD) and calcitonin gene-related peptide; However, it remains unclear about whether and how ERK activity modulates CSD that correlates to migraine aura. Here, we determined the role of ERK in regulating CSD and explored the underlying mechanism involving transient receptor potential ankyrin 1 (TRPA1), a stress-sensing cation channel. CSD was recorded using intrinsic optical imaging in mouse brain slices, and electrophysiology in rats. Phosphorylated ERK (pERK1/2) and interleukin-1β (IL-1β) protein levels were detected using Western blot or enzyme-linked immunosorbent assay, respectively. IL-1β mRNA level was detected using qPCR. The results showed that an ERK inhibitor, SCH77298, markedly prolonged CSD latency and reduced propagation rate in mouse brain slices. Corresponding to this, CSD induction increased levels of cytosolic pERK1/2 in ipsilateral cerebral cortices of rats, the elevation of which correlated to the level of IL-1β mRNA. Mechanistic analysis showed that pre-treatment of an anti-TRPA1 antibody reduced the cytosolic pERK2 level but not pERK1 following CSD in cerebral cortices of rats and this level of pERK2 correlated with that of cerebral cortical IL-1β protein. Furthermore, an ERK activator, AES16-2M, but not its scrambled control, reversed the prolonged CSD latency by a TRPA1 inhibitor, HC-030031, in mouse brain slices. These data revealed a crucial role of ERK activity in regulating CSD, and elevation of pERK and IL-1β production induced by CSD is predominantly TRPA1 channel-dependent, thereby contributing to migraine pathogenesis.
Collapse
Affiliation(s)
- Haoyang Li
- Department of Biological Sciences, Centre for Neuroscience, School of Science, Xi'an Jiaotong-Liverpool University, China
| | - Chenyi Wang
- Department of Biological Sciences, Centre for Neuroscience, School of Science, Xi'an Jiaotong-Liverpool University, China
| | - Ziyang Gong
- Department of Biological Sciences, Centre for Neuroscience, School of Science, Xi'an Jiaotong-Liverpool University, China
| | - Lingdi Nie
- Department of Biological Sciences, Centre for Neuroscience, School of Science, Xi'an Jiaotong-Liverpool University, China
| | - Jiaxin Xu
- Department of Biological Sciences, Centre for Neuroscience, School of Science, Xi'an Jiaotong-Liverpool University, China
| | - Minyan Wang
- Department of Biological Sciences, Centre for Neuroscience, School of Science, Xi'an Jiaotong-Liverpool University, China.
| |
Collapse
|
7
|
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
|
8
|
Silveira Prudente A, Hoon Lee S, Roh J, Luckemeyer DD, Cohen CF, Pertin M, Park CK, Suter MR, Decosterd I, Zhang JM, Ji RR, Berta T. Microglial STING activation alleviates nerve injury-induced neuropathic pain in male but not female mice. Brain Behav Immun 2024; 117:51-65. [PMID: 38190983 PMCID: PMC11034751 DOI: 10.1016/j.bbi.2024.01.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 12/21/2023] [Accepted: 01/03/2024] [Indexed: 01/10/2024] Open
Abstract
Microglia, resident immune cells in the central nervous system, play a role in neuroinflammation and the development of neuropathic pain. We found that the stimulator of interferon genes (STING) is predominantly expressed in spinal microglia and upregulated after peripheral nerve injury. However, mechanical allodynia, as a marker of neuropathic pain following peripheral nerve injury, did not require microglial STING expression. In contrast, STING activation by specific agonists (ADU-S100, 35 nmol) significantly alleviated neuropathic pain in male mice, but not female mice. STING activation in female mice leads to increase in proinflammatory cytokines that may counteract the analgesic effect of ADU-S100. Microglial STING expression and type I interferon-ß (IFN-ß) signaling were required for the analgesic effects of STING agonists in male mice. Mechanistically, downstream activation of TANK-binding kinase 1 (TBK1) and the production of IFN-ß, may partly account for the analgesic effect observed. These findings suggest that STING activation in spinal microglia could be a potential therapeutic intervention for neuropathic pain, particularly in males.
Collapse
Affiliation(s)
- Arthur Silveira Prudente
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH, USA
| | - Sang Hoon Lee
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH, USA
| | - Jueun Roh
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH, USA; Department of Physiology, Gachon Pain Center, Gachon University College of Medicine, Incheon, South Korea
| | - Debora D Luckemeyer
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH, USA
| | - Cinder F Cohen
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH, USA
| | - Marie Pertin
- Pain Center, Department of Anesthesiology, Lausanne University Hospital (CHUV) and University of Lausanne, 1011 Lausanne, Switzerland; Department of Fundamental Neurosciences, Faculty of Biology and Medicine, University of Lausanne, 1011 Lausanne, Switzerland
| | - Chul-Kyu Park
- Department of Physiology, Gachon Pain Center, Gachon University College of Medicine, Incheon, South Korea
| | - Marc R Suter
- Pain Center, Department of Anesthesiology, Lausanne University Hospital (CHUV) and University of Lausanne, 1011 Lausanne, Switzerland; Department of Fundamental Neurosciences, Faculty of Biology and Medicine, University of Lausanne, 1011 Lausanne, Switzerland
| | - Isabelle Decosterd
- Pain Center, Department of Anesthesiology, Lausanne University Hospital (CHUV) and University of Lausanne, 1011 Lausanne, Switzerland; Department of Fundamental Neurosciences, Faculty of Biology and Medicine, University of Lausanne, 1011 Lausanne, Switzerland
| | - Jun-Ming Zhang
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH, USA
| | - Ru-Rong Ji
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA; Departments of Cell Biology and Neurobiology, Duke University Medical Center, Durham, NC, USA
| | - Temugin Berta
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH, USA.
| |
Collapse
|
9
|
Shaikh A, Li YQ, Lu J. Perspectives on pain in Down syndrome. Med Res Rev 2023; 43:1411-1437. [PMID: 36924439 DOI: 10.1002/med.21954] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 01/08/2023] [Accepted: 02/28/2023] [Indexed: 03/18/2023]
Abstract
Down syndrome (DS) or trisomy 21 is a genetic condition often accompanied by chronic pain caused by congenital abnormalities and/or conditions, such as osteoarthritis, recurrent infections, and leukemia. Although DS patients are more susceptible to chronic pain as compared to the general population, the pain experience in these individuals may vary, attributed to the heterogenous structural and functional differences in the central nervous system, which might result in abnormal pain sensory information transduction, transmission, modulation, and perception. We tried to elaborate on some key questions and possible explanations in this review. Further clarification of the mechanisms underlying such abnormal conditions induced by the structural and functional differences is needed to help pain management in DS patients.
Collapse
Affiliation(s)
- Ammara Shaikh
- Department of Human Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning Province, China
| | - Yun-Qing Li
- Department of Anatomy, Histology, and Embryology & K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, Shaanxi Province, China
- Department of Anatomy, Basic Medical College, Zhengzhou University, Zhengzhou, China
| | - Jie Lu
- Department of Human Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning Province, China
| |
Collapse
|
10
|
Franco-Enzástiga Ú, Natarajan K, David ET, Patel KJ, Ravirala A, Price TJ. Vinorelbine causes a neuropathic pain-like state in mice via STING and MNK1 signaling associated with type I interferon induction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.03.543579. [PMID: 37333411 PMCID: PMC10274710 DOI: 10.1101/2023.06.03.543579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Type I interferons (IFNs) increase the excitability of dorsal root ganglion (DRG) neurons via activation of MNK-eIF4E translation signaling to promote pain sensitization in mice. Activation of STING signaling is a key component of type I IFN induction. Manipulation of STING signaling is an active area of investigation in cancer and other therapeutic areas. Vinorelbine is a chemotherapeutic that activates STING and has been shown to cause pain and neuropathy in oncology clinical trials in patients. There are conflicting reports on whether STING signaling promotes or inhibits pain in mice. We hypothesized that vinorelbine would cause a neuropathic pain-like state in mice via STING and signaling pathways in DRG neurons associated with type I IFN induction. Vinorelbine (10 mg/kg, i.v.) induced tactile allodynia and grimacing in WT male and female mice and increased p-IRF3 and type I IFN protein in peripheral nerves. In support of our hypothesis, vinorelbine-mediated pain was absent in male and female StingGt/Gt mice. Vinorelbine also failed to induce IRF3 and type I IFN signaling in these mice. Since type I IFNs engage translational control via MNK1-eIF4E in DRG nociceptors, we assessed vinorelbine-mediated p-eIF4E changes. Vinorelbine increased p-eIF4E in DRG in WT animals but not in StingGt/Gt or Mknk1-/- (MNK1 KO) mice. Consistent with these biochemical findings, vinorelbine had an attenuated pro-nociceptive effect in male and female MNK1 KO mice. Our findings support the conclusion that activation of STING signaling in the peripheral nervous system causes a neuropathic pain-like state that is mediated by type I IFN signaling to DRG nociceptors.
Collapse
Affiliation(s)
- Úrzula Franco-Enzástiga
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080
| | - Keerthana Natarajan
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080
| | - Eric T. David
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080
| | - Krish J. Patel
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080
| | - Abhira Ravirala
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080
| | - Theodore J. Price
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080
| |
Collapse
|
11
|
Ming LG, Hu DX, Zuo C, Zhang WJ. G protein-coupled P2Y12 receptor is involved in the progression of neuropathic pain. Biomed Pharmacother 2023; 162:114713. [PMID: 37084563 DOI: 10.1016/j.biopha.2023.114713] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/13/2023] [Accepted: 04/14/2023] [Indexed: 04/23/2023] Open
Abstract
The pathological mechanism of neuropathic pain is complex, which seriously affects the physical and mental health of patients, and its treatment is also difficult. The role of G protein-coupled P2Y12 receptor in pain has been widely recognized and affirmed. After nerve injury, stimulated cells can release large amounts of nucleotides into the extracellular matrix, act on P2Y12 receptor. Activated P2Y12 receptor activates intracellular signal transduction and is involved in the development of pain. P2Y12 receptor activation can sensitize primary sensory neurons and receive sensory information. By transmitting the integrated information through the dorsal root of the spinal cord to the secondary neurons of the posterior horn of the spinal cord. The integrated information is then transmitted to the higher center through the ascending conduction tract to produce pain. Moreover, activation of P2Y12 receptor can mediate immune cells to release pro-inflammatory factors, increase damage to nerve cells, and aggravate pain. While inhibits the activation of P2Y12 receptor can effectively relieve pain. Therefore, in this article, we described P2Y12 receptor antagonists and their pharmacological properties. In addition, we explored the potential link between P2Y12 receptor and the nervous system, discussed the intrinsic link of P2Y12 receptor and neuropathic pain and as a potential pharmacological target for pain suppression.
Collapse
Affiliation(s)
- Li-Guo Ming
- Department of Gastrointestinal surgery, the Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi Province 343000, China
| | - Dong-Xia Hu
- Department of Rehabilitation Medicine, the Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi Province 343000, China
| | - Cheng Zuo
- Department of Gastrointestinal surgery, the Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi Province 343000, China
| | - Wen-Jun Zhang
- Department of Rehabilitation Medicine, the Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi Province 343000, China.
| |
Collapse
|
12
|
Adhikari UK, Khan R, Mikhael M, Balez R, David MA, Mahns D, Hardy J, Tayebi M. Therapeutic anti-amyloid β antibodies cause neuronal disturbances. Alzheimers Dement 2022. [PMID: 36515320 DOI: 10.1002/alz.12833] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 09/07/2022] [Accepted: 09/19/2022] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Recent published clinical trial safety data showed that 41% of Alzheimer patients experienced amyloid-related imaging abnormalities (ARIA), marks of microhemorrhages and edema in the brain, following administration of Biogen's Aduhelm/aducanumab (amino acids 3-7 of the Aβ peptide). Similarly, Janssen/Pfizer's Bapineuzumab (amino acids 1-5 of the Aβ peptide) and Roche's Gantenerumab (amino acids 2-11/18-27 of the Aβ peptide) also displayed ARIA in clinical trials, including microhemorrhage and focal areas of inflammation or vasogenic edema, respectively. The molecular mechanisms underlying ARIA caused by therapeutic anti-Aβ antibodies remain largely unknown, however, recent reports demonstrated that therapeutic anti-prion antibodies activate neuronal allergenic proteomes following cross-linking cellular prion protein. METHODS Here, we report that treatment of human induced pluripotent stem cells- derived neurons (HSCN) from a non-demented donor, co-cultured with human primary microglia with anti-Aβ1-6, or anti-Aβ17-23 antibodies activate a significant number of allergenic-related proteins as assessed by mass spectrometry. RESULTS Interestingly, a large proportion of the identified proteins included cytokines such as interleukin (IL)-4, IL-12, and IL-13 suggesting a type-1 hypersensitivity response. Following flow cytometry analysis, several proinflammatory cytokines were significantly elevated following anti-Aβ1-6, or anti-Aβ17-23 antibody treatment. DISCUSSION These results justify further and more robust investigation of the molecular mechanisms of ARIA during immunotherapy study trials of AD. HIGHLIGHTS Allergenic-related proteins are often linked with Alzheimer's disease (AD). We investigated the effects of amyloid beta (Aβ) immunotherapy on stem cell derived neurons and primary neuronal cells co-cultured with microglia. Anti-Aβ antibody treatment of neurons or neurons co-cultured with microglia led to activation of a substantial number of allergenic-related genes. These allergenic-related genes are associated with endothelial dysfunction possibly responsible for ARIA.
Collapse
Affiliation(s)
- Utpal Kumar Adhikari
- School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| | - Rizwan Khan
- School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| | - Meena Mikhael
- School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| | - Rachelle Balez
- Illawarra Health and Medical Research Institute, Wollongong, New South Wales, Australia
- School of Chemistry and Molecular Bioscience and Molecular Horizons, University of Wollongong, Wollongong, New South Wales, Australia
| | | | - David Mahns
- School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| | - John Hardy
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London, UK
| | - Mourad Tayebi
- School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| |
Collapse
|
13
|
Tan PH, Ji J, Hsing CH, Tan R, Ji RR. Emerging Roles of Type-I Interferons in Neuroinflammation, Neurological Diseases, and Long-Haul COVID. Int J Mol Sci 2022; 23:ijms232214394. [PMID: 36430870 PMCID: PMC9696119 DOI: 10.3390/ijms232214394] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/11/2022] [Accepted: 11/15/2022] [Indexed: 11/22/2022] Open
Abstract
Interferons (IFNs) are pleiotropic cytokines originally identified for their antiviral activity. IFN-α and IFN-β are both type I IFNs that have been used to treat neurological diseases such as multiple sclerosis. Microglia, astrocytes, as well as neurons in the central and peripheral nervous systems, including spinal cord neurons and dorsal root ganglion neurons, express type I IFN receptors (IFNARs). Type I IFNs play an active role in regulating cognition, aging, depression, and neurodegenerative diseases. Notably, by suppressing neuronal activity and synaptic transmission, IFN-α and IFN-β produced potent analgesia. In this article, we discuss the role of type I IFNs in cognition, neurodegenerative diseases, and pain with a focus on neuroinflammation and neuro-glial interactions and their effects on cognition, neurodegenerative diseases, and pain. The role of type I IFNs in long-haul COVID-associated neurological disorders is also discussed. Insights into type I IFN signaling in neurons and non-neuronal cells will improve our treatments of neurological disorders in various disease conditions.
Collapse
Affiliation(s)
- Ping-Heng Tan
- Department of Anesthesiology, Chi Mei Medical Center, Tainan 701, Taiwan
- Correspondence: (P.-H.T.); (C.-H.H.)
| | - Jasmine Ji
- Neuroscience Department, Wellesley College, Wellesley, MA 02482, USA
| | - Chung-Hsi Hsing
- Department of Anesthesiology, Chi Mei Medical Center, Tainan 701, Taiwan
- Correspondence: (P.-H.T.); (C.-H.H.)
| | - Radika Tan
- Kaohsiung American School, Kaohsiung 81354, Taiwan
| | - Ru-Rong Ji
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
- Departments of Cell Biology and Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
14
|
Braden K, Campolo M, Li Y, Chen Z, Doyle TM, Giancotti LA, Esposito E, Zhang J, Cuzzocrea S, Arnatt CK, Salvemini D. Activation of GPR183 by 7 α,25-Dihydroxycholesterol Induces Behavioral Hypersensitivity through Mitogen-Activated Protein Kinase and Nuclear Factor- κB. J Pharmacol Exp Ther 2022; 383:172-181. [PMID: 36116795 PMCID: PMC9553113 DOI: 10.1124/jpet.122.001283] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 08/17/2022] [Indexed: 01/07/2023] Open
Abstract
Emerging evidence implicates the G-protein coupled receptor (GPCR) GPR183 in the development of neuropathic pain. Further investigation of the signaling pathways downstream of GPR183 is needed to support the development of GPR183 antagonists as analgesics. In rodents, intrathecal injection of its ligand, 7α,25-dihydroxycholesterol (7α,25-OHC), causes time-dependent development of mechano-and cold- allodynia (behavioral hypersensitivity). These effects are blocked by the selective small molecule GPR183 antagonist, SAE-14. However, the molecular mechanisms engaged downstream of GPR183 in the spinal cord are not known. Here, we show that 7α,25-OHC-induced behavioral hypersensitivity is Gα i dependent, but not β-arrestin 2-dependent. Non-biased transcriptomic analyses of dorsal-horn spinal cord (DH-SC) tissues harvested at the time of peak hypersensitivity implicate potential contributions of mitogen-activated protein kinase (MAPK) and nuclear factor κB (NF-κB). In support, we found that the development of 7α,25-OHC/GPR183-induced mechano-allodynia was associated with significant activation of MAPKs (extracellular signal-regulated kinase [ERK], p38) and redox-sensitive transcription factors (NF-κB) and increased formation of inflammatory and neuroexcitatory cytokines. SAE-14 blocked these effects and behavioral hypersensitivity. Our findings provide novel mechanistic insight into how GPR183 signaling in the spinal cord produces hypersensitivity through MAPK and NF-κB activation. SIGNIFICANCE STATEMENT: Using a multi-disciplinary approach, we have characterized the molecular mechanisms underpinning 7α,25-OHC/GPR183-induced hypersensitivity in mice. Intrathecal injections of the GPR183 agonist 7α,25-OHC induce behavioral hypersensitivity, and these effects are blocked by the selective GPR183 antagonist SAE-14. We found that 7α,25-OHC-induced allodynia is dependent on MAPK and NF-κB signaling pathways and results in an increase in pro-inflammatory cytokine expression. This study provides a first insight into how GPR183 signaling in the spinal cord is pronociceptive.
Collapse
Affiliation(s)
- Kathryn Braden
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (K.B., Y.L., Z.C., T.M.D., L.A.G., J.Z., C.K.A., D.S.); Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, Saint Louis, Missouri (K.B., Y.L., Z.C., T.M.D., L.A.G., J.Z., C.K.A.,D.S.); Department of Clinical and Experimental Medicine and Pharmacology, University of Messina, Messina, Italy (M.C., E.E., S.C.); and Department of Chemistry, Saint Louis University, Saint Louis, Missouri (C.K.A.)
| | - Michela Campolo
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (K.B., Y.L., Z.C., T.M.D., L.A.G., J.Z., C.K.A., D.S.); Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, Saint Louis, Missouri (K.B., Y.L., Z.C., T.M.D., L.A.G., J.Z., C.K.A.,D.S.); Department of Clinical and Experimental Medicine and Pharmacology, University of Messina, Messina, Italy (M.C., E.E., S.C.); and Department of Chemistry, Saint Louis University, Saint Louis, Missouri (C.K.A.)
| | - Ying Li
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (K.B., Y.L., Z.C., T.M.D., L.A.G., J.Z., C.K.A., D.S.); Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, Saint Louis, Missouri (K.B., Y.L., Z.C., T.M.D., L.A.G., J.Z., C.K.A.,D.S.); Department of Clinical and Experimental Medicine and Pharmacology, University of Messina, Messina, Italy (M.C., E.E., S.C.); and Department of Chemistry, Saint Louis University, Saint Louis, Missouri (C.K.A.)
| | - Zhoumou Chen
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (K.B., Y.L., Z.C., T.M.D., L.A.G., J.Z., C.K.A., D.S.); Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, Saint Louis, Missouri (K.B., Y.L., Z.C., T.M.D., L.A.G., J.Z., C.K.A.,D.S.); Department of Clinical and Experimental Medicine and Pharmacology, University of Messina, Messina, Italy (M.C., E.E., S.C.); and Department of Chemistry, Saint Louis University, Saint Louis, Missouri (C.K.A.)
| | - Timothy M Doyle
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (K.B., Y.L., Z.C., T.M.D., L.A.G., J.Z., C.K.A., D.S.); Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, Saint Louis, Missouri (K.B., Y.L., Z.C., T.M.D., L.A.G., J.Z., C.K.A.,D.S.); Department of Clinical and Experimental Medicine and Pharmacology, University of Messina, Messina, Italy (M.C., E.E., S.C.); and Department of Chemistry, Saint Louis University, Saint Louis, Missouri (C.K.A.)
| | - Luigino Antonio Giancotti
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (K.B., Y.L., Z.C., T.M.D., L.A.G., J.Z., C.K.A., D.S.); Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, Saint Louis, Missouri (K.B., Y.L., Z.C., T.M.D., L.A.G., J.Z., C.K.A.,D.S.); Department of Clinical and Experimental Medicine and Pharmacology, University of Messina, Messina, Italy (M.C., E.E., S.C.); and Department of Chemistry, Saint Louis University, Saint Louis, Missouri (C.K.A.)
| | - Emanuela Esposito
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (K.B., Y.L., Z.C., T.M.D., L.A.G., J.Z., C.K.A., D.S.); Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, Saint Louis, Missouri (K.B., Y.L., Z.C., T.M.D., L.A.G., J.Z., C.K.A.,D.S.); Department of Clinical and Experimental Medicine and Pharmacology, University of Messina, Messina, Italy (M.C., E.E., S.C.); and Department of Chemistry, Saint Louis University, Saint Louis, Missouri (C.K.A.)
| | - Jinsong Zhang
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (K.B., Y.L., Z.C., T.M.D., L.A.G., J.Z., C.K.A., D.S.); Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, Saint Louis, Missouri (K.B., Y.L., Z.C., T.M.D., L.A.G., J.Z., C.K.A.,D.S.); Department of Clinical and Experimental Medicine and Pharmacology, University of Messina, Messina, Italy (M.C., E.E., S.C.); and Department of Chemistry, Saint Louis University, Saint Louis, Missouri (C.K.A.)
| | - Salvatore Cuzzocrea
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (K.B., Y.L., Z.C., T.M.D., L.A.G., J.Z., C.K.A., D.S.); Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, Saint Louis, Missouri (K.B., Y.L., Z.C., T.M.D., L.A.G., J.Z., C.K.A.,D.S.); Department of Clinical and Experimental Medicine and Pharmacology, University of Messina, Messina, Italy (M.C., E.E., S.C.); and Department of Chemistry, Saint Louis University, Saint Louis, Missouri (C.K.A.)
| | - Christopher Kent Arnatt
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (K.B., Y.L., Z.C., T.M.D., L.A.G., J.Z., C.K.A., D.S.); Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, Saint Louis, Missouri (K.B., Y.L., Z.C., T.M.D., L.A.G., J.Z., C.K.A.,D.S.); Department of Clinical and Experimental Medicine and Pharmacology, University of Messina, Messina, Italy (M.C., E.E., S.C.); and Department of Chemistry, Saint Louis University, Saint Louis, Missouri (C.K.A.)
| | - Daniela Salvemini
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri (K.B., Y.L., Z.C., T.M.D., L.A.G., J.Z., C.K.A., D.S.); Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, Saint Louis, Missouri (K.B., Y.L., Z.C., T.M.D., L.A.G., J.Z., C.K.A.,D.S.); Department of Clinical and Experimental Medicine and Pharmacology, University of Messina, Messina, Italy (M.C., E.E., S.C.); and Department of Chemistry, Saint Louis University, Saint Louis, Missouri (C.K.A.)
| |
Collapse
|
15
|
Mustafa S, Evans S, Barry B, Barratt D, Wang Y, Lin C, Wang X, Hutchinson MR. Toll-Like Receptor 4 in Pain: Bridging Molecules-to-Cells-to-Systems. Handb Exp Pharmacol 2022; 276:239-273. [PMID: 35434749 DOI: 10.1007/164_2022_587] [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] [Indexed: 06/14/2023]
Abstract
Pain impacts the lives of billions of people around the world - both directly and indirectly. It is complex and transcends beyond an unpleasant sensory experience to encompass emotional experiences. To date, there are no successful treatments for sufferers of chronic pain. Although opioids do not provide any benefit to chronic pain sufferers, they are still prescribed, often resulting in more complications such as hyperalgesia and dependence. In order to develop effective and safe medications to manage, and perhaps even treat pain, it is important to evaluate novel contributors to pain pathologies. As such, in this chapter we review the role of Toll-like receptor 4, a receptor of the innate immune system, that continues to gain substantial attention in the field of pain research. Positioned in the nexus of the neuro and immune systems, TLR4 may provide one of the missing pieces in understanding the complexities of pain. Here we consider how TLR4 enables a mechanistical understanding of pain as a multidimensional biopsychosocial state from molecules to cells to systems and back again.
Collapse
Affiliation(s)
- Sanam Mustafa
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia.
- Australian Research Council Centre of Excellence for Nanoscale BioPhotonics, The University of Adelaide, Adelaide, SA, Australia.
| | - Samuel Evans
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
| | - Benjamin Barry
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
| | - Daniel Barratt
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
| | - Yibo Wang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, China
| | - Cong Lin
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, China
| | - Xiaohui Wang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, China
| | - Mark R Hutchinson
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
- Australian Research Council Centre of Excellence for Nanoscale BioPhotonics, The University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
16
|
Abstract
Neuropsychiatric diseases have traditionally been studied from brain, and mind-centric perspectives. However, mounting epidemiological and clinical evidence shows a strong correlation of neuropsychiatric manifestations with immune system activation, suggesting a likely mechanistic interaction between the immune and nervous systems in mediating neuropsychiatric disease. Indeed, immune mediators such as cytokines, antibodies, and complement proteins have been shown to affect various cellular members of the central nervous system in multitudinous ways, such as by modulating neuronal firing rates, inducing cellular apoptosis, or triggering synaptic pruning. These observations have in turn led to the exciting development of clinical therapies aiming to harness this neuro-immune interaction for the treatment of neuropsychiatric disease and symptoms. Besides the clinic, important theoretical fundamentals can be drawn from the immune system and applied to our understanding of the brain and neuropsychiatric disease. These new frameworks could lead to novel insights in the field and further potentiate the development of future therapies to treat neuropsychiatric disease.
Collapse
|
17
|
Tan PH, Ji J, Yeh CC, Ji RR. Interferons in Pain and Infections: Emerging Roles in Neuro-Immune and Neuro-Glial Interactions. Front Immunol 2021; 12:783725. [PMID: 34804074 PMCID: PMC8602180 DOI: 10.3389/fimmu.2021.783725] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 10/19/2021] [Indexed: 12/24/2022] Open
Abstract
Interferons (IFNs) are cytokines that possess antiviral, antiproliferative, and immunomodulatory actions. IFN-α and IFN-β are two major family members of type-I IFNs and are used to treat diseases, including hepatitis and multiple sclerosis. Emerging evidence suggests that type-I IFN receptors (IFNARs) are also expressed by microglia, astrocytes, and neurons in the central and peripheral nervous systems. Apart from canonical transcriptional regulations, IFN-α and IFN-β can rapidly suppress neuronal activity and synaptic transmission via non-genomic regulation, leading to potent analgesia. IFN-γ is the only member of the type-II IFN family and induces central sensitization and microglia activation in persistent pain. We discuss how type-I and type-II IFNs regulate pain and infection via neuro-immune modulations, with special focus on neuroinflammation and neuro-glial interactions. We also highlight distinct roles of type-I IFNs in the peripheral and central nervous system. Insights into IFN signaling in nociceptors and their distinct actions in physiological vs. pathological and acute vs. chronic conditions will improve our treatments of pain after surgeries, traumas, and infections.
Collapse
Affiliation(s)
- Ping-Heng Tan
- Department of Anesthesiology, Chi Mei Medical Center, Tainan City, Taiwan
| | - Jasmine Ji
- Neuroscience Department, Wellesley College, Wellesley, Massachusetts, MA, United States
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC, United States
| | - Chun-Chang Yeh
- Department of Anesthesiology of Tri-Service General Hospital & National Defense Medical Center, Taipei City, Taiwan
| | - Ru-Rong Ji
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC, United States
- Department of Neurobiology, Duke University Medical Center, Durham, NC, United States
- Department of Cell Biology, Duke University Medical Center, Durham, NC, United States
| |
Collapse
|
18
|
Kankowski S, Grothe C, Haastert-Talini K. Neuropathic pain: Spotlighting anatomy, experimental models, mechanisms, and therapeutic aspects. Eur J Neurosci 2021; 54:4475-4496. [PMID: 33942412 DOI: 10.1111/ejn.15266] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 04/19/2021] [Accepted: 04/28/2021] [Indexed: 12/30/2022]
Abstract
The International Association for the Study of Pain defines neuropathic pain as "pain arising as a direct consequence of a lesion or disease affecting the somatosensory system". The associated changes can be observed in the peripheral as well as the central nervous system. The available literature discusses a wide variety of causes as predisposing for the development and amplification of neuropathic pain. Further, key interactions within sensory pathways have been discovered, but no common molecular mechanism leading to neuropathic pain has been identified until now. In the first part of this review, the pain mediating lateral spinothalamic tract is described. Different in vivo models are presented that allow studying trauma-, chemotherapy-, virus-, and diabetes-induced neuropathic pain in rodents. We furthermore discuss approaches to assess neuropathic pain in these models. Second, the current knowledge about cellular and molecular mechanisms suggested to underlie the development of neuropathic pain is presented and discussed. A summary of established therapies that are already applied in the clinic and novel, promising approaches closes the paper. In conclusion, the established animal models are able to emulate the diversity of neuropathic pain observed in the clinics. However, the assessment of neuropathic pain in the presented in vivo models should be improved. The determination of common molecular markers with suitable in vitro models would simplify the assessment of neuropathic pain in vivo. This would furthermore provide insights into common molecular mechanisms of the disease and establish a basis to search for satisfying therapeutic approaches.
Collapse
Affiliation(s)
- Svenja Kankowski
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School (MHH), Hannover, Germany
| | - Claudia Grothe
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School (MHH), Hannover, Germany.,Center for Systems Neuroscience (ZNS) Hannover, Hannover, Germany
| | - Kirsten Haastert-Talini
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School (MHH), Hannover, Germany.,Center for Systems Neuroscience (ZNS) Hannover, Hannover, Germany
| |
Collapse
|
19
|
Gwak YS, Chen G, Abdi S, Kim HK. Calcium-independent phospholipase A2 inhibitor produces an analgesic effect in a rat model of neuropathic pain by reducing central sensitization in the dorsal horn. Neurol Res 2021; 43:683-692. [PMID: 33866950 DOI: 10.1080/01616412.2021.1915079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
OBJECTIVE Phospholipase A2 (PLA2) plays an important role in regulating the production of arachidonic acid and various eicosanoids. The aim of our study was to investigate the analgesic mechanisms of calcium-dependent cytosolic phospholipase A2 and calcium-independent PLA2 (iPLA2) inhibitors in the spinal cord in a rat model of neuropathic pain. METHODS Lumbar 5 spinal nerve ligation was performed in male Sprague-Dawley rats to develop a peripheral neuropathic pain model. Paw withdrawal thresholds in response to von Frey filaments, brush, pressure, and pinch were measured. Lumbar wide dynamic range neuronal firing rates and iPLA2 subtype expression were measured by in vivo extracellular recording and double immunofluorescence staining, respectively. RESULTS In our rat models, oral administration of prednisolone, a non-selective PLA2 inhibitor, and intrathecal injection of bromoenolactone, a iPLA2 inhibitor, significantly increased the ipsilateral hindpaw withdrawal thresholds in response to von Frey filament stimulation, but intrathecal injection of arachidonyl trifluoromethyl ketone, a selective cytosolic PLA2 inhibitor, did not show significant changes. In spinal dorsal horn neurons, bromoenolactone reduced neuronal firing rates in response to withdrawal stimulation and spontaneous firing rates in the ipsilateral side of the spinal dorsal horn. In addition, the expression of iPLA2 was co-localized with astrocytes and neurons on the ipsilateral side of the dorsal horn in rats that underwent spinal nerve ligation. DISCUSSION These data suggest that selective iPLA2 inhibitor produce analgesia in neuropathic rats by reducing central sensitization in the dorsal horn.
Collapse
Affiliation(s)
- Young Seob Gwak
- Department of Anesthesiology and Perioperative Care, University of California, Irvine, CA, USA
| | - Guanxing Chen
- Department of Pain Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Salahadin Abdi
- Department of Pain Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hee Kee Kim
- Department of Pain Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
20
|
STING-ing Pain: How Can Pro-inflammatory Signaling Attenuate Pain? Neurosci Bull 2021; 37:1075-1078. [PMID: 33835401 PMCID: PMC8275733 DOI: 10.1007/s12264-021-00672-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 03/03/2021] [Indexed: 11/04/2022] Open
|
21
|
Increased substance P and synaptic remodeling occur in the trigeminal sensory system with sustained osteoarthritic temporomandibular joint sensitivity. Pain Rep 2021; 6:e911. [PMID: 33977183 PMCID: PMC8104398 DOI: 10.1097/pr9.0000000000000911] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 11/24/2020] [Accepted: 12/18/2020] [Indexed: 12/15/2022] Open
Abstract
Supplemental Digital Content is Available in the Text. Increased substance P and a loss of inhibitory synapses occurs within the brain's trigeminal sensory system with persistent, but not transient, temporomandibular joint sensitivity. Introduction: Temporomandibular joint (TMJ) pain is among the most prevalent musculoskeletal conditions and can result from atypical joint loading. Although TMJ pain is typically self-resolving, 15% of patients develop chronic TMJ pain that is recalcitrant to therapy and may be attributed to changes in pain processing centers. Although TMJ overloading induces pain and osteoarthritis, whether neuronal modifications in the trigeminal sensory system contribute to persistent TMJ pain is unknown. Objective: This study investigates changes in excitatory neuropeptides and synaptic transmission proteins in cases of transient and persistent TMJ sensitivity in a rat model. Methods: Rats underwent repeated jaw loading that produces transient (2N-load) or persistent (3.5N-load) sensitivity. In both groups, immunolabeling was used to assess substance P in the spinal trigeminal nucleus caudalis (Sp5C) and glutamate transporter 1 in the ventroposteriomedial thalamus early after loading. Synaptosomal Western blots were used to measure synaptic proteins in the caudal medulla and thalamus at a later time after loading. Results: Substance P increases transiently in the Sp5C early after loading that induces persistent sensitivity. However, glutamate transporter 1 is unchanged in the ventroposteriomedial thalamus. At a later time, synaptosomal Western blots show loss of the presynaptic tethering protein, synapsin, and the inhibitory scaffolding protein, gephyrin, in the thalamus with persistent, but not transient, sensitivity. No changes are identified in synapsin, phosphorylated synapsin, homer, or gephyrin in the caudal medulla. Conclusions: Substance P in the Sp5C and later loss of inhibitory synapses in the thalamus likely contribute to, or indicate, persistent TMJ pain.
Collapse
|
22
|
Wang ZJ, Yasuhara T. An Examination of Mobile Spinal Cord Stimulators on Treating Parkinson Disease. Brain Circ 2021; 7:8-12. [PMID: 34084970 PMCID: PMC8057101 DOI: 10.4103/bc.bc_6_21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 12/31/2020] [Accepted: 01/03/2021] [Indexed: 12/24/2022] Open
Abstract
In animal models of Parkinson disease (PD), spinal cord stimulation (SCS) exhibits neuroprotective effects. Recent advancements in SCS technology, most importantly mobile stimulators, allow for the conventional limitations of SCS such as limited stimulation time and restricted animal movements to be bypassed, offering potential avenues for improved clinical translation to PD patients. Small devices that could deliver continuous SCS to freely moving parkinsonian rats were shown to significantly improve behavior, preserve neurons and fibers in the substantia Nigra/striatum, reduce microglia infiltration, and increase laminin-positive area of the cerebral cortex. Through possible anti-inflammatory and angiogenic mechanisms, it has been demonstrated that there are behavioral and histological benefits to continuous SCS in a time-dependent manner. This review will discuss the benefits of this technology as well as focus on the limitations of current animal models.
Collapse
Affiliation(s)
- Zhen-Jie Wang
- Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Takao Yasuhara
- Department of Neurological Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| |
Collapse
|
23
|
Zheng J, Ru W, Adolacion JR, Spurgat MS, Liu X, Yuan S, Liang RX, Dong J, Potter AS, Potter SS, Chen K, Chen R, Varadarajan N, Tang SJ. Single-cell RNA-seq analysis reveals compartment-specific heterogeneity and plasticity of microglia. iScience 2021; 24:102186. [PMID: 33718838 PMCID: PMC7921843 DOI: 10.1016/j.isci.2021.102186] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 12/31/2020] [Accepted: 02/09/2021] [Indexed: 12/31/2022] Open
Abstract
Microglia are ubiquitous central nervous system (CNS)-resident macrophages that maintain homeostasis of neural tissues and protect them from pathogen attacks. Yet, their differentiation in different compartments remains elusive. We performed single-cell RNA-seq to compare microglial subtypes in the cortex and the spinal cord. A multi-way comparative analysis was carried out on samples from C57/BL and HIV gp120 transgenic mice at two, four, and eight months of age. The results revealed overlapping but distinct microglial populations in the cortex and the spinal cord. The differential heterogeneity of microglia in these CNS regions was further suggested by their disparity of plasticity in response to life span progression and HIV-1 pathogenic protein gp120. Our findings indicate that microglia in different CNS compartments are adapted to their local environments to fulfill region-specific biological functions.
Collapse
Affiliation(s)
- Junying Zheng
- Department of Neuroscience, Cell Biology, & Anatomy, The University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555, USA
| | - Wenjuan Ru
- Department of Neuroscience, Cell Biology, & Anatomy, The University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555, USA
| | - Jay R. Adolacion
- Department of Chemical & Biomolecular Engineering, University of Houston, Houston, TX 77004, USA
| | - Michael S. Spurgat
- Department of Neuroscience, Cell Biology, & Anatomy, The University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555, USA
| | - Xin Liu
- Department of Neuroscience, Cell Biology, & Anatomy, The University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555, USA
| | - Subo Yuan
- Department of Neuroscience, Cell Biology, & Anatomy, The University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555, USA
| | - Rommel X. Liang
- Department of Pathology, The University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Jianli Dong
- Department of Pathology, The University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Andrew S. Potter
- Division of Developmental Biology, Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - S Steven Potter
- Division of Developmental Biology, Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Ken Chen
- Department of Bioinformatics and Computational Biology, Division of Quantitative Sciences, The University of Texas MD Anderson Cancer Center, Houston 77030, TX, USA
| | - Rui Chen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston 77030, TX, USA
| | - Navin Varadarajan
- Department of Chemical & Biomolecular Engineering, University of Houston, Houston, TX 77004, USA
| | - Shao-Jun Tang
- Department of Neuroscience, Cell Biology, & Anatomy, The University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555, USA
| |
Collapse
|
24
|
Chang C, Liu HK, Yeh CB, Yang ML, Liao WC, Liu CH, Tseng TJ. Cross-Talk of Toll-Like Receptor 5 and Mu-Opioid Receptor Attenuates Chronic Constriction Injury-Induced Mechanical Hyperalgesia through a Protein Kinase C Alpha-Dependent Signaling. Int J Mol Sci 2021; 22:1891. [PMID: 33673008 PMCID: PMC7918001 DOI: 10.3390/ijms22041891] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/30/2021] [Accepted: 02/10/2021] [Indexed: 12/11/2022] Open
Abstract
Recently, Toll-like receptors (TLRs), a family of pattern recognition receptors, are reported as potential modulators for neuropathic pain; however, the desired mechanism is still unexplained. Here, we operated on the sciatic nerve to establish a pre-clinical rodent model of chronic constriction injury (CCI) in Sprague-Dawley rats, which were assigned into CCI and Decompression groups randomly. In Decompression group, the rats were performed with nerve decompression at post-operative week 4. Mechanical hyperalgesia and mechanical allodynia were obviously attenuated after a month. Toll-like receptor 5 (TLR5)-immunoreactive (ir) expression increased in dorsal horn, particularly in the inner part of lamina II. Additionally, substance P (SP) and isolectin B4 (IB4)-ir expressions, rather than calcitonin-gene-related peptide (CGRP)-ir expression, increased in their distinct laminae. Double immunofluorescence proved that increased TLR5-ir expression was co-expressed mainly with IB4-ir expression. Through an intrathecal administration with FLA-ST Ultrapure (a TLR5 agonist, purified flagellin from Salmonella Typhimurium, only the CCI-induced mechanical hyperalgesia was attenuated dose-dependently. Moreover, we confirmed that mu-opioid receptor (MOR) and phospho-protein kinase Cα (pPKCα)-ir expressions but not phospho-protein kinase A RII (pPKA RII)-ir expression, increased in lamina II, where they mostly co-expressed with IB4-ir expression. Go 6976, a potent protein kinase C inhibitor, effectively reversed the FLA-ST Ultrapure- or DAMGO-mediated attenuated trend towards mechanical hyperalgesia by an intrathecal administration in CCI rats. In summary, our current findings suggest that nerve decompression improves CCI-induced mechanical hyperalgesia that might be through the cross-talk of TLR5 and MOR in a PKCα-dependent manner, which opens a novel opportunity for the development of analgesic therapeutics in neuropathic pain.
Collapse
Affiliation(s)
- Ching Chang
- Department of Anatomy, School of Medicine, Chung Shan Medical University, 40201 Taichung, Taiwan; (C.C.); (H.-K.L.); (M.-L.Y.); (W.-C.L.); (C.-H.L.)
| | - Hung-Kai Liu
- Department of Anatomy, School of Medicine, Chung Shan Medical University, 40201 Taichung, Taiwan; (C.C.); (H.-K.L.); (M.-L.Y.); (W.-C.L.); (C.-H.L.)
| | - Chao-Bin Yeh
- Department of Emergency Medicine, Chung Shan Medical University Hospital, 40201 Taichung, Taiwan;
- Department of Emergency Medicine, School of Medicine, Chung Shan Medical University, 40201 Taichung, Taiwan
| | - Ming-Lin Yang
- Department of Anatomy, School of Medicine, Chung Shan Medical University, 40201 Taichung, Taiwan; (C.C.); (H.-K.L.); (M.-L.Y.); (W.-C.L.); (C.-H.L.)
- Department of Medical Education, Chung Shan Medical University Hospital, 40201 Taichung, Taiwan
| | - Wen-Chieh Liao
- Department of Anatomy, School of Medicine, Chung Shan Medical University, 40201 Taichung, Taiwan; (C.C.); (H.-K.L.); (M.-L.Y.); (W.-C.L.); (C.-H.L.)
- Department of Medical Education, Chung Shan Medical University Hospital, 40201 Taichung, Taiwan
| | - Chiung-Hui Liu
- Department of Anatomy, School of Medicine, Chung Shan Medical University, 40201 Taichung, Taiwan; (C.C.); (H.-K.L.); (M.-L.Y.); (W.-C.L.); (C.-H.L.)
- Department of Medical Education, Chung Shan Medical University Hospital, 40201 Taichung, Taiwan
| | - To-Jung Tseng
- Department of Anatomy, School of Medicine, Chung Shan Medical University, 40201 Taichung, Taiwan; (C.C.); (H.-K.L.); (M.-L.Y.); (W.-C.L.); (C.-H.L.)
- Department of Medical Education, Chung Shan Medical University Hospital, 40201 Taichung, Taiwan
| |
Collapse
|
25
|
Liu CC, Lu IC, Wang LK, Chen JY, Li YY, Yang CP, Liu PH, Cheng WJ, Tan PH. Interferon-β suppresses inflammatory pain through activating µ-opioid receptor. Mol Pain 2021; 17:17448069211045211. [PMID: 34517736 PMCID: PMC8642049 DOI: 10.1177/17448069211045211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 08/02/2021] [Accepted: 08/23/2021] [Indexed: 11/16/2022] Open
Abstract
Interferons (IFNs) are cytokines secreted by infected cells that can interfere with viral replication. Besides activating antiviral defenses, type I IFNs also exhibit diverse biological functions. IFN-β has been shown to have a protective effect against neurotoxic and inflammatory insults on neurons. Therefore, we aimed to investigate the possible role of IFN-β in reducing mechanical allodynia caused by Complete Freund's Adjuvant (CFA) injection in rats. We assessed the antinociceptive effect of intrathecal IFN-β in naïve rats and the rats with CFA-induced inflammatory pain. After the behavioral test, the spinal cords of the rats were harvested for western blot and immunohistochemical double staining. We found that intrathecal administration of IFN-β in naïve rats can significantly increase the paw withdrawal threshold and paw withdrawal latency. Further, the intrathecal injection of a neutralizing IFN-β antibody can reduce the paw withdrawal threshold and paw withdrawal latency, suggesting that IFN-β is produced in the spinal cord in normal conditions and serves as a tonic inhibitor of pain. In addition, intrathecal injection of IFN-β at dosages from 1000 U to 10000 U demonstrates a significant transient dose-dependent inhibition of CFA-induced inflammatory pain. This analgesic effect is reversed by intrathecal naloxone, suggesting that IFN-β produces an analgesic effect through central opioid receptor-mediated signaling. Increased expression of phospho-µ-opioid receptors after IFN-β injection was observed on western blot, and immunohistochemical staining showed that µ-opioids co-localized with IFN-α/βR in the dorsal horn of the spinal cord. The findings of this study demonstrate that the analgesic effect of IFN-β is through µ-opioid receptors activation in spial cord.
Collapse
Affiliation(s)
- Chien Cheng Liu
- Department of Anesthesiology, E-Da Hospital/I-Shou University, Kaohsiung City, Taiwan
| | - I Cheng Lu
- Department of Anesthesiology, College of Medicine, Kaohsiung Medical University, Kaohsiung City, Taiwan
| | - Li Kai Wang
- Southern Taiwan University of Science and Technology, Tainan City, Taiwan
- Department of Anesthesiology, Chi Mei Medical Center, Tainan City, Taiwan
| | - Jen Yin Chen
- Department of Anesthesiology, Chi Mei Medical Center, Tainan City, Taiwan
| | - Yu Yu Li
- Department of Anesthesiology, Chi Mei Medical Center, Tainan City, Taiwan
| | - Chih Ping Yang
- Department of Anesthesiology, Chi Mei Medical Center, Tainan City, Taiwan
| | - Ping Hsin Liu
- Department of Anesthesiology, E-Da Hospital/I-Shou University, Kaohsiung City, Taiwan
| | - Wan Jung Cheng
- Department of Anesthesiology, Chi Mei Medical Center, Tainan City, Taiwan
| | - Ping Heng Tan
- Department of Anesthesiology, Chi Mei Medical Center, Tainan City, Taiwan
| |
Collapse
|
26
|
Aw E, Zhang Y, Carroll M. Microglial responses to peripheral type 1 interferon. J Neuroinflammation 2020; 17:340. [PMID: 33183319 PMCID: PMC7659169 DOI: 10.1186/s12974-020-02003-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 10/19/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Interferon α (IFNα) is a cytokine whose production is increased endogenously in response to viral infection and in autoimmune diseases such as systemic lupus erythematosus (SLE). An elevated IFNα signature has been associated with clinically observed neuro-behavioural deficits such as mild cognitive impairment, fatigue, depression and psychosis in these diseases. However, the mechanisms underlying these neuropsychiatric symptoms remain largely unknown, and it is as yet unclear how IFNα signalling might influence central nervous system (CNS) function. Aberrant microglia-mediated synaptic pruning and function has recently been implicated in several neurodegenerative and neuropsychiatric diseases, but whether and how IFNα modulates these functions are not well defined. METHODS Using a model of peripheral IFNα administration, we investigated gene expression changes due to IFNAR signalling in microglia. Bulk RNA sequencing on sorted microglia from wild type and microglia-specific Ifnar1 conditional knockout mice was performed to evaluate IFNα and IFNAR signalling-dependent changes in gene expression. Furthermore, the effects of IFNα on microglia morphology and synapse engulfment were assessed, via immunohistochemistry and flow cytometry. RESULTS We found that IFNα exposure through the periphery induces a unique gene signature in microglia that includes the expected upregulation of multiple interferon-stimulated genes (ISGs), as well as the complement component C4b. We additionally characterized several IFNα-dependent changes in microglial phenotype, including expression of CD45 and CD68, cellular morphology and presynaptic engulfment, that reveal subtle brain region-specific differences. Finally, by specifically knocking down expression of IFNAR1 on microglia, we show that these changes are largely attributable to direct IFNAR signalling on microglia and not from indirect signalling effects through other CNS parenchymal cell types which are capable of IFNα-IFNAR signal transduction. CONCLUSIONS Peripheral IFNα induces unique genetic and phenotypic changes in microglia that are largely dependent on direct signalling through microglial IFNAR. The IFNα-induced upregulation of C4b could play important roles in the context of aberrant synaptic pruning in neuropsychiatric disease.
Collapse
Affiliation(s)
- Ernest Aw
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Division of Medical Sciences, Harvard Medical School, Boston, MA, USA
| | - Yingying Zhang
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
| | - Michael Carroll
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA.
| |
Collapse
|
27
|
Patil MJ, Ru F, Sun H, Wang J, Kolbeck RR, Dong X, Kollarik M, Canning BJ, Undem BJ. Acute activation of bronchopulmonary vagal nociceptors by type I interferons. J Physiol 2020; 598:5541-5554. [PMID: 32924209 DOI: 10.1113/jp280276] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 08/17/2020] [Indexed: 12/14/2022] Open
Abstract
KEY POINTS Type I interferon receptors are expressed by the majority of vagal C-fibre neurons innervating the respiratory tract Interferon alpha and beta acutely and directly activate vagal C-fibers in the airways. The interferon-induced activation of C-fibers occurs secondary to stimulation of type 1 interferon receptors Type 1 interferons may contribute to the symptoms as well as the spread of respiratory viral infections by causing coughing and other defensive reflexes associated with vagal C-fibre activation ABSTRACT: We evaluated the ability of type I interferons to acutely activate airway vagal afferent nerve terminals in mouse lungs. Using single cell RT-PCR of lung-specific vagal neurons we found that IFNAR1 and IFNAR2 were expressed in 70% of the TRPV1-positive neurons (a marker for vagal C-fibre neurons) and 44% of TRPV1-negative neurons. We employed an ex vivo vagal innervated mouse trachea-lung preparation to evaluate the effect of interferons in directly activating airway nerves. Utilizing 2-photon microscopy of the nodose ganglion neurons from Pirt-Cre;R26-GCaMP6s mice we found that applying IFNα or IFNβ to the lungs acutely activated the majority of vagal afferent nerve terminals. When the type 1 interferon receptor, IFNAR1, was blocked with a blocking antibody the response to IFNβ was largely inhibited. The type 2 interferon, IFNγ, also activated airway nerves and this was not inhibited by the IFNAR1 blocking antibody. The Janus kinase inhibitor GLPG0634 (1 μm) virtually abolished the nerve activation caused by IFNβ. Consistent with the activation of vagal afferent C-fibers, infusing IFNβ into the mouse trachea led to defensive breathing reflexes including apneas and gasping. These reflexes were prevented by pretreatment with an IFN type-1 receptor blocking antibody. Finally, using whole cell patch-clamp electrophysiology of lung-specific neurons we found that IFNβ (1000 U ml-1 ) directly depolarized the membrane potential of isolated nodose neurons, in some cases beyond to action potential threshold. This acute non-genomic activation of vagal sensory nerve terminals by interferons may contribute to the incessant coughing that is a hallmark of respiratory viral infections.
Collapse
Affiliation(s)
- Mayur J Patil
- The Johns Hopkins School of Medicine Departments of Medicine, Baltimore, MD
| | - Fei Ru
- The Johns Hopkins School of Medicine Departments of Medicine, Baltimore, MD
| | - Hui Sun
- The Johns Hopkins School of Medicine Departments of Medicine, Baltimore, MD
| | - Jingya Wang
- AstraZeneca BioPharmaceuticals R&D Gaithersburg, MD
| | | | - Xinzhong Dong
- The Johns Hopkins School of Medicine Departments of Neuroscience, Baltimore, MD
| | - Marian Kollarik
- The Johns Hopkins School of Medicine Departments of Medicine, Baltimore, MD
| | - Brendan J Canning
- The Johns Hopkins School of Medicine Departments of Medicine, Baltimore, MD
| | - Bradley J Undem
- The Johns Hopkins School of Medicine Departments of Medicine, Baltimore, MD
| |
Collapse
|
28
|
Kuwahara K, Sasaki T, Yasuhara T, Kameda M, Okazaki Y, Hosomoto K, Kin I, Okazaki M, Yabuno S, Kawauchi S, Tomita Y, Umakoshi M, Kin K, Morimoto J, Lee JY, Tajiri N, Borlongan CV, Date I. Long-Term Continuous Cervical Spinal Cord Stimulation Exerts Neuroprotective Effects in Experimental Parkinson's Disease. Front Aging Neurosci 2020; 12:164. [PMID: 32612523 PMCID: PMC7309445 DOI: 10.3389/fnagi.2020.00164] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Accepted: 05/12/2020] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Spinal cord stimulation (SCS) exerts neuroprotective effects in animal models of Parkinson's disease (PD). Conventional stimulation techniques entail limited stimulation time and restricted movement of animals, warranting the need for optimizing the SCS regimen to address the progressive nature of the disease and to improve its clinical translation to PD patients. OBJECTIVE Recognizing the limitations of conventional stimulation, we now investigated the effects of continuous SCS in freely moving parkinsonian rats. METHODS We developed a small device that could deliver continuous SCS. At the start of the experiment, thirty female Sprague-Dawley rats received the dopamine (DA)-depleting neurotoxin, 6-hydroxydopamine, into the right striatum. The SCS device was fixed below the shoulder area of the back of the animal, and a line from this device was passed under the skin to an electrode that was then implanted epidurally over the dorsal column. The rats were divided into three groups: control, 8-h stimulation, and 24-h stimulation, and behaviorally tested then euthanized for immunohistochemical analysis. RESULTS The 8- and 24-h stimulation groups displayed significant behavioral improvement compared to the control group. Both SCS-stimulated groups exhibited significantly preserved tyrosine hydroxylase (TH)-positive fibers and neurons in the striatum and substantia nigra pars compacta (SNc), respectively, compared to the control group. Notably, the 24-h stimulation group showed significantly pronounced preservation of the striatal TH-positive fibers compared to the 8-h stimulation group. Moreover, the 24-h group demonstrated significantly reduced number of microglia in the striatum and SNc and increased laminin-positive area of the cerebral cortex compared to the control group. CONCLUSIONS This study demonstrated the behavioral and histological benefits of continuous SCS in a time-dependent manner in freely moving PD animals, possibly mediated by anti-inflammatory and angiogenic mechanisms.
Collapse
Affiliation(s)
- Ken Kuwahara
- Department of Neurological Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Tatsuya Sasaki
- Department of Neurological Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Takao Yasuhara
- Department of Neurological Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Masahiro Kameda
- Department of Neurological Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Yosuke Okazaki
- Department of Neurological Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Kakeru Hosomoto
- Department of Neurological Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Ittetsu Kin
- Department of Neurological Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Mihoko Okazaki
- Department of Neurological Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Satoru Yabuno
- Department of Neurological Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Satoshi Kawauchi
- Department of Neurological Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Yousuke Tomita
- Department of Neurological Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Michiari Umakoshi
- Department of Neurological Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Kyohei Kin
- Department of Neurological Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Jun Morimoto
- Department of Neurological Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Jea-Young Lee
- Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Naoki Tajiri
- Department of Neurophysiology and Brain Science, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| | - Cesar V. Borlongan
- Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Isao Date
- Department of Neurological Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| |
Collapse
|
29
|
Type I Interferons Act Directly on Nociceptors to Produce Pain Sensitization: Implications for Viral Infection-Induced Pain. J Neurosci 2020; 40:3517-3532. [PMID: 32245829 DOI: 10.1523/jneurosci.3055-19.2020] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 03/04/2020] [Accepted: 03/19/2020] [Indexed: 12/25/2022] Open
Abstract
One of the first signs of viral infection is body-wide aches and pain. Although this type of pain usually subsides, at the extreme, viral infections can induce painful neuropathies that can last for decades. Neither of these types of pain sensitization is well understood. A key part of the response to viral infection is production of interferons (IFNs), which then activate their specific receptors (IFNRs) resulting in downstream activation of cellular signaling and a variety of physiological responses. We sought to understand how type I IFNs (IFN-α and IFN-β) might act directly on nociceptors in the dorsal root ganglion (DRG) to cause pain sensitization. We demonstrate that type I IFNRs are expressed in small/medium DRG neurons and that their activation produces neuronal hyper-excitability and mechanical pain in mice. Type I IFNs stimulate JAK/STAT signaling in DRG neurons but this does not apparently result in PKR-eIF2α activation that normally induces an anti-viral response by limiting mRNA translation. Rather, type I IFNs stimulate MNK-mediated eIF4E phosphorylation in DRG neurons to promote pain hypersensitivity. Endogenous release of type I IFNs with the double-stranded RNA mimetic poly(I:C) likewise produces pain hypersensitivity that is blunted in mice lacking MNK-eIF4E signaling. Our findings reveal mechanisms through which type I IFNs cause nociceptor sensitization with implications for understanding how viral infections promote pain and can lead to neuropathies.SIGNIFICANCE STATEMENT It is increasingly understood that pathogens interact with nociceptors to alert organisms to infection as well as to mount early host defenses. Although specific mechanisms have been discovered for diverse bacterial and fungal pathogens, mechanisms engaged by viruses have remained elusive. Here we show that type I interferons, one of the first mediators produced by viral infection, act directly on nociceptors to produce pain sensitization. Type I interferons act via a specific signaling pathway (MNK-eIF4E signaling), which is known to produce nociceptor sensitization in inflammatory and neuropathic pain conditions. Our work reveals a mechanism through which viral infections cause heightened pain sensitivity.
Collapse
|
30
|
Herzberg D, Strobel P, Ramirez-Reveco A, Werner M, Bustamante H. Chronic Inflammatory Lameness Increases Cytokine Concentration in the Spinal Cord of Dairy Cows. Front Vet Sci 2020; 7:125. [PMID: 32185190 PMCID: PMC7058553 DOI: 10.3389/fvets.2020.00125] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 02/19/2020] [Indexed: 12/20/2022] Open
Abstract
Lameness in dairy cows is an extremely painful multifactorial condition that affects the welfare of animals and economically impacts the dairy industry worldwide. The aim of this study was to determine the profile of cytokines in the spinal cord dorsal horn of dairy cows with painful chronic inflammatory lameness. Concentrations of 10 cytokines were measured in the spinal cord of seven adult dairy cows with chronic lameness and seven adult dairy cows with no lameness. In all cows lameness was evaluated using a mobility scoring system and registered accordingly. Immediately after euthanasia the spinal cord was removed and 20 cm of lumbar segments (L2–L5) were obtained. After dorsal horn removal and processing, cytokine quantification of tumor necrosis factor-alpha (TNF-α), interleukin-1alpha (IL-1α), interleukin 13 (IL-13), chemokine-10 (CXCL10/IP-10), chemokine-9 (CXCL9/MIG), interferon-alpha (IFN-α), interferon-gamma (IFN-γ), interleukin-21 (IL-21), interleukin-36ra (IL-36ra), and macrophage inflammatory protein-1 beta (MIP-1β) was performed using a multiplex array. Lame cows had higher concentrations of TNF-α, IL-1-α, IL-13, CXCL10, CXCL9, IFN-α, and IFN-γ in their dorsal horn compared to non-lame cows, while IL-21 concentration was decreased. No differences in IL-36ra and MIP-1β concentrations between lame and non-lame cows were observed. Painful chronic inflammation of the hoof in dairy cows leads to a marked increase in cytokine concentration in the dorsal horn of the spinal cord, which could represent a state of neuroinflammation of the Central Nervous System (CNS).
Collapse
Affiliation(s)
- Daniel Herzberg
- Faculty of Veterinary Sciences, Graduate School, Universidad Austral de Chile, Valdivia, Chile
| | - Pablo Strobel
- Faculty of Veterinary Sciences, Animal Science Institute, Universidad Austral de Chile, Valdivia, Chile
| | - Alfredo Ramirez-Reveco
- Faculty of Veterinary Sciences, Animal Science Institute, Universidad Austral de Chile, Valdivia, Chile
| | - Marianne Werner
- Faculty of Veterinary Sciences, Animal Science Institute, Universidad Austral de Chile, Valdivia, Chile
| | - Hedie Bustamante
- Faculty of Veterinary Sciences, Veterinary Clinical Sciences Institute, Universidad Austral de Chile, Valdivia, Chile
| |
Collapse
|
31
|
Gonçalves dos Santos G, Delay L, Yaksh TL, Corr M. Neuraxial Cytokines in Pain States. Front Immunol 2020; 10:3061. [PMID: 32047493 PMCID: PMC6997465 DOI: 10.3389/fimmu.2019.03061] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 12/16/2019] [Indexed: 12/11/2022] Open
Abstract
A high-intensity potentially tissue-injuring stimulus generates a homotopic response to escape the stimulus and is associated with an affective phenotype considered to represent pain. In the face of tissue or nerve injury, the afferent encoding systems display robust changes in the input-output function, leading to an ongoing sensation reported as painful and sensitization of the nociceptors such that an enhanced pain state is reported for a given somatic or visceral stimulus. Our understanding of the mechanisms underlying this non-linear processing of nociceptive stimuli has led to our appreciation of the role played by the functional interactions of neural and immune signaling systems in pain phenotypes. In pathological states, neural systems interact with the immune system through the actions of a variety of soluble mediators, including cytokines. Cytokines are recognized as important mediators of inflammatory and neuropathic pain, supporting system sensitization and the development of a persistent pathologic pain. Cytokines can induce a facilitation of nociceptive processing at all levels of the neuraxis including supraspinal centers where nociceptive input evokes an affective component of the pain state. We review here several key proinflammatory and anti-inflammatory cytokines/chemokines and explore their underlying actions at four levels of neuronal organization: (1) peripheral nociceptor termini; (2) dorsal root ganglia; (3) spinal cord; and (4) supraspinal areas. Thus, current thinking suggests that cytokines by this action throughout the neuraxis play key roles in the induction of pain and the maintenance of the facilitated states of pain behavior generated by tissue injury/inflammation and nerve injury.
Collapse
Affiliation(s)
| | - Lauriane Delay
- Department of Anesthesiology, University of California, San Diego, La Jolla, CA, United States
| | - Tony L. Yaksh
- Department of Anesthesiology, University of California, San Diego, La Jolla, CA, United States
| | - Maripat Corr
- Division of Rheumatology, Allergy and Immunology, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
32
|
Deftu AF, Suter MR. Glia and Pain in Spinal Cord. THE SENSES: A COMPREHENSIVE REFERENCE 2020:235-248. [DOI: 10.1016/b978-0-12-809324-5.24214-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
33
|
Liu S, Karaganis S, Mo RF, Li XX, Wen RX, Song XJ. IFNβ Treatment Inhibits Nerve Injury-induced Mechanical Allodynia and MAPK Signaling By Activating ISG15 in Mouse Spinal Cord. THE JOURNAL OF PAIN 2019; 21:836-847. [PMID: 31785403 DOI: 10.1016/j.jpain.2019.11.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 10/15/2019] [Accepted: 11/11/2019] [Indexed: 01/10/2023]
Abstract
Neuropathic pain is difficult to treat and remains a major clinical challenge worldwide. While the mechanisms which underlie the development of neuropathic pain are incompletely understood, interferon signaling by the immune system is known to play a role. Here, we demonstrate a role for interferon β (IFNβ) in attenuating mechanical allodynia induced by the spared nerve injury in mice. The results show that intrathecal administration of IFNβ (dosages up to 5,000 U) produces significant, transient, and dose-dependent attenuation of mechanical allodynia without observable effects on motor activity or feeding behavior, as is common with IFN administration. This analgesic effect is mediated by the ubiquitin-like protein interferon-stimulated gene 15 (ISG15), which is potently induced within the spinal cord following intrathecal delivery of IFNβ. Both free and conjugated ISG15 are elevated following IFNβ treatment, and this effect is increased in UBP43-/- mice lacking a key deconjugating enzyme. The IFNβ-mediated analgesia reduces MAPK signaling activation following nerve injury, and this effect requires induction of ISG15. These findings highlight a new role for IFNβ, ISG15, and MAPK signaling in immunomodulation of neuropathic pain and may lead to new therapeutic possibilities. PERSPECTIVE: Neuropathic pain is frequently intractable in a clinical setting, and new treatment options are needed. Characterizing the antinociceptive potential of IFNβ and the associated downstream signaling pathways in preclinical models may lead to the development of new therapeutic options for debilitating neuropathies.
Collapse
Affiliation(s)
- Su Liu
- SUSTech Center for Pain Medicine, Medical School, Southern University of Science and Technology, Shenzhen, Guangdong, China; Department of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Stephen Karaganis
- SUSTech Center for Pain Medicine, Medical School, Southern University of Science and Technology, Shenzhen, Guangdong, China; Department of Life, Earth and Environmental Sciences, West Texas A&M University, Amarillo, Texas
| | - Ru-Fan Mo
- SUSTech Center for Pain Medicine, Medical School, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Xiao-Xiao Li
- Department of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Ruo-Xin Wen
- SUSTech Center for Pain Medicine, Medical School, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Xue-Jun Song
- SUSTech Center for Pain Medicine, Medical School, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| |
Collapse
|
34
|
Fitzgibbon M, Kerr DM, Henry RJ, Finn DP, Roche M. Endocannabinoid modulation of inflammatory hyperalgesia in the IFN-α mouse model of depression. Brain Behav Immun 2019; 82:372-381. [PMID: 31505257 DOI: 10.1016/j.bbi.2019.09.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 08/15/2019] [Accepted: 09/05/2019] [Indexed: 12/31/2022] Open
Abstract
Depression is a well-recognised effect of long-term treatment with interferon-alpha (IFN-α), a widely used treatment for chronic viral hepatitis and malignancy. In addition to the emotional disturbances, high incidences of painful symptoms such as headache and joint pain have also been reported following IFN-α treatment. The endocannabinoid system plays an important role in emotional and nociceptive processing, however it is unknown whether repeated IFN-α administration induces alterations in this system. The present study investigated nociceptive responding in the IFN-α-induced mouse model of depression and associated changes in the endocannabinoid system. Furthermore, the effects of modulating peripheral endocannabinoid tone on inflammatory pain-related behaviour in the IFN-α model was examined. Repeated IFN-α administration (8000 IU/g/day) to male C57/Bl6 mice increased immobility in the forced swim test and reduced sucrose preference, without altering body weight gain or locomotor activity, confirming development of the depressive-like phenotype. There was no effect of repeated IFN-α administration on latency to respond in the hot plate test on day 4 or 7 of treatment, however, formalin-evoked nociceptive behaviour was significantly increased in IFN-α treated mice following 8 days of IFN-α administration. 2-Arachidonoyl glycerol (2-AG) levels in the periaqueductal grey (PAG) and rostroventromedial medulla (RVM), and anandamide (AEA) levels in the RVM, were significantly increased in IFN-α-, but not saline-, treated mice following formalin administration. There was no change in endocannabinoid levels in the prefrontal cortex, spinal cord or paw tissue between saline- or IFNα-treated mice in the presence or absence of formalin. Furthermore, repeated IFN-α and/or formalin administration did not alter mRNA expression of genes encoding the endocannabinoid catabolic enzymes (fatty acid amide hydrolase or monoacylglycerol lipase) or endocannabinoid receptor targets (CB1, CB2 or PPARs) in the brain, spinal cord or paw tissue. Intra plantar administration of PF3845 (1 μg/10 μl) or MJN110 (1 μg/10 μl), inhibitors of AEA and 2-AG catabolism respectively, attenuated formalin-evoked hyperalgesia in IFN-α, but not saline-, treated mice. In summary, increasing peripheral endocannabinoid tone attenuates inflammatory hyperalgesia induced following repeated IFN-α administration. These data provide support for the endocannabinoid system in mediating and modulating heightened pain responding associated with IFNα-induced depression.
Collapse
Affiliation(s)
- Marie Fitzgibbon
- Physiology, School of Medicine, National University of Ireland, Galway, Ireland
| | - Daniel M Kerr
- Physiology, School of Medicine, National University of Ireland, Galway, Ireland; Pharmacology and Therapeutics, School of Medicine, National University of Ireland, Galway, Ireland
| | - Rebecca J Henry
- Physiology, School of Medicine, National University of Ireland, Galway, Ireland
| | - David P Finn
- Pharmacology and Therapeutics, School of Medicine, National University of Ireland, Galway, Ireland; NCBES Centre for Pain Research and Galway Neuroscience Centre, National University of Ireland, Galway, Ireland
| | - Michelle Roche
- Physiology, School of Medicine, National University of Ireland, Galway, Ireland; NCBES Centre for Pain Research and Galway Neuroscience Centre, National University of Ireland, Galway, Ireland.
| |
Collapse
|
35
|
Laval K, Van Cleemput J, Vernejoul JB, Enquist LW. Alphaherpesvirus infection of mice primes PNS neurons to an inflammatory state regulated by TLR2 and type I IFN signaling. PLoS Pathog 2019; 15:e1008087. [PMID: 31675371 PMCID: PMC6824567 DOI: 10.1371/journal.ppat.1008087] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 09/17/2019] [Indexed: 12/31/2022] Open
Abstract
Pseudorabies virus (PRV), an alphaherpesvirus closely related to Varicella-Zoster virus (VZV) and Herpes simplex type 1 (HSV1) infects mucosa epithelia and the peripheral nervous system (PNS) of its host. We previously demonstrated that PRV infection induces a specific and lethal inflammatory response, contributing to severe neuropathy in mice. So far, the mechanisms that initiate this neuroinflammation remain unknown. Using a mouse footpad inoculation model, we found that PRV infection rapidly and simultaneously induces high G-CSF and IL-6 levels in several mouse tissues, including the footpad, PNS and central nervous system (CNS) tissues. Interestingly, this global increase occurred before PRV had replicated in dorsal root ganglia (DRGs) neurons and also was independent of systemic inflammation. These high G-CSF and IL-6 levels were not caused by neutrophil infiltration in PRV infected tissues, as we did not detect any neutrophils. Efficient PRV replication and spread in the footpad was sufficient to activate DRGs to produce cytokines. Finally, by using knockout mice, we demonstrated that TLR2 and IFN type I play crucial roles in modulating the early neuroinflammatory response and clinical outcome of PRV infection in mice. Overall, these results give new insights into the initiation of virus-induced neuroinflammation during herpesvirus infections. Herpesviruses are major pathogens worldwide. Pseudorabies virus (PRV) is an alphaherpesvirus related to varicella-zoster virus (VZV) and herpes simplex virus type 1 (HSV1). The natural host is the pig, but PRV can infect most mammals. In these non-natural hosts, the virus causes a severe pruritus called the ‘mad itch’. Interestingly, PRV infects the peripheral nervous system (PNS) and induces a specific and lethal inflammatory response in mice, yet little is know about how this neuroinflammatory response is initiated. In this study, we demonstrated for the first time how PNS neurons tightly regulate the inflammatory response during PRV infection and contribute to severe clinical outcome in mice. Our work provides new insights into the process of alphaherpesvirus-induced neuropathies, leading to the development of innovative therapeutic strategies.
Collapse
Affiliation(s)
- Kathlyn Laval
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
- * E-mail:
| | - Jolien Van Cleemput
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
| | - Jonah B. Vernejoul
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
| | - Lynn W. Enquist
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
| |
Collapse
|
36
|
Abstract
Astrocytes are critical for maintaining the homeostasis of the CNS. Increasing evidence suggests that a number of neurological and neuropsychiatric disorders, including chronic pain, may result from astrocyte 'gliopathy'. Indeed, in recent years there has been substantial progress in our understanding of how astrocytes can regulate nociceptive synaptic transmission via neuronal-glial and glial-glial cell interactions, as well as the involvement of spinal and supraspinal astrocytes in the modulation of pain signalling and the maintenance of neuropathic pain. A role of astrocytes in the pathogenesis of chronic itch is also emerging. These developments suggest that targeting the specific pathways that are responsible for astrogliopathy may represent a novel approach to develop therapies for chronic pain and chronic itch.
Collapse
|
37
|
Li P, Zhao G, Ding Y, Wang T, Flores J, Ocak U, Wu P, Zhang T, Mo J, Zhang JH, Tang J. Rh-IFN-α attenuates neuroinflammation and improves neurological function by inhibiting NF-κB through JAK1-STAT1/TRAF3 pathway in an experimental GMH rat model. Brain Behav Immun 2019; 79:174-185. [PMID: 30711510 PMCID: PMC6591046 DOI: 10.1016/j.bbi.2019.01.028] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 01/04/2019] [Accepted: 01/30/2019] [Indexed: 02/07/2023] Open
Abstract
Neuroinflammation occurs after germinal matrix hemorrhage (GMH) and induces secondary brain injury. Interferon-α (IFN-α) has been shown to exert anti-inflammatory effects in infectious diseases via activating IFNAR and its downstream signaling. We aimed to investigate the anti-inflammatory effects of Recombinant human IFN-α (rh-IFN-α) and the underlying mechanisms in a rat GMH model. Two hundred and eighteen P7 rat pups of both sexes were subjected to GMH by an intraparenchymal injection of bacterial collagenase. Rh-IFN-α was administered intraperitoneally. Small interfering RNA (siRNA) of IFNAR, and siRNA of tumor necrosis factor receptor associated factor 3 (TRAF3) were administered through intracerebroventricular (i.c.v.) injections. JAK1 inhibitor ruxolitinib was given by oral lavage. Post-GMH evaluation included neurobehavioral function, Nissl staining, Western blot analysis, and immunofluorescence. Our results showed that endogenous IFN-α and phosphorylated IFNAR levels were increased after GMH. Administration of rh-IFN-α improved neurological functions, attenuated neuroinflammation, inhibited microglial activation, and ameliorated post-hemorrhagic hydrocephalus after GMH. These observations were concomitant with IFNAR activation, increased expression of phosphorylated JAK1, phosphorylated STAT1 and TRAF3, and decreased levels of phosphorylated NF-κB, IL-6 and TNF-α. Specifically, knockdown of IFNAR, JAK1 and TRAF3 abolished the protective effects of rh-IFN-α. In conclusion, our findings demonstrated that rh-IFN-α treatment attenuated neuroinflammation, neurological deficits and hydrocephalus formation through inhibiting microglial activation after GMH, which might be mediated by IFNAR/JAK1-STAT1/TRAF3/NF-κB signaling pathway. Rh-IFN-α may be a promising therapeutic agent to attenuate brain injury via its anti-inflammatory effect.
Collapse
Affiliation(s)
- Peng Li
- Department of Physiology and Pharmacology, Basic Science, School of Medicine, Loma Linda University, Loma Linda, CA 92354, United States; Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou 510180, China; Guangzhou First People's Hospital, the Second Affiliated Hospital of South China University of Technology, Guangzhou 510180, China
| | - Gang Zhao
- Department of Physiology and Pharmacology, Basic Science, School of Medicine, Loma Linda University, Loma Linda, CA 92354, United States; Department of Emergency Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming 650101, China; Traumatic Research Center of Yunnan Province, Kunming 650101, China
| | - Yan Ding
- Department of Physiology and Pharmacology, Basic Science, School of Medicine, Loma Linda University, Loma Linda, CA 92354, United States
| | - Tianyi Wang
- Department of Physiology and Pharmacology, Basic Science, School of Medicine, Loma Linda University, Loma Linda, CA 92354, United States
| | - Jerry Flores
- Department of Physiology and Pharmacology, Basic Science, School of Medicine, Loma Linda University, Loma Linda, CA 92354, United States
| | - Umut Ocak
- Department of Physiology and Pharmacology, Basic Science, School of Medicine, Loma Linda University, Loma Linda, CA 92354, United States
| | - Pei Wu
- Department of Physiology and Pharmacology, Basic Science, School of Medicine, Loma Linda University, Loma Linda, CA 92354, United States
| | - Tongyu Zhang
- Department of Physiology and Pharmacology, Basic Science, School of Medicine, Loma Linda University, Loma Linda, CA 92354, United States
| | - Jun Mo
- Department of Physiology and Pharmacology, Basic Science, School of Medicine, Loma Linda University, Loma Linda, CA 92354, United States
| | - John H Zhang
- Department of Physiology and Pharmacology, Basic Science, School of Medicine, Loma Linda University, Loma Linda, CA 92354, United States; Departments of Anesthesiology, Neurosurgery and Neurology, Loma Linda University School of Medicine, Loma Linda, CA 92354, United States
| | - Jiping Tang
- Department of Physiology and Pharmacology, Basic Science, School of Medicine, Loma Linda University, Loma Linda, CA 92354, United States.
| |
Collapse
|
38
|
Nelson NA, Wang X, Cook D, Carey EM, Nimmerjahn A. Imaging spinal cord activity in behaving animals. Exp Neurol 2019; 320:112974. [PMID: 31175843 DOI: 10.1016/j.expneurol.2019.112974] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 06/02/2019] [Accepted: 06/04/2019] [Indexed: 01/06/2023]
Abstract
The spinal cord is the primary neurological link between the brain and peripheral organs. How important it is in everyday life is apparent in patients with spinal cord injury or motoneuron disease, who have dramatically reduced musculoskeletal control or capacity to sense their environment. Despite its crucial role in sensory and motor processing little is known about the cellular and molecular signaling events that underlie spinal cord function under naturalistic conditions. While genetic, electrophysiological, pharmacological, and circuit tracing studies have revealed important roles for different molecularly defined neurons, these approaches insufficiently describe the moment-to-moment neuronal and non-neuronal activity patterns that underlie sensory-guided motor behaviors in health and disease. The recent development of imaging methods for real-time interrogation of cellular activity in the spinal cord of behaving mice has removed longstanding technical obstacles to spinal cord research and allowed new insight into how different cell types encode sensory information from mechanoreceptors and nociceptors in the skin. Here, we review the current state-of-the-art in interrogating cellular and microcircuit function in the spinal cord of behaving mammals and discuss current opportunities and technological challenges.
Collapse
Affiliation(s)
- Nicholas A Nelson
- Waitt Advanced Biophotonics Center, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA; Biologial Sciences Graduate Program, University of California, San Diego, La Jolla, CA 92037, USA
| | - Xiang Wang
- Waitt Advanced Biophotonics Center, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Daniela Cook
- Waitt Advanced Biophotonics Center, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Erin M Carey
- Waitt Advanced Biophotonics Center, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Axel Nimmerjahn
- Waitt Advanced Biophotonics Center, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|
39
|
Associations of cigarette smoking with disease phenotype and type I interferon expression in primary Sjögren’s syndrome. Rheumatol Int 2019; 39:1575-1584. [DOI: 10.1007/s00296-019-04335-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 05/23/2019] [Indexed: 12/31/2022]
|
40
|
Guo SH, Lin JP, Huang LE, Yang Y, Chen CQ, Li NN, Su MY, Zhao X, Zhu SM, Yao YX. Silencing of spinal Trpv1 attenuates neuropathic pain in rats by inhibiting CAMKII expression and ERK2 phosphorylation. Sci Rep 2019; 9:2769. [PMID: 30808963 PMCID: PMC6391380 DOI: 10.1038/s41598-019-39184-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 01/14/2019] [Indexed: 12/15/2022] Open
Abstract
Accumulating evidence suggests a potential role of transient receptor potential vanilloid 1 (TRPV1) channels in inflammatory and cancer-related pain. However, the role of TRPV1 in the maintenance of neuropathic pain remains elusive. The current study investigated the effects of transient Trpv1 gene silencing using a small interference RNA (siRNA) on neuropathic pain induced by chronic constriction injury (CCI) of the sciatic nerve in rats. Seven days after CCI, the TRPV1 siRNA was intrathecally administered (5 µg/15 µl, once daily for 2 days). TRPV1 and Ca2+/calmodulin-dependent protein kinase II (CAMKII) expression and extracellular signal-regulated kinase (ERK) phosphorylation in the spinal cord were detected using western blotting. The thresholds to mechanical and thermal stimuli were determined before and after intrathecal TRPV1 siRNA administration. TRPV1 and CAMKII expression and ERK2 phosphorylation in the spinal cord were upregulated after CCI. Intrathecal administration of the TRPV1 siRNA not only attenuated behavioural hyperalgesia but also reduced the expression of TRPV1 and CAMKII, as well as ERK2 phosphorylation. Based on these results, silencing of the TRPV1 gene in the spinal cord attenuates the maintenance of neuropathic pain by inhibiting CAMKII/ERK2 activation and suggests that TRPV1 represents a potential target in pain therapy.
Collapse
Affiliation(s)
- Shao-Hui Guo
- Department of Anaesthesia, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, P. R. China
| | - Jia-Piao Lin
- Department of Anaesthesia, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, P. R. China
| | - Ling-Er Huang
- Department of Anaesthesia, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, P. R. China
| | - Yan Yang
- Zhejiang University School of Medicine, Centre for Neuroscience, Hangzhou, 310016, P. R. China
| | - Chao-Qin Chen
- Department of Anaesthesia, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, P. R. China
| | - Na-Na Li
- Department of Anaesthesia, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, P. R. China
| | - Meng-Yun Su
- Department of Anaesthesia, Shulan (Hangzhou) Hospital, Hangzhou, 310022, P. R. China
| | - Xian Zhao
- Department of Anaesthesia, Shulan (Hangzhou) Hospital, Hangzhou, 310022, P. R. China
| | - Sheng-Mei Zhu
- Department of Anaesthesia, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, P. R. China
| | - Yong-Xing Yao
- Department of Anaesthesia, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, P. R. China.
| |
Collapse
|
41
|
Woller SA, Ocheltree C, Wong SY, Bui A, Fujita Y, Gonçalves Dos Santos G, Yaksh TL, Corr M. Neuraxial TNF and IFN-beta co-modulate persistent allodynia in arthritic mice. Brain Behav Immun 2019; 76:151-158. [PMID: 30465880 PMCID: PMC6396982 DOI: 10.1016/j.bbi.2018.11.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Revised: 11/03/2018] [Accepted: 11/18/2018] [Indexed: 02/06/2023] Open
Abstract
In rheumatoid arthritis, joint pain can persist despite resolution of swelling. Similarly, in the murine K/BxN serum transfer model, a persistent tactile allodynia is observed after the resolution of joint inflammation (post-inflammatory pain) in male mice. Here, we found female wild type (WT) mice show inflammatory, but reduced post-inflammatory tactile allodynia. The transition to the post-inflammatory phenotype is dependent on TLR4 signaling. At the spinal level, we found differences in TNF and IFNβ mRNA expression in WT and TLR4 deficient males. In wild type male and female mice, there is differential temporal spinal expression of TNF and IFNβ. In WT males, blockade of TNF or administration of IFNβ was insufficient to affect the persistent allodynia. However, co-administration of intrathecal (IT) IFNβ and anti-TNF antibodies in male WT mice permanently reversed tactile allodynia. IT IFNβ treatment induces expression of anti-inflammatory proteins, contributing to the beneficial effect. Together, these experiments illustrated differences in the transition to chronic tactile allodynia in male and female animals and the complexities of effective pharmacologic interventions.
Collapse
Affiliation(s)
- Sarah A Woller
- Departments of Anesthesiology, University of California, San Diego, La Jolla, CA, USA
| | - Cody Ocheltree
- Division of Rheumatology, Allergy and Immunology, University of California, San Diego, La Jolla, CA, USA
| | - Stephanie Y Wong
- Division of Rheumatology, Allergy and Immunology, University of California, San Diego, La Jolla, CA, USA
| | - Anthony Bui
- Division of Rheumatology, Allergy and Immunology, University of California, San Diego, La Jolla, CA, USA
| | - Yuya Fujita
- Division of Rheumatology, Allergy and Immunology, University of California, San Diego, La Jolla, CA, USA
| | | | - Tony L Yaksh
- Departments of Anesthesiology, University of California, San Diego, La Jolla, CA, USA
| | - Maripat Corr
- Division of Rheumatology, Allergy and Immunology, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
42
|
Chamessian A, Young M, Qadri Y, Berta T, Ji RR, Van de Ven T. Transcriptional Profiling of Somatostatin Interneurons in the Spinal Dorsal Horn. Sci Rep 2018; 8:6809. [PMID: 29717160 PMCID: PMC5931607 DOI: 10.1038/s41598-018-25110-7] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 04/13/2018] [Indexed: 01/08/2023] Open
Abstract
The spinal dorsal horn (SDH) is comprised of distinct neuronal populations that process different somatosensory modalities. Somatostatin (SST)-expressing interneurons in the SDH have been implicated specifically in mediating mechanical pain. Identifying the transcriptomic profile of SST neurons could elucidate the unique genetic features of this population and enable selective analgesic targeting. To that end, we combined the Isolation of Nuclei Tagged in Specific Cell Types (INTACT) method and Fluorescence Activated Nuclei Sorting (FANS) to capture tagged SST nuclei in the SDH of adult male mice. Using RNA-sequencing (RNA-seq), we uncovered more than 13,000 genes. Differential gene expression analysis revealed more than 900 genes with at least 2-fold enrichment. In addition to many known dorsal horn genes, we identified and validated several novel transcripts from pharmacologically tractable functional classes: Carbonic Anhydrase 12 (Car12), Phosphodiesterase 11 A (Pde11a), and Protease-Activated Receptor 3 (F2rl2). In situ hybridization of these novel genes showed differential expression patterns in the SDH, demonstrating the presence of transcriptionally distinct subpopulations within the SST population. Overall, our findings provide new insights into the gene repertoire of SST dorsal horn neurons and reveal several novel targets for pharmacological modulation of this pain-mediating population and treatment of pathological pain.
Collapse
Affiliation(s)
- Alexander Chamessian
- Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, 27710, USA. .,Medical Scientist Training Program, Duke University School of Medicine, Durham, North Carolina, 27710, USA. .,Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, 27710, USA.
| | - Michael Young
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina, 27710, USA
| | - Yawar Qadri
- Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, 27710, USA
| | - Temugin Berta
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, Ohio, 45267, USA
| | - Ru-Rong Ji
- Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, 27710, USA.,Department of Neurobiology, Duke University Medical Center, Durham, North Carolina, 27710, USA
| | - Thomas Van de Ven
- Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, 27710, USA
| |
Collapse
|
43
|
Peripheral GABA A receptor-mediated signaling facilitates persistent inflammatory hypersensitivity. Neuropharmacology 2018; 135:572-580. [PMID: 29634983 DOI: 10.1016/j.neuropharm.2018.04.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 03/30/2018] [Accepted: 04/06/2018] [Indexed: 01/26/2023]
Abstract
Unlike in the central nervous system (CNS), in the adult peripheral nervous system (PNS), activation of GABAA receptors (GABAAR) is excitatory because of the relatively high concentration of intracellular chloride in these neurons. Indeed, exogenous GABA and muscimol, a GABAAR agonist, exacerbate acute inflammatory hypersensitivity in rodents. However, it remains unclear whether peripheral GABAAR and the endogenous GABA play an important role in persistent inflammatory hypersensitivity. In this study, we thus investigated how peripheral GABAAR affects pain hypersensitivity by using the complete Freund's adjuvant (CFA)-induced persistent inflammatory pain mouse model. We found that intraplantar (i.pl.) administration of GABAAR antagonists, picrotoxin, and 1(S),9(R)-(-)-bicuculline methiodide significantly inhibited both spontaneous nociceptive (paw licking and flinching) behavior and mechanical hypersensitivity in CFA-injected mice at day 3 (D3), but not in naïve mice. Interestingly, CFA-induced mechanical hypersensitivity was significantly reversed by anti-GABA antibody (anti-GABA, i.pl.). In addition, RT-qPCR revealed that glutamate decarboxylase Gad1 (GAD 67) and Gad2 (GAD 65) mRNA expression was also upregulated in the ipsilateral hind paw of CFA-injected mice at D3. Finally, 5α-pregnan-3α-ol-20-one (3α,5α-THP), a selective positive allosteric modulator of GABAAR, produced mechanical hypersensitivity in naïve mice in a dose-dependent manner. Taken together, our results indicate that peripheral GABAAR and endogenous GABA, possibly produced by the inflamed tissue, potentiate CFA-induced persistent inflammatory hypersensitivity, suggesting that they can be used as a therapeutic target for alleviating inflammatory pain.
Collapse
|
44
|
Abstract
One of the fundamental mechanisms whereby the innate immune system coordinates inflammatory signal transduction is through Toll-like receptors (TLRs), which function to protect and defend the host organism by initiating inflammatory signaling cascades in response to tissue damage or injury. TLRs are positioned at the neuroimmune interface, and accumulating evidence suggests that the inflammatory consequences of TLR activation on glia (including microglia and astrocytes), sensory neurons, and other cell types can influence nociceptive processing and lead to states of exaggerated and unresolved pain. In this review, we summarize our current understanding of how different TLRs and their accessory or adaptor molecules can contribute to the development and maintenance of persistent pain. The challenges and opportunities of targeting TLRs for new treatment strategies against chronic pain are discussed, including the therapeutic context of TLR-mediated signaling in opioid analgesia and chemotherapy-induced pain. Considering the prevalence of persistent pain and the insufficient efficacy and safety of current treatment options, a deeper understanding of Toll-like receptors holds the promise of novel therapies for managing pathological pain.
Collapse
|
45
|
Xie RG, Gao YJ, Park CK, Lu N, Luo C, Wang WT, Wu SX, Ji RR. Spinal CCL2 Promotes Central Sensitization, Long-Term Potentiation, and Inflammatory Pain via CCR2: Further Insights into Molecular, Synaptic, and Cellular Mechanisms. Neurosci Bull 2017; 34:13-21. [PMID: 28265898 DOI: 10.1007/s12264-017-0106-5] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 01/22/2017] [Indexed: 12/17/2022] Open
Abstract
Mounting evidence supports an important role of chemokines, produced by spinal cord astrocytes, in promoting central sensitization and chronic pain. In particular, CCL2 (C-C motif chemokine ligand 2) has been shown to enhance N-methyl-D-aspartate (NMDA)-induced currents in spinal outer lamina II (IIo) neurons. However, the exact molecular, synaptic, and cellular mechanisms by which CCL2 modulates central sensitization are still unclear. We found that spinal injection of the CCR2 antagonist RS504393 attenuated CCL2- and inflammation-induced hyperalgesia. Single-cell RT-PCR revealed CCR2 expression in excitatory vesicular glutamate transporter subtype 2-positive (VGLUT2+) neurons. CCL2 increased NMDA-induced currents in CCR2+/VGLUT2+ neurons in lamina IIo; it also enhanced the synaptic NMDA currents evoked by dorsal root stimulation; and furthermore, it increased the total and synaptic NMDA currents in somatostatin-expressing excitatory neurons. Finally, intrathecal RS504393 reversed the long-term potentiation evoked in the spinal cord by C-fiber stimulation. Our findings suggest that CCL2 directly modulates synaptic plasticity in CCR2-expressing excitatory neurons in spinal lamina IIo, and this underlies the generation of central sensitization in pathological pain.
Collapse
Affiliation(s)
- Rou-Gang Xie
- Department of Neurobiology and Collaborative Innovation Center for Brain Science, The Fourth Military Medical University, Xi'an, 710032, China. .,Department of Anesthesiology, Duke University Medical Center, Durham, NC, 27710, USA.
| | - Yong-Jing Gao
- Pain Research Laboratory, Institute of Nautical Medicine, Jiangsu Key laboratory of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Chul-Kyu Park
- Department of Physiology, College of Medicine, Gachon University, Incheon, 21999, Republic of Korea
| | - Ning Lu
- Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Ceng Luo
- Department of Neurobiology and Collaborative Innovation Center for Brain Science, The Fourth Military Medical University, Xi'an, 710032, China
| | - Wen-Ting Wang
- Department of Neurobiology and Collaborative Innovation Center for Brain Science, The Fourth Military Medical University, Xi'an, 710032, China
| | - Sheng-Xi Wu
- Department of Neurobiology and Collaborative Innovation Center for Brain Science, The Fourth Military Medical University, Xi'an, 710032, China
| | - Ru-Rong Ji
- Department of Anesthesiology, Duke University Medical Center, Durham, NC, 27710, USA. .,Department of Neurobiology, Duke University Medical Center, Durham, NC, 27710, USA.
| |
Collapse
|