51
|
Wang P, Zhang Q, Dias FC, Suttle A, Dong X, Chen Y. TMEM100, a regulator of TRPV1-TRPA1 interaction, contributes to temporomandibular disorder pain. Front Mol Neurosci 2023; 16:1160206. [PMID: 37033371 PMCID: PMC10077888 DOI: 10.3389/fnmol.2023.1160206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 02/27/2023] [Indexed: 04/11/2023] Open
Abstract
There is an unmet need to identify new therapeutic targets for temporomandibular disorder (TMD) pain because current treatments are limited and unsatisfactory. TMEM100, a two-transmembrane protein, was recently identified as a regulator to weaken the TRPA1-TRPV1 physical association, resulting in disinhibition of TRPA1 activity in sensory neurons. Recent studies have also shown that Tmem100, Trpa1, and Trpv1 mRNAs were upregulated in trigeminal ganglion (TG) after inflammation of the temporomandibular joint (TMJ) associated tissues. These findings raise a critical question regarding whether TMEM100 in TG neurons is involved in TMD pain via regulating the TRPA1-TRPV1 functional interaction. Here, using two mouse models of TMD pain induced by TMJ inflammation or masseter muscle injury, we found that global knockout or systemic inhibition of TRPA1 and TRPV1 attenuated pain. In line with their increased genes, mice exhibited significant upregulation of TMEM100, TRPA1, and TRPV1 at the protein levels in TG neurons after TMD pain. Importantly, TMEM100 co-expressed with TRPA1 and TRPV1 in TG neurons-innervating the TMJ and masseter muscle and their co-expression was increased after TMD pain. Moreover, the enhanced activity of TRPA1 in TG neurons evoked by TMJ inflammation or masseter muscle injury was suppressed by inhibition of TMEM100. Selective deletion of Tmem100 in TG neurons or local administration of TMEM100 inhibitor into the TMJ or masseter muscle attenuated TMD pain. Together, these results suggest that TMEM100 in TG neurons contributes to TMD pain by regulating TRPA1 activity within the TRPA1-TRPV1 complex. TMEM100 therefore represents a potential novel target-of-interest for TMD pain.
Collapse
Affiliation(s)
- Peng Wang
- Department of Neurology, Duke University, Durham, NC, United States
| | - Qiaojuan Zhang
- Department of Neurology, Duke University, Durham, NC, United States
| | - Fabiana C. Dias
- Department of Neurology, Duke University, Durham, NC, United States
| | - Abbie Suttle
- Department of Neurology, Duke University, Durham, NC, United States
| | - Xinzhong Dong
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Yong Chen
- Department of Neurology, Duke University, Durham, NC, United States
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University, Durham, NC, United States
- Department of Pathology, Duke University, Durham, NC, United States
- *Correspondence: Yong Chen,
| |
Collapse
|
52
|
Peach CJ, Edgington-Mitchell LE, Bunnett NW, Schmidt BL. Protease-activated receptors in health and disease. Physiol Rev 2023; 103:717-785. [PMID: 35901239 PMCID: PMC9662810 DOI: 10.1152/physrev.00044.2021] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 07/08/2022] [Accepted: 07/10/2022] [Indexed: 11/22/2022] Open
Abstract
Proteases are signaling molecules that specifically control cellular functions by cleaving protease-activated receptors (PARs). The four known PARs are members of the large family of G protein-coupled receptors. These transmembrane receptors control most physiological and pathological processes and are the target of a large proportion of therapeutic drugs. Signaling proteases include enzymes from the circulation; from immune, inflammatory epithelial, and cancer cells; as well as from commensal and pathogenic bacteria. Advances in our understanding of the structure and function of PARs provide insights into how diverse proteases activate these receptors to regulate physiological and pathological processes in most tissues and organ systems. The realization that proteases and PARs are key mediators of disease, coupled with advances in understanding the atomic level structure of PARs and their mechanisms of signaling in subcellular microdomains, has spurred the development of antagonists, some of which have advanced to the clinic. Herein we review the discovery, structure, and function of this receptor system, highlight the contribution of PARs to homeostatic control, and discuss the potential of PAR antagonists for the treatment of major diseases.
Collapse
Affiliation(s)
- Chloe J Peach
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York
- Department of Neuroscience and Physiology and Neuroscience Institute, Grossman School of Medicine, New York University, New York, New York
| | - Laura E Edgington-Mitchell
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
- Bluestone Center for Clinical Research, Department of Oral and Maxillofacial Surgery, New York University College of Dentistry, New York, New York
| | - Nigel W Bunnett
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York
- Department of Neuroscience and Physiology and Neuroscience Institute, Grossman School of Medicine, New York University, New York, New York
| | - Brian L Schmidt
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York
- Bluestone Center for Clinical Research, Department of Oral and Maxillofacial Surgery, New York University College of Dentistry, New York, New York
| |
Collapse
|
53
|
Wang Q, #, Zhang Y, #, Du Q, Zhao X, Wang W, Zhai Q, Xiang M. SKF96365 impedes spinal glutamatergic transmission-mediated neuropathic allodynia. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2023; 27:39-48. [PMID: 36575932 PMCID: PMC9806642 DOI: 10.4196/kjpp.2023.27.1.39] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 08/18/2022] [Accepted: 09/08/2022] [Indexed: 12/29/2022]
Abstract
Spinal nerve injury causes mechanical allodynia and structural imbalance of neurotransmission, which were typically associated with calcium overload. Store-operated calcium entry (SOCE) is considered crucial elements-mediating intracellular calcium homeostasis, ion channel activity, and synaptic plasticity. However, the underlying mechanism of SOCE in mediating neuronal transmitter release and synaptic transmission remains ambiguous in neuropathic pain. Neuropathic rats were operated by spinal nerve ligations. Neurotransmissions were assessed by whole-cell recording in substantia gelatinosa. Immunofluorescence staining of STIM1 with neuronal and glial biomarkers in the spinal dorsal horn. The endoplasmic reticulum stress level was estimated from qRT-PCR. Intrathecal injection of SOCE antagonist SKF96365 dose-dependently alleviated mechanical allodynia in ipsilateral hind paws of neuropathic rats with ED50 of 18 μg. Immunofluorescence staining demonstrated that STIM1 was specifically and significantly expressed in neurons but not astrocytes and microglia in the spinal dorsal horn. Bath application of SKF96365 inhibited enhanced miniature excitatory postsynaptic currents in a dosage-dependent manner without affecting miniature inhibitory postsynaptic currents. Mal-adaption of SOCE was commonly related to endoplasmic reticulum (ER) stress in the central nervous system. SKF96365 markedly suppressed ER stress levels by alleviating mRNA expression of C/EBP homologous protein and heat shock protein 70 in neuropathic rats. Our findings suggested that nerve injury might promote SOCE-mediated calcium levels, resulting in long-term imbalance of spinal synaptic transmission and behavioral sensitization, SKF96365 produces antinociception by alleviating glutamatergic transmission and ER stress. This work demonstrated the involvement of SOCE in neuropathic pain, implying that SOCE might be a potential target for pain management.
Collapse
Affiliation(s)
- Qiru Wang
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Minhang Branch, Shanghai 200240, China
| | - #
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Minhang Branch, Shanghai 200240, China
| | - Yang Zhang
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai 200240, China
| | - #
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Minhang Branch, Shanghai 200240, China
| | - Qiong Du
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Minhang Branch, Shanghai 200240, China
| | - Xinjie Zhao
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Minhang Branch, Shanghai 200240, China
| | - Wei Wang
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai 200240, China,Correspondence Ming Xiang, E-mail: , Qing Zhai, E-mail: , Wei Wang, E-mail:
| | - Qing Zhai
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Minhang Branch, Shanghai 200240, China,Correspondence Ming Xiang, E-mail: , Qing Zhai, E-mail: , Wei Wang, E-mail:
| | - Ming Xiang
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Minhang Branch, Shanghai 200240, China,Correspondence Ming Xiang, E-mail: , Qing Zhai, E-mail: , Wei Wang, E-mail:
| |
Collapse
|
54
|
Upregulation of DRG protein TMEM100 facilitates dry-skin-induced pruritus by enhancing TRPA1 channel function. Acta Biochim Biophys Sin (Shanghai) 2022; 55:404-416. [PMID: 36514220 PMCID: PMC10160222 DOI: 10.3724/abbs.2022180] [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: 11/30/2022] Open
Abstract
The dry skin tortures numerous patients with severe itch. The transient receptor potential cation channel V member 1 (TRPV1) and A member 1 (TRPA1) are two essential receptors for peripheral neural coding of itch sensory, mediating histaminergic and nonhistaminergic itch separately. In the dorsal root ganglion, transmembrane protein 100 (TMEM100) is structurally related to both TRPV1 and TRPA1 receptors, but the exact role of TMEM100 in itch sensory coding is still unknown. Here, in this study, we find that TMEM100 + DRG neurons account for the majority of activated neurons in an acetone-ether-water (AEW)-induced dry skin itch model, and some TMEM100 + DRG neurons are colocalized with both TRPA1 and the chloroquine-related Mrgpr itch receptor family. Both the expression and function of TRPA1 channels, but not TRPV1 channels, are upregulated in the AEW model, and specific DRG Tmem100 gene knockdown alleviates AEW-induced itch and rescues the expression and functional changes of TRPA1. Our results strongly suggest that TMEM100 protein in DRG is the main facilitating factor for dry-skin-related chronic itch, and specific suppression of TMEM100 in DRG could be a novel effective treatment strategy for patients who suffer from dry skin-induced itch.
Collapse
|
55
|
Xu LL, Yan Y, Yuan YM, Li Y, Jiang J, Zhang LC. TRPV1 and GABAB1 in the Cerebrospinal Fluid-Contacting Nucleus are Jointly Involved in Chronic Inflammatory Pain in Rats. J Pain Res 2022; 15:3931-3939. [DOI: 10.2147/jpr.s385810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 12/06/2022] [Indexed: 12/15/2022] Open
|
56
|
Zhang YY, Liu F, Fang ZH, Li YL, Liao HL, Song QX, Zhou C, Shen JF. Differential roles of NMDAR subunits 2A and 2B in mediating peripheral and central sensitization contributing to orofacial neuropathic pain. Brain Behav Immun 2022; 106:129-146. [PMID: 36038077 DOI: 10.1016/j.bbi.2022.08.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 07/27/2022] [Accepted: 08/23/2022] [Indexed: 11/29/2022] Open
Abstract
The spinal N-methyl-d-aspartate receptor (NMDAR), particularly their subtypes NR2A and NR2B, plays pivotal roles in neuropathic and inflammatory pain. However, the roles of NR2A and NR2B in orofacial pain and the exact molecular and cellular mechanisms mediating nervous system sensitization are still poorly understood. Here, we exhaustively assessed the regulatory effect of NMDAR in mediating peripheral and central sensitization in orofacial neuropathic pain. Von-Frey filament tests showed that the inferior alveolar nerve transection (IANX) induced ectopic allodynia behavior in the whisker pad of mice. Interestingly, mechanical allodynia was reversed in mice lacking NR2A and NR2B. IANX also promoted the production of peripheral sensitization-related molecules, such as interleukin (IL)-1β, tumor necrosis factor (TNF)-α, brain-derived neurotrophic factor (BDNF), and chemokine upregulation (CC motif) ligand 2 (CCL2), and decreased the inward potassium channel (Kir) 4.1 on glial cells in the trigeminal ganglion, but NR2A conditional knockout (CKO) mice prevented these alterations. In contrast, NR2B CKO only blocked the changes of Kir4.1, IL-1β, and TNF-α and further promoted the production of CCL2. Central sensitization-related c-fos, glial fibrillary acidic protein (GFAP), and ionized calcium-binding adaptor molecule 1 (Iba-1) were promoted and Kir4.1 was reduced in the spinal trigeminal caudate nucleus by IANX. Differential actions of NR2A and NR2B in mediating central sensitization were also observed. Silencing of NR2B was effective in reducing c-fos, GFAP, and Iba-1 but did not affect Kir4.1. In contrast, NR2A CKO only altered Iba-1 and Kir4.1 and further increased c-fos and GFAP. Gain-of-function and loss-of-function approaches provided insight into the differential roles of NR2A and NR2B in mediating peripheral and central nociceptive sensitization induced by IANX, which may be a fundamental basis for advancing knowledge of the neural mechanisms' reaction to nerve injury.
Collapse
Affiliation(s)
- Yan-Yan Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Fei Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zhong-Han Fang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yue-Ling Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Hong-Lin Liao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qin-Xuan Song
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Cheng Zhou
- Laboratory of Anesthesia and Critical Care Medicine, Translational Neuroscience Center, West China Hospital of Sichuan University, Chengdu, China
| | - Jie-Fei Shen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
57
|
Jia YZ, Li HT, Zhang GM, Wu HY, Zhang SS, Zhi HW, Wang YH, Zhu JW, Wang YF, Xu XQ, Tian CJ, Cui WQ. Electroacupuncture alleviates orofacial allodynia and anxiety-like behaviors by regulating synaptic plasticity of the CA1 hippocampal region in a mouse model of trigeminal neuralgia. Front Mol Neurosci 2022; 15:979483. [PMID: 36277498 PMCID: PMC9582442 DOI: 10.3389/fnmol.2022.979483] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 09/08/2022] [Indexed: 11/13/2022] Open
Abstract
Objective Trigeminal neuralgia (TN), one of the most severe and debilitating chronic pain conditions, is often accompanied by mood disorders, such as anxiety and depression. Electroacupuncture (EA) is a characteristic therapy of Traditional Chinese Medicine with analgesic and anxiolytic effects. This study aimed to investigate whether EA ameliorates abnormal TN orofacial pain and anxiety-like behavior by altering synaptic plasticity in the hippocampus CA1. Materials and methods A mouse infraorbital nerve transection model (pT-ION) of neuropathic pain was established, and EA or sham EA was used to treat ipsilateral acupuncture points (GV20-Baihui and ST7-Xiaguan). Golgi-Cox staining and transmission electron microscopy (TEM) were administrated to observe the changes of synaptic plasticity in the hippocampus CA1. Results Stable and persistent orofacial allodynia and anxiety-like behaviors induced by pT-ION were related to changes in hippocampal synaptic plasticity. Golgi stainings showed a decrease in the density of dendritic spines, especially mushroom-type dendritic spines, in hippocampal CA1 neurons of pT-ION mice. TEM results showed that the density of synapses, membrane thickness of the postsynaptic density, and length of the synaptic active zone were decreased, whereas the width of the synaptic cleft was increased in pT-ION mice. EA attenuated pT-ION-induced orofacial allodynia and anxiety-like behaviors and effectively reversed the abnormal changes in dendritic spines and synapse of the hippocampal CA1 region. Conclusion EA modulates synaptic plasticity of hippocampal CA1 neurons, thereby reducing abnormal orofacial pain and anxiety-like behavior. This provides evidence for a TN treatment strategy.
Collapse
Affiliation(s)
- Yu-Zhi Jia
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Hai-Tao Li
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Guang-Ming Zhang
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Hong-Yun Wu
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Si-Shuo Zhang
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Hong-Wei Zhi
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ya-Han Wang
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jing-Wen Zhu
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yi-Fan Wang
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiang-Qing Xu
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Cai-Jun Tian
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wen-Qiang Cui
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
58
|
Liu YJ, Li YL, Fang ZH, Liao HL, Zhang YY, Lin J, Liu F, Shen JF. NMDARs mediate peripheral and central sensitization contributing to chronic orofacial pain. Front Cell Neurosci 2022; 16:999509. [PMID: 36238833 PMCID: PMC9553029 DOI: 10.3389/fncel.2022.999509] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 08/22/2022] [Indexed: 11/28/2022] Open
Abstract
Peripheral and central sensitizations of the trigeminal nervous system are the main mechanisms to promote the development and maintenance of chronic orofacial pain characterized by allodynia, hyperalgesia, and ectopic pain after trigeminal nerve injury or inflammation. Although the pathomechanisms of chronic orofacial pain are complex and not well known, sufficient clinical and preclinical evidence supports the contribution of the N-methyl-D-aspartate receptors (NMDARs, a subclass of ionotropic glutamate receptors) to the trigeminal nociceptive signal processing pathway under various pathological conditions. NMDARs not only have been implicated as a potential mediator of pain-related neuroplasticity in the peripheral nervous system (PNS) but also mediate excitatory synaptic transmission and synaptic plasticity in the central nervous system (CNS). In this review, we focus on the pivotal roles and mechanisms of NMDARs in the trigeminal nervous system under orofacial neuropathic and inflammatory pain. In particular, we summarize the types, components, and distribution of NMDARs in the trigeminal nervous system. Besides, we discuss the regulatory roles of neuron-nonneuronal cell/neuron-neuron communication mediated by NMDARs in the peripheral mechanisms of chronic orofacial pain following neuropathic injury and inflammation. Furthermore, we review the functional roles and mechanisms of NMDARs in the ascending and descending circuits under orofacial neuropathic and inflammatory pain conditions, which contribute to the central sensitization. These findings are not only relevant to understanding the underlying mechanisms, but also shed new light on the targeted therapy of chronic orofacial pain.
Collapse
Affiliation(s)
- Ya-Jing Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, Chengdu, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yue-Ling Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, Chengdu, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zhong-Han Fang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, Chengdu, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Hong-Lin Liao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, Chengdu, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yan-Yan Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, Chengdu, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jiu Lin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, Chengdu, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Fei Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, Chengdu, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- *Correspondence: Jie-Fei Shen Fei Liu
| | - Jie-Fei Shen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, West China School of Stomatology, Sichuan University, Chengdu, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- *Correspondence: Jie-Fei Shen Fei Liu
| |
Collapse
|
59
|
Wu Z, Yang M, Zhao P, Zou F, Peng J, Deng Q, Duan G, Li H. The Association Between Long-Term Spicy-Food Consumption and the Incidence of Chronic Postsurgical Pain After Cesarean Delivery: An Observational Study. J Pain Res 2022; 15:2833-2844. [PMID: 36124036 PMCID: PMC9482407 DOI: 10.2147/jpr.s373030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 09/06/2022] [Indexed: 11/23/2022] Open
Abstract
Background Our previous study found that a long-term diet incorporating spicy foods can reduce the human basal pain threshold. Capsaicin is the pungent ingredient in chili peppers. Transient receptor potential vanilloid type1 is the capsaicin receptor expressed in the oral cavity and is the primary sensory neuron of the “pain” pathway. Few studies have examined the association between long-term spicy diet and chronic postsurgical pain (CPSP). Women who underwent elective cesarean section (eCS) have consistent characteristics of CPSP. This study aimed to investigate the relationship between a long-term spicy diet and the incidence of CPSP after eCS. Methods Participants were divided into a low frequency group (LF, numerical rating scale (NRS)<5) for spicy food consumption and a high frequency group (HF, NRS≥5) by receiver operator characteristic analysis. The primary outcome was the incidence of CPSP three months after eCS. Propensity score matching (PSM) analysis was performed between the two frequency groups. Stepwise logistic regression analysis was then performed. Results Of the 1029 enrolled patients, data from 982 were analyzed 3 months after eCS. After PSM, the incidence of CPSP in the HF group (30.1% [108/359]) was higher than that in the LF group (19.8% [71/359]; P = 0.001). Compared with the LF group, the risk of CPSP in the HF group increased 1.61 times by 3 months (95% CI 1.18–2.20, P = 0.003). PSM results found that 1 year, the incidence of CPSP in the HF group (15.2% [56/369]) was higher than that in the LF group (8.1% [30/369], P = 0.003). Conclusion With an NRS≥5 as a boundary, women who consumed spicy food ≥ 2 days/week were more likely to have CPSP than those who consumed spicy food < 2 days/week.
Collapse
Affiliation(s)
- Zhuoxi Wu
- Department of Anesthesiology, Second Affiliated Hospital of Army Medical University, People's Liberation Army of China, Chongqing, People's Republic of China
| | - Mi Yang
- Department of Anesthesiology, Second Affiliated Hospital of Army Medical University, People's Liberation Army of China, Chongqing, People's Republic of China
| | - Peng Zhao
- Department of Anesthesiology, Second Affiliated Hospital of Army Medical University, People's Liberation Army of China, Chongqing, People's Republic of China.,Department of Anesthesiology, Chinese People's Liberation Army of China (PLA) No. 964 Hospital, Changchun, People's Republic of China
| | - Feng Zou
- Department of Anesthesiology, Second Affiliated Hospital of Army Medical University, People's Liberation Army of China, Chongqing, People's Republic of China
| | - Jing Peng
- Department of Anesthesiology, Second Affiliated Hospital of Army Medical University, People's Liberation Army of China, Chongqing, People's Republic of China
| | - Qiangting Deng
- Editorial Office of Journal of Third Military Medical University, Army Medical University, People's Liberation Army of China, Chongqing, People's Republic of China
| | - Guangyou Duan
- Department of Anesthesiology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, People's Republic of China
| | - Hong Li
- Department of Anesthesiology, Second Affiliated Hospital of Army Medical University, People's Liberation Army of China, Chongqing, People's Republic of China
| |
Collapse
|
60
|
Zhuang ZF, Wu HY, Song YY, Li L, Cui X, Yang J, Xu XQ, Cui WQ. N-Methyl D-aspartate receptor subtype 2B/Ca2+/calmodulin-dependent protein kinase II signaling in the lateral habenula regulates orofacial allodynia and anxiety-like behaviors in a mouse model of trigeminal neuralgia. Front Cell Neurosci 2022; 16:981190. [PMID: 36187288 PMCID: PMC9521491 DOI: 10.3389/fncel.2022.981190] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 08/22/2022] [Indexed: 11/18/2022] Open
Abstract
Trigeminal neuralgia (TN) is a peripheral nerve disorder often accompanied by abnormalities in mood. The lateral habenula (LHb) plays important roles in the modulation of pain and emotion. In the present study, we investigated the involvement of the LHb in the mechanisms underlying allodynia and anxiety induced by partial transection of the infraorbital nerve (pT-ION) in mice. Our results indicated that pT-ION induced persistent orofacial allodynia and anxiety-like behaviors, which were correlated with increased phosphorylation of N-Methyl D-aspartate receptor (NMDAR) subtype 2B (p-NR2B) and Ca2+/calmodulin-dependent protein kinase II (p-CaMKII) in LHb neurons. Bilateral inhibition of NMDARs and CaMKII in the LHb attenuated the allodynia and anxiety-like behavior induced by pT-ION. Furthermore, bilateral activation of NMDARs in the LHb increased the expression of p-NR2B and p-CaMKII and induced orofacial allodynia and anxiety-like behaviors in naive mice. Adeno-associated virus (AAV)-mediated expression of hM3D(Gq) in CaMKII+ neurons of the bilateral LHb, followed by clozapine-N-oxide (CNO) administration, also triggered orofacial allodynia and anxiety-like behaviors in naïve mice with successful virus infection in LHb neurons (verified based on immunofluorescence). In conclusion, these findings suggest that activation of NMDA/CaMKII signaling in the LHb contributes to the occurrence and development of TN and related anxiety-like behaviors. Therefore, suppressing the activity of CaMKII+ neurons in the bilateral LHb by targeting NMDA/CaMKII may represent a novel strategy for treating pain and anxiety associated with TN.
Collapse
Affiliation(s)
- Zi-Fan Zhuang
- College of First Clinical Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Hong-Yun Wu
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ya-Yi Song
- Department of Traditional Chinese Medicine, Lianyungang Maternal and Child Health Hospital, Lianyungang, China
| | - Lei Li
- Department of Cardiology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xia Cui
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jie Yang
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiang-Qing Xu
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
- Xiang-Qing Xu,
| | - Wen-Qiang Cui
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
- *Correspondence: Wen-Qiang Cui,
| |
Collapse
|
61
|
High-fat diet causes mechanical allodynia in the absence of injury or diabetic pathology. Sci Rep 2022; 12:14840. [PMID: 36050326 PMCID: PMC9437006 DOI: 10.1038/s41598-022-18281-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 08/09/2022] [Indexed: 12/04/2022] Open
Abstract
Understanding the interactions between diet, obesity, and diabetes is important to tease out mechanisms in painful pathology. Western diet is rich in fats, producing high amounts of circulating bioactive metabolites. However, no research has assessed how a high-fat diet (HFD) alone may sensitize an individual to non-painful stimuli in the absence of obesity or diabetic pathology. To investigate this, we tested the ability of a HFD to stimulate diet-induced hyperalgesic priming, or diet sensitization in male and female mice. Our results revealed that 8 weeks of HFD did not alter baseline pain sensitivity, but both male and female HFD-fed animals exhibited robust mechanical allodynia when exposed to a subthreshold dose of intraplantar Prostaglandin E2 (PGE2) compared to mice on chow diet. Furthermore, calcium imaging in isolated primary sensory neurons of both sexes revealed HFD induced an increased percentage of capsaicin-responsive neurons compared to their chow counterparts. Immunohistochemistry (IHC) showed a HFD-induced upregulation of ATF3, a neuronal marker of injury, in lumbar dorsal root ganglia (DRG). This suggests that a HFD induces allodynia in the absence of a pre-existing condition or injury via dietary components. With this new understanding of how a HFD can contribute to the onset of pain, we can understand the dissociation behind the comorbidities associated with obesity and diabetes to develop pharmacological interventions to treat them more efficiently.
Collapse
|
62
|
Shannonhouse J, Bernabucci M, Gomez R, Son H, Zhang Y, Ai CH, Ishida H, Kim YS. Meclizine and Metabotropic Glutamate Receptor Agonists Attenuate Severe Pain and Ca 2+ Activity of Primary Sensory Neurons in Chemotherapy-Induced Peripheral Neuropathy. J Neurosci 2022; 42:6020-6037. [PMID: 35772967 PMCID: PMC9351649 DOI: 10.1523/jneurosci.1064-21.2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 06/02/2022] [Accepted: 06/22/2022] [Indexed: 02/05/2023] Open
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) affects ∼68% of patients undergoing chemotherapy, causing debilitating neuropathic pain and reducing quality of life. Cisplatin is a commonly used platinum-based chemotherapeutic drug known to cause CIPN, possibly by causing oxidative stress damage to primary sensory neurons. Metabotropic glutamate receptors (mGluRs) are widely hypothesized to be involved in pain processing and pain mitigation. Meclizine is an H1 histamine receptor antagonist known to have neuroprotective effects, including an anti-oxidative effect. Here, we used a mouse model of cisplatin-induced CIPN using male and female mice to test agonists of mGluR8 and Group II mGluR as well as meclizine as interventions to reduce cisplatin-induced pain. We performed behavioral pain tests, and we imaged Ca2+ activity of the large population of dorsal root ganglia (DRG) neurons in vivo For the latter, we used a genetically-encoded Ca2+ indicator, Pirt-GCaMP3, which enabled us to monitor different drug interventions at the level of the intact DRG neuronal ensemble. We found that CIPN increased spontaneous Ca2+ activity in DRG neurons, increased number of Ca2+ transients, and increased hyper-responses to mechanical, thermal, and chemical stimuli. We found that mechanical and thermal pain caused by CIPN was significantly attenuated by the mGluR8 agonist, (S)-3,4-DCPG, the Group II mGluR agonist, LY379268, and the H1 histamine receptor antagonist, meclizine. DRG neuronal Ca2+ activity elevated by CIPN was attenuated by LY379268 and meclizine, but not by (S)-3,4-DCPG. Furthermore, meclizine and LY379268 attenuated cisplatin-induced weight loss. These results suggest that Group II mGluR agonist, mGluR8 agonist, and meclizine are promising candidates as new treatment options for CIPN, and studies of their mechanisms are warranted.SIGNIFICANCE STATEMENT Chemotherapy-induced peripheral neuropathy (CIPN) is a painful condition that affects most chemotherapy patients and persists several months or longer after treatment ends. Research on CIPN mechanism is extensive but has produced only few clinically useful treatments. Using in vivo GCaMP Ca2+ imaging in live animals over 1800 neurons/dorsal root ganglia (DRG) at once, we have characterized the effects of the chemotherapeutic drug, cisplatin and three treatments that decrease CIPN pain. Cisplatin increases sensory neuronal Ca2+ activity and develops various sensitization. Metabotropic glutamate receptor (mGluR) agonist, LY379268 or the H1 histamine receptor antagonist, meclizine decreases cisplatin's effects on neuronal Ca2+ activity and reduces pain hypersensitivity. Our results and experiments provide insights into cellular effects of cisplatin and drugs preventing CIPN pain.
Collapse
Affiliation(s)
| | - Matteo Bernabucci
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ 08854
| | - Ruben Gomez
- Department of Oral & Maxillofacial Surgery, School of Dentistry
| | - Hyeonwi Son
- Department of Oral & Maxillofacial Surgery, School of Dentistry
| | - Yan Zhang
- Department of Oral & Maxillofacial Surgery, School of Dentistry
| | - Chih-Hsuan Ai
- Department of Oral & Maxillofacial Surgery, School of Dentistry
| | - Hirotake Ishida
- Department of Oral & Maxillofacial Surgery, School of Dentistry
| | - Yu Shin Kim
- Department of Oral & Maxillofacial Surgery, School of Dentistry
- Programs in Integrated Biomedical Sciences, Translational Sciences, Biomedical Engineering, Radiological Sciences, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229
| |
Collapse
|
63
|
Chen Z, Huang Q, Song X, Ford NC, Zhang C, Xu Q, Lay M, He SQ, Dong X, Hanani M, Guan Y. Purinergic signaling between neurons and satellite glial cells of mouse dorsal root ganglia modulates neuronal excitability in vivo. Pain 2022; 163:1636-1647. [PMID: 35027518 PMCID: PMC9771604 DOI: 10.1097/j.pain.0000000000002556] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 11/24/2021] [Indexed: 10/19/2022]
Abstract
ABSTRACT Primary sensory neurons in dorsal root ganglia (DRG) are wrapped by satellite glial cells (SGCs), and neuron-SGC interaction may affect somatosensation, especially nociceptive transmission. P2-purinergic receptors (P2Rs) are key elements in the two-way interactions between DRG neurons and SGCs. However, because the cell types are in such close proximity, conventional approaches such as in vitro culture and electrophysiologic recordings are not adequate to investigate the physiologically relevant responses of these cells at a population level. Here, we performed in vivo calcium imaging to survey the activation of hundreds of DRG neurons in Pirt-GCaMP6s mice and to assess SGC activation in GFAP-GCaMP6s mice in situ. By combining pharmacologic and electrophysiologic techniques, we investigated how ganglionic purinergic signaling initiated by α,β-methyleneadenosine 5'-triphosphate (α,β-MeATP) modulates neuronal activity and excitability at a population level. We found that α,β-MeATP induced robust activation of small neurons-likely nociceptors-through activation of P2X3R. Large neurons, which are likely non-nociceptive, were also activated by α,β-MeATP, but with a delay. Blocking pannexin 1 channels attenuated the late phase response of DRG neurons, indicating that P2R stimulation may subsequently induce paracrine ATP release, which could further activate cells in the ganglion. Moreover, ganglionic α,β-MeATP treatment in vivo sensitized small neurons and enhanced responses of spinal wide-dynamic-range neurons to subsequent C-fiber inputs, suggesting that modulation via ganglionic P2R signaling could significantly affect nociceptive neuron excitability and pain transmission. Therefore, targeting functional P2Rs within ganglia may represent an important new strategy for pain modulation.
Collapse
Affiliation(s)
- Zhiyong Chen
- Department of Anesthesiology and Critical Care Medicine, the Johns Hopkins University, School of Medicine, Baltimore, MD, United States
| | - Qian Huang
- Department of Anesthesiology and Critical Care Medicine, the Johns Hopkins University, School of Medicine, Baltimore, MD, United States
| | - Xiaodan Song
- Department of Anesthesiology and Critical Care Medicine, the Johns Hopkins University, School of Medicine, Baltimore, MD, United States
| | - Neil C. Ford
- Department of Anesthesiology and Critical Care Medicine, the Johns Hopkins University, School of Medicine, Baltimore, MD, United States
| | - Chi Zhang
- Department of Anesthesiology and Critical Care Medicine, the Johns Hopkins University, School of Medicine, Baltimore, MD, United States
| | - Qian Xu
- The Solomon H. Snyder Department of Neuroscience, Center for Sensory Biology, Johns Hopkins University, School of Medicine, Baltimore, MD, United States
- Howard Hughes Medical Institute, Johns Hopkins University, School of Medicine, Baltimore, MD, United States
| | - Mark Lay
- The Solomon H. Snyder Department of Neuroscience, Center for Sensory Biology, Johns Hopkins University, School of Medicine, Baltimore, MD, United States
| | - Shao-Qiu He
- Department of Anesthesiology and Critical Care Medicine, the Johns Hopkins University, School of Medicine, Baltimore, MD, United States
| | - Xinzhong Dong
- The Solomon H. Snyder Department of Neuroscience, Center for Sensory Biology, Johns Hopkins University, School of Medicine, Baltimore, MD, United States
- Howard Hughes Medical Institute, Johns Hopkins University, School of Medicine, Baltimore, MD, United States
| | - Menachem Hanani
- Laboratory of Experimental Surgery, Hadassah-Hebrew University Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Mount Scopus, Jerusalem, Israel
| | - Yun Guan
- Department of Anesthesiology and Critical Care Medicine, the Johns Hopkins University, School of Medicine, Baltimore, MD, United States
- Department of Neurological Surgery, Johns Hopkins University, School of Medicine, Baltimore, MD, United States
| |
Collapse
|
64
|
Nguyen E, Smith KM, Cramer N, Holland RA, Bleimeister IH, Flores-Felix K, Silberberg H, Keller A, Le Pichon CE, Ross SE. Medullary kappa-opioid receptor neurons inhibit pain and itch through a descending circuit. Brain 2022; 145:2586-2601. [PMID: 35598161 PMCID: PMC9612802 DOI: 10.1093/brain/awac189] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 04/01/2022] [Accepted: 04/27/2022] [Indexed: 11/13/2022] Open
Abstract
In perilous and stressful situations, the ability to suppress pain can be critical for survival. The rostral ventromedial medulla contains neurons that robustly inhibit nocioception at the level of the spinal cord through a top-down modulatory pathway. Although much is known about the role of the rostral ventromedial medulla in the inhibition of pain, the precise ability to directly manipulate pain-inhibitory neurons in the rostral ventromedial medulla has never been achieved. We now expose a cellular circuit that inhibits nocioception and itch in mice. Through a combination of molecular, tracing and behavioural approaches, we found that rostral ventromedial medulla neurons containing the kappa-opioid receptor inhibit itch and nocioception. With chemogenetic inhibition, we uncovered that these neurons are required for stress-induced analgesia. Using intersectional chemogenetic and pharmacological approaches, we determined that rostral ventromedial medulla kappa-opioid receptor neurons inhibit nocioception and itch through a descending circuit. Lastly, we identified a dynorphinergic pathway arising from the periaqueductal grey that modulates nociception within the rostral ventromedial medulla. These discoveries highlight a distinct population of rostral ventromedial medulla neurons capable of broadly and robustly inhibiting itch and nocioception.
Collapse
Affiliation(s)
- Eileen Nguyen
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Kelly M Smith
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Nathan Cramer
- Department of Anatomy and Neurobiology, Program in Neuroscience, University of Maryland, School of Medicine, Baltimore, MD 21201, USA
| | - Ruby A Holland
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Isabel H Bleimeister
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Krystal Flores-Felix
- Department of Anatomy and Neurobiology, Program in Neuroscience, University of Maryland, School of Medicine, Baltimore, MD 21201, USA
| | - Hanna Silberberg
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Asaf Keller
- Department of Anatomy and Neurobiology, Program in Neuroscience, University of Maryland, School of Medicine, Baltimore, MD 21201, USA
| | - Claire E Le Pichon
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sarah E Ross
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| |
Collapse
|
65
|
Cooper AH, Hedden NS, Prasoon P, Qi Y, Taylor BK. Postsurgical Latent Pain Sensitization Is Driven by Descending Serotonergic Facilitation and Masked by µ-Opioid Receptor Constitutive Activity in the Rostral Ventromedial Medulla. J Neurosci 2022; 42:5870-5881. [PMID: 35701159 PMCID: PMC9337598 DOI: 10.1523/jneurosci.2038-21.2022] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 05/22/2022] [Accepted: 05/27/2022] [Indexed: 01/29/2023] Open
Abstract
Following tissue injury, latent sensitization (LS) of nociceptive signaling can persist indefinitely, kept in remission by compensatory µ-opioid receptor constitutive activity (MORCA) in the dorsal horn of the spinal cord. To demonstrate LS, we conducted plantar incision in mice and then waited 3-4 weeks for hypersensitivity to resolve. At this time (remission), systemic administration of the opioid receptor antagonist/inverse agonist naltrexone reinstated mechanical and heat hypersensitivity. We first tested the hypothesis that LS extends to serotonergic neurons in the rostral ventral medulla (RVM) that convey pronociceptive input to the spinal cord. We report that in male and female mice, hypersensitivity was accompanied by increased Fos expression in serotonergic neurons of the RVM, abolished on chemogenetic inhibition of RVM 5-HT neurons, and blocked by intrathecal injection of the 5-HT3R antagonist ondansetron; the 5-HT2AR antagonist MDL-11 939 had no effect. Second, to test for MORCA, we microinjected the MOR inverse agonist d-Phe-Cys-Tyr-D-Trp-Arg-Thr-Pen-Thr-NH2 (CTAP) and/or neutral opioid receptor antagonist 6β-naltrexol. Intra-RVM CTAP produced mechanical hypersensitivity at both hindpaws; 6β-naltrexol had no effect by itself, but blocked CTAP-induced hypersensitivity. This indicates that MORCA, rather than an opioid ligand-dependent mechanism, maintains LS in remission. We conclude that incision establishes LS in descending RVM 5-HT neurons that drives pronociceptive 5-HT3R signaling in the dorsal horn, and this LS is tonically opposed by MORCA in the RVM. The 5-HT3 receptor is a promising therapeutic target for the development of drugs to prevent the transition from acute to chronic postsurgical pain.SIGNIFICANCE STATEMENT Surgery leads to latent pain sensitization and a compensatory state of endogenous pain control that is maintained long after tissue healing. Here, we show that either chemogenetic inhibition of serotonergic neuron activity in the RVM or pharmacological inhibition of 5-HT3 receptor signaling at the spinal cord blocks behavioral signs of postsurgical latent sensitization. We conclude that MORCA in the RVM opposes descending serotonergic facilitation of LS and that the 5-HT3 receptor is a promising therapeutic target for the development of drugs to prevent the transition from acute to chronic postsurgical pain.
Collapse
Affiliation(s)
- Andrew H Cooper
- Department of Anesthesiology and Perioperative Medicine, Pittsburgh Center for Pain Research, Pittsburgh Project to End Opioid Misuse, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213
| | - Naomi S Hedden
- Department of Anesthesiology and Perioperative Medicine, Pittsburgh Center for Pain Research, Pittsburgh Project to End Opioid Misuse, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213
| | - Pranav Prasoon
- Department of Anesthesiology and Perioperative Medicine, Pittsburgh Center for Pain Research, Pittsburgh Project to End Opioid Misuse, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213
| | - Yanmei Qi
- Department of Anesthesiology and Perioperative Medicine, Pittsburgh Center for Pain Research, Pittsburgh Project to End Opioid Misuse, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213
| | - Bradley K Taylor
- Department of Anesthesiology and Perioperative Medicine, Pittsburgh Center for Pain Research, Pittsburgh Project to End Opioid Misuse, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213
| |
Collapse
|
66
|
BzATP Activates Satellite Glial Cells and Increases the Excitability of Dorsal Root Ganglia Neurons In Vivo. Cells 2022; 11:cells11152280. [PMID: 35892578 PMCID: PMC9330736 DOI: 10.3390/cells11152280] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 07/18/2022] [Accepted: 07/21/2022] [Indexed: 12/14/2022] Open
Abstract
The purinergic system plays an important role in pain transmission. Recent studies have suggested that activation of P2-purinergic receptors (P2Rs) may be involved in neuron-satellite glial cell (SGC) interactions in the dorsal root ganglia (DRG), but the details remain unclear. In DRG, P2X7R is selectively expressed in SGCs, which closely surround neurons, and is highly sensitive to 3’-O-(4-Benzoyl) benzoyl-ATP (BzATP). Using calcium imaging in intact mice to survey a large number of DRG neurons and SGCs, we examined how intra-ganglionic purinergic signaling initiated by BzATP affects neuronal activities in vivo. We developed GFAP-GCaMP6s and Pirt-GCaMP6s mice to express the genetically encoded calcium indicator GGCaM6s in SGCs and DRG neurons, respectively. The application of BzATP to the ganglion induced concentration-dependent activation of SGCs in GFAP-GCaMP6s mice. In Pirt-GCaMP6s mice, BzATP initially activated more large-size neurons than small-size ones. Both glial and neuronal responses to BzATP were blocked by A438079, a P2X7R-selective antagonist. Moreover, blockers to pannexin1 channels (probenecid) and P2X3R (A317491) also reduced the actions of BzATP, suggesting that P2X7R stimulation may induce the opening of pannexin1 channels, leading to paracrine ATP release, which could further excite neurons by acting on P2X3Rs. Importantly, BzATP increased the responses of small-size DRG neurons and wide-dynamic range spinal neurons to subsequent peripheral stimuli. Our findings suggest that intra-ganglionic purinergic signaling initiated by P2X7R activation could trigger SGC-neuron interaction in vivo and increase DRG neuron excitability.
Collapse
|
67
|
Ma P, Mo R, Liao H, Qiu C, Wu G, Yang C, Zhang Y, Zhao Y, Song XJ. Gut microbiota depletion by antibiotics ameliorates somatic neuropathic pain induced by nerve injury, chemotherapy, and diabetes in mice. J Neuroinflammation 2022; 19:169. [PMID: 35764988 PMCID: PMC9237999 DOI: 10.1186/s12974-022-02523-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 06/12/2022] [Indexed: 12/02/2022] Open
Abstract
Background Gut microbiota has been found involved in neuronal functions and neurological disorders. Whether and how gut microbiota impacts chronic somatic pain disorders remain elusive.
Methods Neuropathic pain was produced by different forms of injury or diseases, the chronic constriction injury (CCI) of the sciatic nerves, oxaliplatin (OXA) chemotherapy, and streptozocin (STZ)-induced diabetes in mice. Continuous feeding of antibiotics (ABX) cocktail was used to cause major depletion of the gut microbiota. Fecal microbiota, biochemical changes in the spinal cord and dorsal root ganglion (DRG), and the behaviorally expressed painful syndromes were assessed. Results Under condition of gut microbiota depletion, CCI, OXA, or STZ treatment-induced thermal hyperalgesia or mechanical allodynia were prevented or completely suppressed. Gut microbiota depletion also prevented CCI or STZ treatment-induced glial cell activation in the spinal cord and inhibited cytokine production in DRG in OXA model. Interestingly, STZ treatment failed to induce the diabetic high blood glucose and painful hypersensitivity in animals with the gut microbiota depletion. ABX feeding starting simultaneously with CCI, OXA, or STZ treatment resulted in instant analgesia in all the animals. ABX feeding starting after establishment of the neuropathic pain in CCI- and STZ-, but not OXA-treated animals produced significant alleviation of the thermal hyeralgesia or mechanical allodynia. Transplantation of fecal bacteria from SPF mice to ABX-treated mice partially restored the gut microbiota and fully rescued the behaviorally expressed neuropathic pain, of which, Akkermansia, Bacteroides, and Desulfovibrionaceae phylus may play a key role. Conclusion This study demonstrates distinct roles of gut microbiota in the pathogenesis of chronic painful conditions with nerve injury, chemotherapy and diabetic neuropathy and supports the clinical significance of fecal bacteria transplantation.
Collapse
Affiliation(s)
- Pingchuan Ma
- Department of Medical Neurocience and SUSTech Center for Pain Medicine, School of Medicine, Southern University of Science and Technology, 1088 Xueyuan Ave, Nanshan District, Shenzhen, 518055, Guangdong, China.,Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education of China), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Rufan Mo
- Department of Medical Neurocience and SUSTech Center for Pain Medicine, School of Medicine, Southern University of Science and Technology, 1088 Xueyuan Ave, Nanshan District, Shenzhen, 518055, Guangdong, China
| | - Huabao Liao
- Department of Medical Neurocience and SUSTech Center for Pain Medicine, School of Medicine, Southern University of Science and Technology, 1088 Xueyuan Ave, Nanshan District, Shenzhen, 518055, Guangdong, China
| | - Chengjie Qiu
- Department of Medical Neurocience and SUSTech Center for Pain Medicine, School of Medicine, Southern University of Science and Technology, 1088 Xueyuan Ave, Nanshan District, Shenzhen, 518055, Guangdong, China
| | - Genhao Wu
- Department of Medical Neurocience and SUSTech Center for Pain Medicine, School of Medicine, Southern University of Science and Technology, 1088 Xueyuan Ave, Nanshan District, Shenzhen, 518055, Guangdong, China
| | - Caixia Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education of China), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Yunxiao Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education of China), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Yiran Zhao
- Department of Medical Neurocience and SUSTech Center for Pain Medicine, School of Medicine, Southern University of Science and Technology, 1088 Xueyuan Ave, Nanshan District, Shenzhen, 518055, Guangdong, China
| | - Xue-Jun Song
- Department of Medical Neurocience and SUSTech Center for Pain Medicine, School of Medicine, Southern University of Science and Technology, 1088 Xueyuan Ave, Nanshan District, Shenzhen, 518055, Guangdong, China. .,Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education of China), Peking University Cancer Hospital and Institute, Beijing, 100142, China.
| |
Collapse
|
68
|
Mahadevan AS, Long BL, Hu CW, Ryan DT, Grandel NE, Britton GL, Bustos M, Gonzalez Porras MA, Stojkova K, Ligeralde A, Son H, Shannonhouse J, Robinson JT, Warmflash A, Brey EM, Kim YS, Qutub AA. cytoNet: Spatiotemporal network analysis of cell communities. PLoS Comput Biol 2022; 18:e1009846. [PMID: 35696439 PMCID: PMC9191702 DOI: 10.1371/journal.pcbi.1009846] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 01/18/2022] [Indexed: 11/18/2022] Open
Abstract
We introduce cytoNet, a cloud-based tool to characterize cell populations from microscopy images. cytoNet quantifies spatial topology and functional relationships in cell communities using principles of network science. Capturing multicellular dynamics through graph features, cytoNet also evaluates the effect of cell-cell interactions on individual cell phenotypes. We demonstrate cytoNet’s capabilities in four case studies: 1) characterizing the temporal dynamics of neural progenitor cell communities during neural differentiation, 2) identifying communities of pain-sensing neurons in vivo, 3) capturing the effect of cell community on endothelial cell morphology, and 4) investigating the effect of laminin α4 on perivascular niches in adipose tissue. The analytical framework introduced here can be used to study the dynamics of complex cell communities in a quantitative manner, leading to a deeper understanding of environmental effects on cellular behavior. The versatile, cloud-based format of cytoNet makes the image analysis framework accessible to researchers across domains.
Collapse
Affiliation(s)
- Arun S. Mahadevan
- Department of Bioengineering, University of Pennsylvania; Philadelphia, Pennsylvania, United States of America
- Department of Bioengineering, Rice University, Houston, Texas, United States of America
| | - Byron L. Long
- Department of Bioengineering, Rice University, Houston, Texas, United States of America
- Department of Biomedical Engineering, University of Texas at San Antonio, San Antonio, Texas, United States of America
- Department of Computer Science, University of Texas at San Antonio, San Antonio, Texas, United States of America
| | - Chenyue W. Hu
- Department of Bioengineering, Rice University, Houston, Texas, United States of America
| | - David T. Ryan
- Department of Bioengineering, Rice University, Houston, Texas, United States of America
| | - Nicolas E. Grandel
- Systems, Synthetic and Physical Biology Program, Rice University, Houston, Texas, United States of America
| | - George L. Britton
- Systems, Synthetic and Physical Biology Program, Rice University, Houston, Texas, United States of America
| | - Marisol Bustos
- Department of Biomedical Engineering, University of Texas at San Antonio, San Antonio, Texas, United States of America
| | - Maria A. Gonzalez Porras
- Department of Biomedical Engineering, University of Texas at San Antonio, San Antonio, Texas, United States of America
| | - Katerina Stojkova
- Department of Biomedical Engineering, University of Texas at San Antonio, San Antonio, Texas, United States of America
| | - Andrew Ligeralde
- Biophysics Graduate Program, University of California, Berkeley, California, United States of America
| | - Hyeonwi Son
- Department of Oral & Maxillofacial Surgery, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - John Shannonhouse
- Department of Oral & Maxillofacial Surgery, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Jacob T. Robinson
- Department of Electrical and Computer Engineering, Rice University, Houston, Texas, United States of America
| | - Aryeh Warmflash
- Systems, Synthetic and Physical Biology Program, Rice University, Houston, Texas, United States of America
- Department of Biosciences, Rice University, Houston, Texas, United States of America
| | - Eric M. Brey
- Department of Biomedical Engineering, University of Texas at San Antonio, San Antonio, Texas, United States of America
- UTSA–UT Health Joint Graduate Group in Biomedical Engineering, San Antonio, Texas, United States of America
| | - Yu Shin Kim
- Department of Oral & Maxillofacial Surgery, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
- UTSA–UT Health Joint Graduate Group in Biomedical Engineering, San Antonio, Texas, United States of America
- Programs in Integrated Biomedical Sciences, Translational Sciences, Radiological Sciences, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Amina A. Qutub
- Department of Biomedical Engineering, University of Texas at San Antonio, San Antonio, Texas, United States of America
- UTSA–UT Health Joint Graduate Group in Biomedical Engineering, San Antonio, Texas, United States of America
- UTSA AI MATRIX Consortium, San Antonio, Texas, United States of America
- * E-mail:
| |
Collapse
|
69
|
Shen CL, Castro L, Fang CY, Castro M, Sherali S, White S, Wang R, Neugebauer V. Bioactive compounds for neuropathic pain: An update on preclinical studies and future perspectives. J Nutr Biochem 2022; 104:108979. [PMID: 35245654 DOI: 10.1016/j.jnutbio.2022.108979] [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: 09/22/2021] [Revised: 01/21/2022] [Accepted: 02/21/2022] [Indexed: 12/19/2022]
Abstract
Among different types of chronic pain, neuropathic pain (NP), arising from damage to the nervous system, including peripheral fibers and central neurons, is notoriously difficult to treat and affects 7-10% of the general population. Currently available treatment options for NP are limited and opioid analgesics have severe side effects and can result in opioid use disorder. Recent studies have exhibited the role of dietary bioactive compounds in the mitigation of NP. Here, we assessed the effects of commonly consumed bioactive compounds (ginger, curcumin, omega-3 polyunsaturated fatty acids, epigallocatechin gallate, resveratrol, soy isoflavones, lycopene, and naringin) on NP and NP-related neuroinflammation. Cellular studies demonstrated that these bioactive compounds reduce inflammation via suppression of NF-κB and MAPK signaling pathways that regulate apoptosis/cell survival, antioxidant, and anti-inflammatory responses. Animal studies strongly suggest that these regularly consumed bioactive compounds have a pronounced anti-NP effect as shown by decreased mechanical allodynia, mechanical hyperalgesia, thermal hyperalgesia, and cold hyperalgesia. The proposed molecular mechanisms include (1) the enhancement of neuron survival, (2) the reduction of neuronal hyperexcitability by activation of antinociceptive cannabinoid 1 receptors and opioid receptors, (3) the suppression of sodium channel current, and (4) enhancing a potassium outward current in NP-affected animals, triggering a cascade of chemical changes within, and between neurons for pain relief. Human studies administered in this area have been limited. Future randomized controlled trials are warranted to confirm the findings of preclinical efficacies using bioactive compounds in patients with NP.
Collapse
Affiliation(s)
- Chwan-Li Shen
- Department of Pathology, Texas Tech University Health Sciences Center, Lubbock, Texas, USA; Center of Excellence for Integrative Health, Texas Tech University Health Sciences Center, Lubbock, Texas, USA; Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, Texas, USA.
| | - Luis Castro
- School of Medicine, Texas Tech University Health Sciences, Lubbock, Texas, USA
| | - Chih-Yu Fang
- School of Medicine, Texas Tech University Health Sciences, Lubbock, Texas, USA
| | - Maribel Castro
- School of Medicine, Texas Tech University Health Sciences, Lubbock, Texas, USA
| | - Samir Sherali
- School of Medicine, Texas Tech University Health Sciences, Lubbock, Texas, USA
| | - Steely White
- Department of Microbiology, Texas Tech University, Lubbock, Texas, USA
| | - Rui Wang
- Department of Pathology, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| | - Volker Neugebauer
- Center of Excellence for Integrative Health, Texas Tech University Health Sciences Center, Lubbock, Texas, USA; Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, Texas, USA; Department of Pharmacology & Neuroscience, Texas Tech University Health Sciences Center, Lubbock, Texas, USA; Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| |
Collapse
|
70
|
Talluri B, Hoelzel F, Medda BK, Terashvili M, Sanvanson P, Shaker R, Banerjee A, Sengupta JN, Banerjee B. Identification and characterization of rostral ventromedial medulla neurons synaptically connected to the urinary bladder afferents in female rats with or without neonatal cystitis. J Comp Neurol 2022; 530:1129-1147. [PMID: 34628661 PMCID: PMC8967775 DOI: 10.1002/cne.25260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 09/29/2021] [Accepted: 10/04/2021] [Indexed: 11/06/2022]
Abstract
The neurons in the rostral ventromedial medulla (RVM) play a major role in pain modulation. We have previously shown that early-life noxious bladder stimuli in rats resulted in an overall spinal GABAergic disinhibition and a long-lasting bladder/colon sensitization when tested in adulthood. However, the neuromolecular alterations within RVM neurons in the pathophysiology of early life bladder inflammation have not been elucidated. In this study, we have identified and characterized RVM neurons that are synaptically linked to the bladder and colon and examined the effect of neonatal bladder inflammation on molecular expressions of these neurons. A transient bladder inflammation was induced by intravesicular instillation of protamine sulfate and zymosan during postnatal days 14 through 16 (P14-16) followed by pseudorabies virus PRV-152 and PRV-614 injections into the bladder and colon, respectively, on postnatal day P60. Tissues were examined 96 h postinoculation for serotonergic, GABAergic, and enkephalinergic expressions using in situ hybridization and/or immunohistochemistry techniques. The results revealed that > 50% of RVM neurons that are synaptically connected to the bladder (i.e., PRV-152+) were GABAergic, 40% enkephalinergic, and about 14% expressing serotonergic marker tryptophan hydroxylase 2 (TpH2). Neonatal cystitis resulted in a significant increase in converging neurons in RVM receiving dual synaptic inputs from the bladder and colon. In addition, neonatal cystitis significantly downregulated vesicular GABA transporter (VGAT) with a concomitant increase in TpH2 expression in bladder-linked RVM neurons, suggesting an alteration in supraspinal signaling. These alterations of synaptic connectivity and GABAergic/serotonergic expressions in RVM neurons may contribute to bladder pain modulation and cross-organ visceral sensitivity.
Collapse
Affiliation(s)
- Bhavana Talluri
- Division of Gastroenterology and Hepatology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Faith Hoelzel
- Division of Gastroenterology and Hepatology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Bidyut K. Medda
- Division of Gastroenterology and Hepatology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Maia Terashvili
- Division of Gastroenterology and Hepatology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Patrick Sanvanson
- Division of Gastroenterology and Hepatology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Reza Shaker
- Division of Gastroenterology and Hepatology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Anjishnu Banerjee
- Division of Biostatistics, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Jyoti N. Sengupta
- Division of Gastroenterology and Hepatology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Banani Banerjee
- Division of Gastroenterology and Hepatology, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
71
|
Gupta S, Gautam S, Kumar U, Arora T, Dada R. Potential Role of Yoga Intervention in the Management of Chronic Non-malignant Pain. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:5448671. [PMID: 35668780 PMCID: PMC9167073 DOI: 10.1155/2022/5448671] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 03/18/2022] [Accepted: 05/13/2022] [Indexed: 12/22/2022]
Abstract
Pain is an unpleasant and upsetting experience. Persistent pain has an impact on an individual's quality of life which causes stress and mood disorders. There are currently no pain-relieving techniques available that can eliminate pain and offer relief without causing any adverse effects. These factors draw attention to traditional treatments like yoga and meditation, which can reduce biological stress and hence increase immunity, as well as alleviate the psychological and emotional suffering produced by pain. Yoga reduces the stress response and the pain cascade via the downregulation of the hypothalamus-pituitary-adrenal (HPA) axis and vagal stimulation. Yoga is a cost-effective growing health practice that, unlike pharmaceuticals, has no side effects and can help patients stay in remission for longer periods of time with fewer relapses. Yoga not only reduces stress and depression severity but also improves functional status and reduces pain perception. This article highlights the impact of yoga on pain management and on a malfunctioning immune system, which leads to improved health, pain reduction, disease management, and improvement in overall quality of life.
Collapse
Affiliation(s)
- Shivani Gupta
- Laboratory for Molecular Reproduction and Genetics, Department of Anatomy, All India Institute of Medical Sciences, New Delhi, India
| | - Surabhi Gautam
- Laboratory for Molecular Reproduction and Genetics, Department of Anatomy, All India Institute of Medical Sciences, New Delhi, India
| | - Uma Kumar
- Department of Rheumatology, All India Institute of Medical Sciences, New Delhi, India
| | - Taruna Arora
- Division of Reproductive Biology, Maternal & Child Health, Indian Council of Medical Research, New Delhi, India
| | - Rima Dada
- Laboratory for Molecular Reproduction and Genetics, Department of Anatomy, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
72
|
Zhou F, Metzner K, Engel P, Balzulat A, Sisignano M, Ruth P, Lukowski R, Schmidtko A, Lu R. Slack Potassium Channels Modulate TRPA1-Mediated Nociception in Sensory Neurons. Cells 2022; 11:cells11101693. [PMID: 35626730 PMCID: PMC9140117 DOI: 10.3390/cells11101693] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 05/15/2022] [Accepted: 05/17/2022] [Indexed: 12/13/2022] Open
Abstract
The transient receptor potential (TRP) ankyrin type 1 (TRPA1) channel is highly expressed in a subset of sensory neurons where it acts as an essential detector of painful stimuli. However, the mechanisms that control the activity of sensory neurons upon TRPA1 activation remain poorly understood. Here, using in situ hybridization and immunostaining, we found TRPA1 to be extensively co-localized with the potassium channel Slack (KNa1.1, Slo2.2, or Kcnt1) in sensory neurons. Mice lacking Slack globally (Slack−/−) or conditionally in sensory neurons (SNS-Slack−/−) demonstrated increased pain behavior after intraplantar injection of the TRPA1 activator allyl isothiocyanate. By contrast, pain behavior induced by the TRP vanilloid 1 (TRPV1) activator capsaicin was normal in Slack-deficient mice. Patch-clamp recordings in sensory neurons and in a HEK cell line transfected with TRPA1 and Slack revealed that Slack-dependent potassium currents (IKS) are modulated in a TRPA1-dependent manner. Taken together, our findings highlight Slack as a modulator of TRPA1-mediated, but not TRPV1-mediated, activation of sensory neurons.
Collapse
Affiliation(s)
- Fangyuan Zhou
- Institute of Pharmacology and Clinical Pharmacy, Goethe University, 60438 Frankfurt am Main, Germany; (F.Z.); (K.M.); (P.E.); (A.B.); (A.S.)
| | - Katharina Metzner
- Institute of Pharmacology and Clinical Pharmacy, Goethe University, 60438 Frankfurt am Main, Germany; (F.Z.); (K.M.); (P.E.); (A.B.); (A.S.)
| | - Patrick Engel
- Institute of Pharmacology and Clinical Pharmacy, Goethe University, 60438 Frankfurt am Main, Germany; (F.Z.); (K.M.); (P.E.); (A.B.); (A.S.)
| | - Annika Balzulat
- Institute of Pharmacology and Clinical Pharmacy, Goethe University, 60438 Frankfurt am Main, Germany; (F.Z.); (K.M.); (P.E.); (A.B.); (A.S.)
| | - Marco Sisignano
- Institute of Clinical Pharmacology, Pharmazentrum Frankfurt/ZAFES, Goethe University, 60590 Frankfurt am Main, Germany;
| | - Peter Ruth
- Department of Pharmacology, Toxicology and Clinical Pharmacy, University of Tübingen, 72076 Tübingen, Germany; (P.R.); (R.L.)
| | - Robert Lukowski
- Department of Pharmacology, Toxicology and Clinical Pharmacy, University of Tübingen, 72076 Tübingen, Germany; (P.R.); (R.L.)
| | - Achim Schmidtko
- Institute of Pharmacology and Clinical Pharmacy, Goethe University, 60438 Frankfurt am Main, Germany; (F.Z.); (K.M.); (P.E.); (A.B.); (A.S.)
| | - Ruirui Lu
- Institute of Pharmacology and Clinical Pharmacy, Goethe University, 60438 Frankfurt am Main, Germany; (F.Z.); (K.M.); (P.E.); (A.B.); (A.S.)
- Correspondence: ; Tel.: +49-69-798-29377
| |
Collapse
|
73
|
Zhang YY, Liu F, Lin J, Li YL, Fang ZH, Zhou C, Li CJ, Shen JF. Activation of the N-methyl-D-aspartate receptor contributes to orofacial neuropathic and inflammatory allodynia by facilitating calcium-calmodulin-dependent protein kinase II phosphorylation in mice. Brain Res Bull 2022; 185:174-192. [DOI: 10.1016/j.brainresbull.2022.05.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 02/28/2022] [Accepted: 05/04/2022] [Indexed: 12/17/2022]
|
74
|
Wank I, Kutsche L, Kreitz S, Reeh P, Hess A. Imaging the influence of peripheral TRPV1-signaling on cerebral nociceptive processing applying fMRI-based graph theory in a resiniferatoxin rat model. PLoS One 2022; 17:e0266669. [PMID: 35482725 PMCID: PMC9049522 DOI: 10.1371/journal.pone.0266669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 03/24/2022] [Indexed: 11/19/2022] Open
Abstract
Resiniferatoxin (RTX), an extract from the spurge plant Euphorbia resinifera, is a potent agonist of the transient receptor potential cation channel subfamily V member 1 (TRPV1), mainly expressed on peripheral nociceptors-a prerequisite for nociceptive heat perception. Systemic overdosing of RTX can be used to desensitize specifically TRPV1-expressing neurons, and was therefore utilized here to selectively characterize the influence of TRPV1-signaling on central nervous system (CNS) temperature processing. Resting state and CNS temperature processing of male rats were assessed via functional magnetic resonance imaging before and after RTX injection. General linear model-based and graph-theoretical network analyses disentangled the underlying distinct CNS circuitries. At baseline, rats displayed an increase of nociception-related response amplitude and activated brain volume that correlated highly with increasing stimulation temperatures. In contrast, RTX-treated rats showed a clear disruption of thermal nociception, reflected in a missing increase of CNS responses to temperatures above 48°C. Graph-theoretical analyses revealed two distinct brain subnetworks affected by RTX: one subcortical (brainstem, lateral and medial thalamus, hippocampus, basal ganglia and amygdala), and one cortical (primary sensory, motor and association cortices). Resting state analysis revealed first, that peripheral desensitization of TRPV1-expressing neurons did not disrupt the basic resting-state-network of the brain. Second, only at baseline, but not after RTX, noxious stimulation modulated the RS-network in regions associated with memory formation (e.g. hippocampus). Altogether, the combination of whole-brain functional magnetic resonance imaging and RTX-mediated desensitization of TRPV1-signaling provided further detailed insight into cerebral processing of noxious temperatures.
Collapse
Affiliation(s)
- Isabel Wank
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Lisa Kutsche
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Silke Kreitz
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Peter Reeh
- Institute of Physiology and Pathophysiology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Andreas Hess
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
75
|
Hu M, Doyle AD, Yamada KM, Kulkarni AB. Visualization of trigeminal ganglion sensory neuronal signaling regulated by Cdk5. Cell Rep 2022; 38:110458. [PMID: 35263573 PMCID: PMC9004325 DOI: 10.1016/j.celrep.2022.110458] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 11/24/2021] [Accepted: 02/08/2022] [Indexed: 02/04/2023] Open
Abstract
The mechanisms underlying facial pain are still incompletely understood, posing major therapeutic challenges. Cyclin-dependent kinase 5 (Cdk5) is a key neuronal kinase involved in pain signaling. However, the regulatory roles of Cdk5 in facial pain signaling and the possibility of therapeutic intervention at the level of mouse trigeminal ganglion primary neurons remain elusive. In this study, we use optimized intravital imaging to directly compare trigeminal neuronal activities after mechanical, thermal, and chemical stimulation. We then test whether facial inflammatory pain in mice could be alleviated by the Cdk5 inhibitor peptide TFP5. We demonstrate regulation of total Ca2+ intensity by Cdk5 activity using transgenic and knockout mouse models. In mice with vibrissal pad inflammation, application of TFP5 specifically decreases total Ca2+ intensity in response to noxious stimuli. It also alleviates inflammation-induced allodynia by inhibiting activation of trigeminal peripheral sensory neurons. Cdk5 inhibitors may provide promising non-opioid candidates for pain treatment.
Collapse
Affiliation(s)
- Minghan Hu
- Functional Genomics Section and Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Andrew D Doyle
- Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Kenneth M Yamada
- Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA.
| | - Ashok B Kulkarni
- Functional Genomics Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, MSC 4359, Bethesda, MD, USA.
| |
Collapse
|
76
|
Abstract
Transient receptor potential vanilloid type 1 (TRPV1) is a nonselective cation channel that is intensively expressed in the peripheral nerve system and involved in a variety of physiological and pathophysiological processes in mammals. Its activity is of great significance in transmitting pain or itch signals from peripheral sensory neurons to the central nervous system. The alteration or hypersensitivity of TRPV1 channel is well evidenced under various pathological conditions. Moreover, accumulative studies have revealed that TRPV1-expressing (TRPV1+) sensory neurons mediate the neuroimmune crosstalk by releasing neuropeptides to innervated tissues as well as immune cells. In the central projection, TRPV1+ terminals synapse with the secondary neurons for the transmission of pain and itch signalling. The intense involvement of TRPV1 and TRPV1+ neurons in pain and itch makes it a potential pharmaceutical target. Over decades, the basis of TRPV1 channel structure, the nature of its activity, and its modulation in pathological processes have been broadly studied and well documented. Herein, we highlight the role of TRPV1 and its associated neurons in sensing pain and itch. The fundamental understandings of TRPV1-involved nociception, pruriception, neurogenic inflammation, and cell-specific modulation will help bring out more effective strategies of TRPV1 modulation in treating pain- and itch-related diseases.
Collapse
|
77
|
Antoniazzi C, Belinskaia M, Zurawski T, Kaza SK, Dolly JO, Lawrence GW. Botulinum Neurotoxin Chimeras Suppress Stimulation by Capsaicin of Rat Trigeminal Sensory Neurons In Vivo and In Vitro. Toxins (Basel) 2022; 14:116. [PMID: 35202143 PMCID: PMC8878885 DOI: 10.3390/toxins14020116] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/14/2022] [Accepted: 02/01/2022] [Indexed: 02/06/2023] Open
Abstract
Chimeras of botulinum neurotoxin (BoNT) serotype A (/A) combined with /E protease might possess improved analgesic properties relative to either parent, due to inheriting the sensory neurotropism of the former with more extensive disabling of SNAP-25 from the latter. Hence, fusions of /E protease light chain (LC) to whole BoNT/A (LC/E-BoNT/A), and of the LC plus translocation domain (HN) of /E with the neuronal acceptor binding moiety (HC) of /A (BoNT/EA), created previously by gene recombination and expression in E. coli., were used. LC/E-BoNT/A (75 units/kg) injected into the whisker pad of rats seemed devoid of systemic toxicity, as reflected by an absence of weight loss, but inhibited the nocifensive behavior (grooming, freezing, and reduced mobility) induced by activating TRPV1 with capsaicin, injected at various days thereafter. No sex-related differences were observed. c-Fos expression was increased five-fold in the trigeminal nucleus caudalis ipsi-lateral to capsaicin injection, relative to the contra-lateral side and vehicle-treated controls, and this increase was virtually prevented by LC/E-BoNT/A. In vitro, LC/E-BoNT/A or /EA diminished CGRP exocytosis from rat neonate trigeminal ganglionic neurons stimulated with up to 1 µM capsaicin, whereas BoNT/A only substantially reduced the release in response to 0.1 µM or less of the stimulant, in accordance with the /E protease being known to prevent fusion of exocytotic vesicles.
Collapse
Affiliation(s)
| | | | | | | | | | - Gary W. Lawrence
- International Centre for Neurotherapeutics, Dublin City University, Collins Avenue, D09 V209 Dublin, Ireland; (C.A.); (M.B.); (T.Z.); (S.K.K.); (J.O.D.)
| |
Collapse
|
78
|
Li F, Wang C, Hu D, Zhang X, Shen R, Zhou Y, Yang Y, Zhu C, Tang Z, Yu G. mMrgprA3
/
mMrgprC11
/
hMrgprX1
: potential therapeutic targets for allergic contact dermatitis induced pruritus in mice and human. Contact Dermatitis 2022; 86:286-294. [DOI: 10.1111/cod.14051] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 01/06/2022] [Accepted: 01/18/2022] [Indexed: 11/27/2022]
Affiliation(s)
- Fengxian Li
- Department of Anesthesiology Zhujiang Hospital of Southern Medical University Guangzhou Guangdong China
| | - Changming Wang
- School of Medicine & Holistic Integrative Medicine Nanjing University of Chinese Medicine Nanjing Jiangsu China
- Key Laboratory for Chinese Medicine of Prevention and Treatment in Neurological Diseases Nanjing University of Chinese Medicine Nanjing Jiangsu China
| | - Danyou Hu
- School of Medicine & Holistic Integrative Medicine Nanjing University of Chinese Medicine Nanjing Jiangsu China
- Key Laboratory for Chinese Medicine of Prevention and Treatment in Neurological Diseases Nanjing University of Chinese Medicine Nanjing Jiangsu China
| | - Xinyu Zhang
- School of Medicine & Holistic Integrative Medicine Nanjing University of Chinese Medicine Nanjing Jiangsu China
- Key Laboratory for Chinese Medicine of Prevention and Treatment in Neurological Diseases Nanjing University of Chinese Medicine Nanjing Jiangsu China
| | - Ran Shen
- School of Medicine & Holistic Integrative Medicine Nanjing University of Chinese Medicine Nanjing Jiangsu China
- Key Laboratory for Chinese Medicine of Prevention and Treatment in Neurological Diseases Nanjing University of Chinese Medicine Nanjing Jiangsu China
| | - Yuan Zhou
- School of Medicine & Holistic Integrative Medicine Nanjing University of Chinese Medicine Nanjing Jiangsu China
- Key Laboratory for Chinese Medicine of Prevention and Treatment in Neurological Diseases Nanjing University of Chinese Medicine Nanjing Jiangsu China
| | - Yan Yang
- School of Medicine & Holistic Integrative Medicine Nanjing University of Chinese Medicine Nanjing Jiangsu China
- Key Laboratory for Chinese Medicine of Prevention and Treatment in Neurological Diseases Nanjing University of Chinese Medicine Nanjing Jiangsu China
| | - Chan Zhu
- School of Medicine & Holistic Integrative Medicine Nanjing University of Chinese Medicine Nanjing Jiangsu China
- Key Laboratory for Chinese Medicine of Prevention and Treatment in Neurological Diseases Nanjing University of Chinese Medicine Nanjing Jiangsu China
| | - Zongxiang Tang
- School of Medicine & Holistic Integrative Medicine Nanjing University of Chinese Medicine Nanjing Jiangsu China
- Key Laboratory for Chinese Medicine of Prevention and Treatment in Neurological Diseases Nanjing University of Chinese Medicine Nanjing Jiangsu China
| | - Guang Yu
- School of Medicine & Holistic Integrative Medicine Nanjing University of Chinese Medicine Nanjing Jiangsu China
- Key Laboratory for Chinese Medicine of Prevention and Treatment in Neurological Diseases Nanjing University of Chinese Medicine Nanjing Jiangsu China
| |
Collapse
|
79
|
Iannone LF, De Logu F, Geppetti P, De Cesaris F. The role of TRP ion channels in migraine and headache. Neurosci Lett 2022; 768:136380. [PMID: 34861342 DOI: 10.1016/j.neulet.2021.136380] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 10/08/2021] [Accepted: 10/17/2021] [Indexed: 12/15/2022]
Abstract
Migraine afflicts more than 10% of the general population. Although its mechanism is poorly understood, recent preclinical and clinical evidence has identified calcitonin gene related peptide (CGRP) as a major mediator of migraine pain. CGRP, which is predominantly expressed in a subset of primary sensory neurons, including trigeminal afferents, when released from peripheral terminals of nociceptors, elicits arteriolar vasodilation and mechanical allodynia, a hallmark of migraine attack. Transient receptor potential (TRP) channels include several cationic channels with pleiotropic functions and ubiquitous distribution in various cells and tissues. Some members of the TRP channel family, such as the ankyrin 1 (TRPA1), vanilloid 1 and 4 (TRPV1 and TRPV4, respectively), and TRPM3, are abundantly expressed in primary sensory neurons and are recognized as sensors of chemical-, heat- and mechanical-induced pain, and play a primary role in several models of pain diseases, including inflammatory, neuropathic cancer pain, and migraine pain. In addition, TRP channel stimulation results in CGRP release, which can be activated or sensitized by various endogenous and exogenous stimuli, some of which have been proven to trigger or worsen migraine attacks. Moreover, some antimigraine medications seem to act through TRPA1 antagonism. Here we review the preclinical and clinical evidence that highlights the role of TRP channels, and mainly TRPA1, in migraine pathophysiology and may be proposed as new targets for its treatment.
Collapse
Affiliation(s)
- Luigi Francesco Iannone
- Headache Center and Clinical Pharmacology Unit, Careggi University Hospital, Florence, Italy
| | - Francesco De Logu
- Section of Clinical Pharmacology and Oncology, Department of Health Sciences, University of Florence, Florence, Italy
| | - Pierangelo Geppetti
- Headache Center and Clinical Pharmacology Unit, Careggi University Hospital, Florence, Italy; Section of Clinical Pharmacology and Oncology, Department of Health Sciences, University of Florence, Florence, Italy
| | - Francesco De Cesaris
- Headache Center and Clinical Pharmacology Unit, Careggi University Hospital, Florence, Italy.
| |
Collapse
|
80
|
Belinskaia M, Zurawski T, Kaza SK, Antoniazzi C, Dolly JO, Lawrence GW. NGF Enhances CGRP Release Evoked by Capsaicin from Rat Trigeminal Neurons: Differential Inhibition by SNAP-25-Cleaving Proteases. Int J Mol Sci 2022; 23:ijms23020892. [PMID: 35055082 PMCID: PMC8778182 DOI: 10.3390/ijms23020892] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/08/2022] [Accepted: 01/11/2022] [Indexed: 11/23/2022] Open
Abstract
Nerve growth factor (NGF) is known to intensify pain in various ways, so perturbing pertinent effects without negating its essential influences on neuronal functions could help the search for much-needed analgesics. Towards this goal, cultured neurons from neonatal rat trigeminal ganglia—a locus for craniofacial sensory nerves—were used to examine how NGF affects the Ca2+-dependent release of a pain mediator, calcitonin gene-related peptide (CGRP), that is triggered by activating a key signal transducer, transient receptor potential vanilloid 1 (TRPV1) with capsaicin (CAP). Measurements utilised neurons fed with or deprived of NGF for 2 days. Acute re-introduction of NGF induced Ca2+-dependent CGRP exocytosis that was inhibited by botulinum neurotoxin type A (BoNT/A) or a chimera of/E and/A (/EA), which truncated SNAP-25 (synaptosomal-associated protein with Mr = 25 k) at distinct sites. NGF additionally caused a Ca2+-independent enhancement of the neuropeptide release evoked by low concentrations (<100 nM) of CAP, but only marginally increased the peak response to ≥100 nM. Notably, BoNT/A inhibited CGRP exocytosis evoked by low but not high CAP concentrations, whereas/EA effectively reduced responses up to 1 µM CAP and inhibited to a greater extent its enhancement by NGF. In addition to establishing that sensitisation of sensory neurons to CAP by NGF is dependent on SNARE-mediated membrane fusion, insights were gleaned into the differential ability of two regions in the C-terminus of SNAP-25 (181–197 and 198–206) to support CAP-evoked Ca2+-dependent exocytosis at different intensities of stimulation.
Collapse
|
81
|
Iseppon F, Linley JE, Wood JN. Calcium imaging for analgesic drug discovery. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2022; 11:100083. [PMID: 35079661 PMCID: PMC8777277 DOI: 10.1016/j.ynpai.2021.100083] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/17/2021] [Accepted: 12/27/2021] [Indexed: 11/24/2022]
Abstract
Somatosensation and pain are complex phenomena involving a rangeofspecialised cell types forming different circuits within the peripheral and central nervous systems. In recent decades, advances in the investigation of these networks, as well as their function in sensation, resulted from the constant evolution of electrophysiology and imaging techniques to allow the observation of cellular activity at the population level both in vitro and in vivo. Genetically encoded indicators of neuronal activity, combined with recent advances in DNA engineering and modern microscopy, offer powerful tools to dissect and visualise the activity of specific neuronal subpopulations with high spatial and temporal resolution. In recent years various groups developed in vivo imaging techniques to image calcium transients in the dorsal root ganglia, the spinal cord and the brain of anesthetised and awake, behaving animals to address fundamental questions in both the physiology and pathophysiology of somatosensation and pain. This approach, besides giving unprecedented details on the circuitry of innocuous and painful sensation, can be a very powerful tool for pharmacological research, from the characterisation of new potential drugs to the discovery of new, druggable targets within specific neuronal subpopulations. Here we summarise recent developments in calcium imaging for pain research, discuss technical challenges and advances, and examine the potential positive impact of this technique in early preclinical phases of the analgesic drug discovery process.
Collapse
Affiliation(s)
- Federico Iseppon
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, Gower Street, WC1E 6BT London, UK
- Discovery UK, Neuroscience, Biopharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - John E. Linley
- Discovery UK, Neuroscience, Biopharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - John N. Wood
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, Gower Street, WC1E 6BT London, UK
| |
Collapse
|
82
|
Zheng XQ, Wu YH, Huang JF, Wu AM. Neurophysiological mechanisms of cancer-induced bone pain. J Adv Res 2022; 35:117-127. [PMID: 35003797 PMCID: PMC8721251 DOI: 10.1016/j.jare.2021.06.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 05/23/2021] [Accepted: 06/08/2021] [Indexed: 12/16/2022] Open
Abstract
Background Cancer-induced Bone Pain (CIBP) is an important factor affecting their quality of life of cancer survivors. In addition, current clinical practice and scientific research suggest that neuropathic pain is a representative component of CIBP. However, given the variability of cancer conditions and the complexity of neuropathic pain, related mechanisms have been continuously supplemented but have not been perfected. Aim of Review Therefore, the current review highlights the latest progress in basic research on the field and proposes potential therapeutic targets, representative drugs and upcoming therapies. Key Scientific Concepts of Review Notably, factors such as central sensitization, neuroinflammation, glial cell activation and an acidic environment are considered to be related to neuropathic pain in CIBP. Nonetheless, further research is needed to ascertain the mechanism of CIBP in order to develop highly effective drugs. Moreover, more attention needs to be paid to the care of patients with advanced cancer.
Collapse
Affiliation(s)
- Xuan-Qi Zheng
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, Zhejiang, 325027, China
- Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Yu-hao Wu
- Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Jin-feng Huang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, Zhejiang, 325027, China
- Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Ai-Min Wu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, Zhejiang, 325027, China
- Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| |
Collapse
|
83
|
Puja G, Sonkodi B, Bardoni R. Mechanisms of Peripheral and Central Pain Sensitization: Focus on Ocular Pain. Front Pharmacol 2021; 12:764396. [PMID: 34916942 PMCID: PMC8669969 DOI: 10.3389/fphar.2021.764396] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 11/09/2021] [Indexed: 12/14/2022] Open
Abstract
Persistent ocular pain caused by corneal inflammation and/or nerve injury is accompanied by significant alterations along the pain axis. Both primary sensory neurons in the trigeminal nerves and secondary neurons in the spinal trigeminal nucleus are subjected to profound morphological and functional changes, leading to peripheral and central pain sensitization. Several studies using animal models of inflammatory and neuropathic ocular pain have provided insight about the mechanisms involved in these maladaptive changes. Recently, the advent of new techniques such as optogenetics or genetic neuronal labelling has allowed the investigation of identified circuits involved in nociception, both at the spinal and trigeminal level. In this review, we will describe some of the mechanisms that contribute to the perception of ocular pain at the periphery and at the spinal trigeminal nucleus. Recent advances in the discovery of molecular and cellular mechanisms contributing to peripheral and central pain sensitization of the trigeminal pathways will be also presented.
Collapse
Affiliation(s)
- Giulia Puja
- Department of Life Sciences, University of Modena and Reggio Emilia, Emilia-Romagna, Italy
| | - Balazs Sonkodi
- Department of Health Sciences and Sport Medicine, University of Physical Education, Budapest, Hungary
| | - Rita Bardoni
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Emilia-Romagna, Italy
| |
Collapse
|
84
|
Crawford J, Liu S, Tao F. A Multidisciplinary Approach to Simultaneously Monitoring Real-Time Neuronal Activity and Pain Behaviors During Optogenetic Stimulation of Brain Neurons in Freely Moving Mice. J Pain Res 2021; 14:3503-3509. [PMID: 34785947 PMCID: PMC8590449 DOI: 10.2147/jpr.s334256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 10/09/2021] [Indexed: 11/23/2022] Open
Abstract
Background Highlighted by the current opioid epidemic, identifying novel therapies to treat chronic trigeminal neuropathic pain is a critical need. To develop these treatments, it is necessary to have viable targets in the brain to act on. Historically, neural tracing studies have been extremely useful in determining connections between brain areas but do not provide information about the functionality of these connections. Combining optogenetics and behavioral observation allows researchers to determine whether a particular brain area is involved in the regulation of such behavior. The addition of multi-channel electrophysiological recording provides information on real-time neuronal activity in the specific neuronal pathway. Methods Male C57/BL/6J mice (8-week-old) underwent either chronic constriction injury of infraorbital nerve (CCI-ION) or a sham surgery and were injected with either channelrhodopsin (ChR2) or a control virus in the hypothalamic A11 nucleus. Two weeks after CCI-ION, they were tested in real-time place preference (RTPP), while neuronal activity in the spinal trigeminal nucleus caudalis (Sp5C) was recorded. Results Optogenetic excitation of the A11 neurons results in more time spent in the stimulation chamber during RTPP testing. Additionally, stimulation of the A11 results in a greater number of neuronal activity increase in the Sp5C in animals with the injection of AAV carrying ChR2 compared to animals injected with a control virus or that underwent a sham surgery. Conclusion In vivo multi-channel electrophysiological recording, optogenetic stimulation, and behavioral observation can be combined in a mouse model of chronic trigeminal neuropathic pain to validate brain areas involved in the modulation of such pain.
Collapse
Affiliation(s)
- Joshua Crawford
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX, 75246, USA
| | - Sufang Liu
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX, 75246, USA
| | - Feng Tao
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX, 75246, USA
| |
Collapse
|
85
|
Mercer Lindsay N, Chen C, Gilam G, Mackey S, Scherrer G. Brain circuits for pain and its treatment. Sci Transl Med 2021; 13:eabj7360. [PMID: 34757810 DOI: 10.1126/scitranslmed.abj7360] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Nicole Mercer Lindsay
- Department of Cell Biology and Physiology, UNC Neuroscience Center, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,Department of Biology, CNC Program, Department of Applied Physics, Stanford University, Stanford, CA 94305, USA
| | - Chong Chen
- Department of Cell Biology and Physiology, UNC Neuroscience Center, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Gadi Gilam
- Division of Pain Medicine, Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Sean Mackey
- Division of Pain Medicine, Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Grégory Scherrer
- Department of Cell Biology and Physiology, UNC Neuroscience Center, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,New York Stem Cell Foundation-Robertson Investigator, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
86
|
Cui W, Wu H, Yu X, Song T, Xu X, Xu F. The Calcium Channel α2δ1 Subunit: Interactional Targets in Primary Sensory Neurons and Role in Neuropathic Pain. Front Cell Neurosci 2021; 15:699731. [PMID: 34658790 PMCID: PMC8514986 DOI: 10.3389/fncel.2021.699731] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 09/10/2021] [Indexed: 11/13/2022] Open
Abstract
Neuropathic pain is mainly triggered after nerve injury and associated with plasticity of the nociceptive pathway in primary sensory neurons. Currently, the treatment remains a challenge. In order to identify specific therapeutic targets, it is necessary to clarify the underlying mechanisms of neuropathic pain. It is well established that primary sensory neuron sensitization (peripheral sensitization) is one of the main components of neuropathic pain. Calcium channels act as key mediators in peripheral sensitization. As the target of gabapentin, the calcium channel subunit α2δ1 (Cavα2δ1) is a potential entry point in neuropathic pain research. Numerous studies have demonstrated that the upstream and downstream targets of Cavα2δ1 of the peripheral primary neurons, including thrombospondins, N-methyl-D-aspartate receptors, transient receptor potential ankyrin 1 (TRPA1), transient receptor potential vanilloid family 1 (TRPV1), and protein kinase C (PKC), are involved in neuropathic pain. Thus, we reviewed and discussed the role of Cavα2δ1 and the associated signaling axis in neuropathic pain conditions.
Collapse
Affiliation(s)
- Wenqiang Cui
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Hongyun Wu
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiaowen Yu
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ting Song
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiangqing Xu
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Fei Xu
- Department of Geriatric Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
87
|
Iwai H, Ataka K, Suzuki H, Dhar A, Kuramoto E, Yamanaka A, Goto T. Tissue-resident M2 macrophages directly contact primary sensory neurons in the sensory ganglia after nerve injury. J Neuroinflammation 2021; 18:227. [PMID: 34645458 PMCID: PMC8513227 DOI: 10.1186/s12974-021-02283-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 09/27/2021] [Indexed: 05/13/2023] Open
Abstract
Background Macrophages in the peripheral nervous system are key players in the repair of nerve tissue and the development of neuropathic pain due to peripheral nerve injury. However, there is a lack of information on the origin and morphological features of macrophages in sensory ganglia after peripheral nerve injury, unlike those in the brain and spinal cord. We analyzed the origin and morphological features of sensory ganglionic macrophages after nerve ligation or transection using wild-type mice and mice with bone-marrow cell transplants. Methods After protecting the head of C57BL/6J mice with lead caps, they were irradiated and transplanted with bone-marrow-derived cells from GFP transgenic mice. The infraorbital nerve of a branch of the trigeminal nerve of wild-type mice was ligated or the infraorbital nerve of GFP-positive bone-marrow-cell-transplanted mice was transected. After immunostaining the trigeminal ganglion, the structures of the ganglionic macrophages, neurons, and satellite glial cells were analyzed using two-dimensional or three-dimensional images. Results The number of damaged neurons in the trigeminal ganglion increased from day 1 after infraorbital nerve ligation. Ganglionic macrophages proliferated from days 3 to 5. Furthermore, the numbers of macrophages increased from days 3 to 15. Bone-marrow-derived macrophages increased on day 7 after the infraorbital nerve was transected in the trigeminal ganglion of GFP-positive bone-marrow-cell-transplanted mice but most of the ganglionic macrophages were composed of tissue-resident cells. On day 7 after infraorbital nerve ligation, ganglionic macrophages increased in volume, extended their processes between the neurons and satellite glial cells, and contacted these neurons. Most of the ganglionic macrophages showed an M2 phenotype when contact was observed, and little neuronal cell death occurred. Conclusion Most of the macrophages that appear after a nerve injury are tissue-resident, and these make direct contact with damaged neurons that act in a tissue-protective manner in the M2 phenotype. These results imply that tissue-resident macrophages signal to neurons directly through physical contact. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02283-z.
Collapse
Affiliation(s)
- Haruki Iwai
- Department of Oral Anatomy and Cell Biology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, Kagoshima, 890-8544, Japan.
| | - Koji Ataka
- Department of Psychosomatic Internal Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, Kagoshima, 890-8544, Japan.,Laboratory of Medical Biochemistry, Kobe Pharmaceutical University, 4-19-1 Motoyamakita-machi, Higashinada-ku, Kobe, 658-8558, Japan
| | - Hajime Suzuki
- Department of Oral and Maxillofacial Surgery, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, Kagoshima, 890-8544, Japan
| | - Ashis Dhar
- Department of Oral Anatomy and Cell Biology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, Kagoshima, 890-8544, Japan
| | - Eriko Kuramoto
- Department of Oral Anatomy and Cell Biology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, Kagoshima, 890-8544, Japan
| | - Atsushi Yamanaka
- Department of Oral Anatomy and Cell Biology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, Kagoshima, 890-8544, Japan
| | - Tetsuya Goto
- Department of Oral Anatomy and Cell Biology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, Kagoshima, 890-8544, Japan
| |
Collapse
|
88
|
Ishida H, Zhang Y, Gomez R, Shannonhouse J, Son H, Banik R, Kim YS. In Vivo Calcium Imaging Visualizes Incision-Induced Primary Afferent Sensitization and Its Amelioration by Capsaicin Pretreatment. J Neurosci 2021; 41:8494-8507. [PMID: 34452938 PMCID: PMC8513701 DOI: 10.1523/jneurosci.0457-21.2021] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 07/27/2021] [Accepted: 08/23/2021] [Indexed: 02/06/2023] Open
Abstract
Previous studies have shown that infiltration of capsaicin into the surgical site can prevent incision-induced spontaneous pain like behaviors and heat hyperalgesia. In the present study, we aimed to monitor primary sensory neuron Ca2+ activity in the intact dorsal root ganglia (DRG) using Pirt-GCaMP3 male and female mice pretreated with capsaicin or vehicle before the plantar incision. Intraplantar injection of capsaicin (0.05%) significantly attenuated spontaneous pain, mechanical, and heat hypersensitivity after plantar incision. The Ca2+ response in in vivo DRG and in in situ spinal cord was significantly enhanced in the ipsilateral side compared with contralateral side or naive control. Primary sensory nerve fiber length was significantly decreased in the incision skin area in capsaicin-pretreated animals detected by immunohistochemistry and placental alkaline phosphatase (PLAP) staining. Thus, capsaicin pretreatment attenuates incisional pain by suppressing Ca2+ response because of degeneration of primary sensory nerve fibers in the skin.SIGNIFICANCE STATEMENT Postoperative surgery pain is a major health and economic problem worldwide with ∼235 million major surgical procedures annually. Approximately 50% of these patients report uncontrolled or poorly controlled postoperative pain. However, mechanistic studies of postoperative surgery pain in primary sensory neurons have been limited to in vitro models or small numbers of neurons. Using an innovative, distinctive, and interdisciplinary in vivo populational dorsal root ganglia (DRG) imaging (>1800 neurons/DRG) approach, we revealed increased DRG neuronal Ca2+ activity from postoperative pain mouse model. This indicates widespread DRG primary sensory neuron plasticity. Increased neuronal Ca2+ activity occurs among various sizes of neurons but mostly in small-diameter and medium-diameter nociceptors. Capsaicin pretreatment as a therapeutic option significantly attenuates Ca2+ activity and postoperative pain.
Collapse
Affiliation(s)
- Hirotake Ishida
- Department of Oral and Maxillofacial Surgery, University of Texas Health and Science Center at San Antonio, Texas, 78229
| | - Yan Zhang
- Department of Oral and Maxillofacial Surgery, University of Texas Health and Science Center at San Antonio, Texas, 78229
| | - Ruben Gomez
- Department of Oral and Maxillofacial Surgery, University of Texas Health and Science Center at San Antonio, Texas, 78229
| | - John Shannonhouse
- Department of Oral and Maxillofacial Surgery, University of Texas Health and Science Center at San Antonio, Texas, 78229
| | - Hyeonwi Son
- Department of Oral and Maxillofacial Surgery, University of Texas Health and Science Center at San Antonio, Texas, 78229
| | - Ratan Banik
- Department of Anesthesiology, School of Medicine, University of Minnesota, Minneapolis, Minnesota, 55455
| | - Yu Shin Kim
- Department of Oral and Maxillofacial Surgery, University of Texas Health and Science Center at San Antonio, Texas, 78229
- Programs in Integrated Biomedical Sciences, Translational Sciences, Biomedical Engineering, Radiological Sciences, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229
| |
Collapse
|
89
|
Hovhannisyan AH, Son H, Mecklenburg J, Barba-Escobedo PA, Tram M, Gomez R, Shannonhouse J, Zou Y, Weldon K, Ruparel S, Lai Z, Tumanov AV, Kim YS, Akopian AN. Pituitary hormones are specifically expressed in trigeminal sensory neurons and contribute to pain responses in the trigeminal system. Sci Rep 2021; 11:17813. [PMID: 34497285 PMCID: PMC8426369 DOI: 10.1038/s41598-021-97084-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Accepted: 08/20/2021] [Indexed: 11/29/2022] Open
Abstract
Trigeminal (TG), dorsal root (DRG), and nodose/jugular (NG/JG) ganglia each possess specialized and distinct functions. We used RNA sequencing of two-cycle sorted Pirt-positive neurons to identify genes exclusively expressing in L3-L5 DRG, T10-L1 DRG, NG/JG, and TG mouse ganglion neurons. Transcription factor Phox2b and Efcab6 are specifically expressed in NG/JG while Hoxa7 is exclusively present in both T10-L1 and L3-L5 DRG neurons. Cyp2f2, Krt18, and Ptgds, along with pituitary hormone prolactin (Prl), growth hormone (Gh), and proopiomelanocortin (Pomc) encoding genes are almost exclusively in TG neurons. Immunohistochemistry confirmed selective expression of these hormones in TG neurons and dural nerves; and showed GH expression in subsets of TRPV1+ and CGRP+ TG neurons. We next examined GH roles in hypersensitivity in the spinal versus trigeminal systems. Exogenous GH produced mechanical hypersensitivity when injected intrathecally, but not intraplantarly. GH-induced thermal hypersensitivity was not detected in the spinal system. GH dose-dependently generated orofacial and headache-like periorbital mechanical hypersensitivity after administration into masseter muscle and dura, respectively. Periorbital mechanical hypersensitivity was reversed by a GH receptor antagonist, pegvisomant. Overall, pituitary hormone genes are selective for TG versus other ganglia somatotypes; and GH has distinctive functional significance in the trigeminal versus spinal systems.
Collapse
Affiliation(s)
- Anahit H Hovhannisyan
- Departments of Endodontics, The School of Dentistry, The University of Texas Health Science Center at San Antonio (UTHSCSA), 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA
| | - Hyeonwi Son
- Departments of Oral and Maxillofacial Surgery, The School of Dentistry, The University of Texas Health Science Center at San Antonio (UTHSCSA), San Antonio, TX, 78229, USA
| | - Jennifer Mecklenburg
- Departments of Endodontics, The School of Dentistry, The University of Texas Health Science Center at San Antonio (UTHSCSA), 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA
| | - Priscilla Ann Barba-Escobedo
- Departments of Endodontics, The School of Dentistry, The University of Texas Health Science Center at San Antonio (UTHSCSA), 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA
| | - Meilinn Tram
- Departments of Endodontics, The School of Dentistry, The University of Texas Health Science Center at San Antonio (UTHSCSA), 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA
- Programs in Integrated Biomedical Sciences and Translational Sciences, The School of Medicine, UTHSCSA, San Antonio, TX, 78229, USA
| | - Ruben Gomez
- Departments of Oral and Maxillofacial Surgery, The School of Dentistry, The University of Texas Health Science Center at San Antonio (UTHSCSA), San Antonio, TX, 78229, USA
| | - John Shannonhouse
- Departments of Oral and Maxillofacial Surgery, The School of Dentistry, The University of Texas Health Science Center at San Antonio (UTHSCSA), San Antonio, TX, 78229, USA
| | - Yi Zou
- Greehey Children's Cancer Research Institute, UTHSCSA, San Antonio, TX, 78229, USA
| | - Korri Weldon
- Greehey Children's Cancer Research Institute, UTHSCSA, San Antonio, TX, 78229, USA
| | - Shivani Ruparel
- Departments of Endodontics, The School of Dentistry, The University of Texas Health Science Center at San Antonio (UTHSCSA), 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA
- Programs in Integrated Biomedical Sciences and Translational Sciences, The School of Medicine, UTHSCSA, San Antonio, TX, 78229, USA
| | - Zhao Lai
- Departments of Molecular Medicine, Programs in Integrated Biomedical Sciences and Translational Sciences, The School of Medicine, UTHSCSA, San Antonio, TX, 78229, USA
- Greehey Children's Cancer Research Institute, UTHSCSA, San Antonio, TX, 78229, USA
| | - Alexei V Tumanov
- Departments of Microbiology, Immunology and Molecular Genetics, Programs in Integrated Biomedical Sciences and Translational Sciences, The School of Medicine, UTHSCSA, San Antonio, TX, 78229, USA
- Programs in Integrated Biomedical Sciences and Translational Sciences, The School of Medicine, UTHSCSA, San Antonio, TX, 78229, USA
| | - Yu Shin Kim
- Departments of Oral and Maxillofacial Surgery, The School of Dentistry, The University of Texas Health Science Center at San Antonio (UTHSCSA), San Antonio, TX, 78229, USA
- Programs in Integrated Biomedical Sciences and Translational Sciences, The School of Medicine, UTHSCSA, San Antonio, TX, 78229, USA
| | - Armen N Akopian
- Departments of Endodontics, The School of Dentistry, The University of Texas Health Science Center at San Antonio (UTHSCSA), 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA.
- Programs in Integrated Biomedical Sciences and Translational Sciences, The School of Medicine, UTHSCSA, San Antonio, TX, 78229, USA.
| |
Collapse
|
90
|
Wang S, Chung MK. Orthodontic force induces nerve injury-like transcriptomic changes driven by TRPV1-expressing afferents in mouse trigeminal ganglia. Mol Pain 2021; 16:1744806920973141. [PMID: 33215551 PMCID: PMC7686596 DOI: 10.1177/1744806920973141] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Orthodontic force produces mechanical irritation and localized inflammation in
the periodontium, which causes pain in most patients. Nocifensive behaviors
resulting from orthodontic force in mice can be substantially attenuated by
intraganglionic injection of resiniferatoxin (RTX), a neurotoxin that
specifically ablates a subset of neurons expressing transient receptor potential
vanilloid 1 (TRPV1). In the current study, we determined changes in the
transcriptomic profiles in the trigeminal ganglia (TG) following the application
of orthodontic force, and assessed the roles of TRPV1-expressing afferents in
these transcriptomic changes. RTX or vehicle was injected into the TG of mice a
week before the placement of an orthodontic spring exerting 10 g of force. After
2 days, the TG were collected for RNA sequencing. The application of orthodontic
force resulted in 1279 differentially expressed genes (DEGs) in the TG. Gene
ontology analysis showed downregulation of gliogenesis and ion channel
activities, especially of voltage-gated potassium channels. DEGs produced by
orthodontic force correlated more strongly with DEGs resulting from nerve injury
than from inflammation. Orthodontic force resulted in the differential
expression of multiple genes involved in pain regulation, including upregulation
of Atf3, Adcyap1, Bdnf, and
Csf1, and downregulation of Scn10a,
Kcna2, Kcnj10, and P2ry1.
Orthodontic force-induced DEGs correlated with DEGs specific to multiple
neuronal and non-neuronal subtypes following nerve injury. These transcriptomic
changes were abolished in the mice that received the RTX injection. These
results suggest that orthodontic force produces transcriptomic changes
resembling nerve injury in the TG and that nociceptive inputs through
TRPV1-expressing afferents leads to subsequent changes in gene expression not
only in TRPV1-positive neurons, but also in TRPV1-negative neurons and
non-neuronal cells throughout the ganglia. Orthodontic force-induced
transcriptomic changes might be an active regenerative program of trigeminal
ganglia in response to axonal injury following orthodontic force.
Collapse
Affiliation(s)
- Sheng Wang
- Department of Neural and Pain Sciences, Center to Advance Chronic Pain Research, University of Maryland Dental School, Baltimore, MD, USA
| | - Man-Kyo Chung
- Department of Neural and Pain Sciences, Center to Advance Chronic Pain Research, University of Maryland Dental School, Baltimore, MD, USA
| |
Collapse
|
91
|
Tobori S, Hiyama H, Miyake T, Yano Y, Nagayasu K, Shirakawa H, Nakagawa T, Mori Y, Kaneko S. MrgprB4 in trigeminal neurons expressing TRPA1 modulates unpleasant sensations. J Pharmacol Sci 2021; 146:200-205. [PMID: 34116733 DOI: 10.1016/j.jphs.2021.04.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 04/18/2021] [Accepted: 04/22/2021] [Indexed: 11/25/2022] Open
Abstract
Gentle touch such as stroking of the skin produces a pleasant feeling, which is detected by a rare subset of sensory neurons that express Mas-related G protein-coupled receptor B4 (MrgprB4) in mice. We examined small populations of MrgprB4-positive neurons in the trigeminal ganglion and the dorsal root ganglion, and most of these were sensitive to transient receptor potential ankyrin 1 (TRPA1) agonist but not TRPV1, TRPM8, or TRPV4 agonists. Deficiency of MrgprB4 did not affect noxious pain or itch behaviors in the hairless plantar and hairy cheek. Although behavior related to acetone-induced cold sensing in the hind paw was not changed, unpleasant sensory behaviors in response to acetone application or sucrose splash to the cheek were significantly enhanced in Mrgprb4-knockout mice as well as in TRPA1-knockout mice. These results suggest that MrgprB4 in the trigeminal neurons produces pleasant sensations in cooperation with TRPA1, rather than noxious or cold sensations. Pleasant sensations may modulate unpleasant sensations on the cheek via MrgprB4.
Collapse
Affiliation(s)
- Shota Tobori
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Haruka Hiyama
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Takahito Miyake
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan; Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Yuichi Yano
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Kazuki Nagayasu
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Hisashi Shirakawa
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan.
| | - Takayuki Nakagawa
- Department of Clinical Pharmacology and Therapeutics, Kyoto University Hospital, 54 Shogoin -Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Yasuo Mori
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Building A4, Katsura Campus, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Shuji Kaneko
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| |
Collapse
|
92
|
Chen Y, Wang ZL, Yeo M, Zhang QJ, López-Romero AE, Ding HP, Zhang X, Zeng Q, Morales-Lázaro SL, Moore C, Jin YA, Yang HH, Morstein J, Bortsov A, Krawczyk M, Lammert F, Abdelmalek M, Diehl AM, Milkiewicz P, Kremer AE, Zhang JY, Nackley A, Reeves TE, Ko MC, Ji RR, Rosenbaum T, Liedtke W. Epithelia-Sensory Neuron Cross Talk Underlies Cholestatic Itch Induced by Lysophosphatidylcholine. Gastroenterology 2021; 161:301-317.e16. [PMID: 33819485 PMCID: PMC9093619 DOI: 10.1053/j.gastro.2021.03.049] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 03/16/2021] [Accepted: 03/22/2021] [Indexed: 01/16/2023]
Abstract
BACKGROUND & AIMS Limited understanding of pruritus mechanisms in cholestatic liver diseases hinders development of antipruritic treatments. Previous studies implicated lysophosphatidic acid (LPA) as a potential mediator of cholestatic pruritus. METHODS Pruritogenicity of lysophosphatidylcholine (LPC), LPA's precursor, was examined in naïve mice, cholestatic mice, and nonhuman primates. LPC's pruritogenicity involving keratinocyte TRPV4 was studied using genetic and pharmacologic approaches, cultured keratinocytes, ion channel physiology, and structural computational modeling. Activation of pruriceptor sensory neurons by microRNA-146a (miR-146a), secreted from keratinocytes, was identified by in vitro and ex vivo Ca2+ imaging assays. Sera from patients with primary biliary cholangitis were used for measuring the levels of LPC and miR-146a. RESULTS LPC was robustly pruritic in mice. TRPV4 in skin keratinocytes was essential for LPC-induced itch and itch in mice with cholestasis. Three-dimensional structural modeling, site-directed mutagenesis, and channel function analysis suggested a TRPV4 C-terminal motif for LPC binding and channel activation. In keratinocytes, TRPV4 activation by LPC induced extracellular release of miR-146a, which activated TRPV1+ sensory neurons to cause itch. LPC and miR-146a levels were both elevated in sera of patients with primary biliary cholangitis with itch and correlated with itch intensity. Moreover, LPC and miR-146a were also increased in sera of cholestatic mice and elicited itch in nonhuman primates. CONCLUSIONS We identified LPC as a novel cholestatic pruritogen that induces itch through epithelia-sensory neuron cross talk, whereby it directly activates skin keratinocyte TRPV4, which rapidly releases miR-146a to activate skin-innervating TRPV1+ pruriceptor sensory neurons. Our findings support the new concept of the skin, as a sensory organ, playing a critical role in cholestatic itch, beyond liver, peripheral sensory neurons, and central neural pathways supporting pruriception.
Collapse
Affiliation(s)
- Yong Chen
- Department of Neurology, Duke University, Durham, North Carolina.
| | - Zi-Long Wang
- Department of Neurology, Duke University, Durham, North Carolina; Center for Translational Pain Medicine, Department of Anesthesiology, Duke University, Durham, North Carolina
| | - Michele Yeo
- Department of Neurology, Duke University, Durham, North Carolina
| | - Qiao-Juan Zhang
- Department of Neurology, Duke University, Durham, North Carolina
| | - Ana E López-Romero
- Departamento de Neurociencia Cognitiva, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Coyoacan, Mexico City, Mexico
| | - Hui-Ping Ding
- Department of Physiology and Pharmacology, Wake Forest University, Winston-Salem, North Carolina
| | - Xin Zhang
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University, Durham, North Carolina
| | - Qian Zeng
- Department of Neurology, Duke University, Durham, North Carolina
| | - Sara L Morales-Lázaro
- Departamento de Neurociencia Cognitiva, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Coyoacan, Mexico City, Mexico
| | - Carlene Moore
- Department of Neurology, Duke University, Durham, North Carolina
| | - Ying-Ai Jin
- Department of Dermatology, Duke University, Durham, North Carolina
| | - Huang-He Yang
- Department of Biochemistry, Duke University, Durham, North Carolina; Department of Neurobiology, Duke University, Durham, North Carolina
| | | | - Andrey Bortsov
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University, Durham, North Carolina
| | - Marcin Krawczyk
- Department of Medicine II, Saarland University Medical Center, Saarland University, Homburg, Germany; Laboratory of Metabolic Liver Diseases, Center for Preclinical Research, Department of General, Transplant and Liver Surgery, Medical University of Warsaw, Warsaw, Poland
| | - Frank Lammert
- Department of Medicine II, Saarland University Medical Center, Saarland University, Homburg, Germany; Hannover Medical School MHH, Hannover, Germany
| | - Manal Abdelmalek
- Division of Gastroenterology, Department of Medicine, Duke University, Durham, North Carolina
| | - Anna Mae Diehl
- Division of Gastroenterology, Department of Medicine, Duke University, Durham, North Carolina
| | - Piotr Milkiewicz
- Liver and Internal Medicine Unit, Department of General, Transplant and Liver Surgery, Medical University of Warsaw, Warsaw, Poland; Translation Medicine Group, Pomeranian Medical University, Szczecin, Poland
| | - Andreas E Kremer
- Department of Medicine 1, Gastroenterology, Hepatology, Pneumology and Endocrinology, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Jennifer Y Zhang
- Department of Dermatology, Duke University, Durham, North Carolina; Department of Pathology, Duke University, Durham, North Carolina
| | - Andrea Nackley
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University, Durham, North Carolina; Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina
| | - Tony E Reeves
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Mei-Chuan Ko
- Department of Physiology and Pharmacology, Wake Forest University, Winston-Salem, North Carolina
| | - Ru-Rong Ji
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University, Durham, North Carolina; Department of Neurobiology, Duke University, Durham, North Carolina
| | - Tamara Rosenbaum
- Departamento de Neurociencia Cognitiva, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Coyoacan, Mexico City, Mexico
| | - Wolfgang Liedtke
- Department of Neurology, Duke University, Durham, North Carolina; Center for Translational Pain Medicine, Department of Anesthesiology, Duke University, Durham, North Carolina; Department of Neurobiology, Duke University, Durham, North Carolina; Neurology Clinics for Headache, Head-Pain and Trigeminal Sensory Disorders, Duke University, Durham, North Carolina; Clinics for Innovative Pain Therapy, Department of Anesthesiology, Duke University, Raleigh, North Carolina.
| |
Collapse
|
93
|
17β-Estradiol Exacerbated Experimental Occlusal Interference-Induced Chronic Masseter Hyperalgesia by Increasing the Neuronal Excitability and TRPV1 Function of Trigeminal Ganglion in Ovariectomized Rats. Int J Mol Sci 2021; 22:ijms22136945. [PMID: 34203300 PMCID: PMC8269106 DOI: 10.3390/ijms22136945] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/14/2021] [Accepted: 06/20/2021] [Indexed: 12/17/2022] Open
Abstract
Pain symptoms in temporomandibular disorders (TMD) predominantly affect reproductive women, suggesting that estrogen regulates pain perception. However, how estrogen contributes to chronic TMD pain remains largely unclear. In the present study, we performed behavioral tests, electrophysiology, Western blot and immunofluorescence to investigate the role and underlying mechanisms of estrogen in dental experimental occlusal interference (EOI)-induced chronic masseter mechanical hyperalgesia in rats. We found that long-term 17β-estradiol (E2) replacement exacerbated EOI-induced masseter hyperalgesia in a dose-dependent manner in ovariectomized (OVX) rats. Whole-cell patch-clamp recordings demonstrated that E2 (100 nM) treatment enhanced the excitability of isolated trigeminal ganglion (TG) neurons in OVX and OVX EOI rats, and EOI increased the functional expression of transient receptor potential vanilloid-1 (TRPV1). In addition, E2 replacement upregulated the protein expression of TRPV1 in EOI-treated OVX rats. Importantly, intraganglionic administration of the TRPV1 antagonist AMG-9810 strongly attenuated the facilitatory effect of E2 on EOI-induced masseter mechanical sensitivity. These results demonstrate that E2 exacerbated EOI-induced chronic masseter mechanical hyperalgesia by increasing TG neuronal excitability and TRPV1 function. Our study helps to elucidate the E2 actions in chronic myogenic TMD pain and may provide new therapeutic targets for relieving estrogen-sensitive pain.
Collapse
|
94
|
Huang Z, Tatti R, Loeven AM, Landi Conde DR, Fadool DA. Modulation of Neural Microcircuits That Control Complex Dynamics in Olfactory Networks. Front Cell Neurosci 2021; 15:662184. [PMID: 34239417 PMCID: PMC8259627 DOI: 10.3389/fncel.2021.662184] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 05/10/2021] [Indexed: 11/13/2022] Open
Abstract
Neuromodulation influences neuronal processing, conferring neuronal circuits the flexibility to integrate sensory inputs with behavioral states and the ability to adapt to a continuously changing environment. In this original research report, we broadly discuss the basis of neuromodulation that is known to regulate intrinsic firing activity, synaptic communication, and voltage-dependent channels in the olfactory bulb. Because the olfactory system is positioned to integrate sensory inputs with information regarding the internal chemical and behavioral state of an animal, how olfactory information is modulated provides flexibility in coding and behavioral output. Herein we discuss how neuronal microcircuits control complex dynamics of the olfactory networks by homing in on a special class of local interneurons as an example. While receptors for neuromodulation and metabolic peptides are widely expressed in the olfactory circuitry, centrifugal serotonergic and cholinergic inputs modulate glomerular activity and are involved in odor investigation and odor-dependent learning. Little is known about how metabolic peptides and neuromodulators control specific neuronal subpopulations. There is a microcircuit between mitral cells and interneurons that is comprised of deep-short-axon cells in the granule cell layer. These local interneurons express pre-pro-glucagon (PPG) and regulate mitral cell activity, but it is unknown what initiates this type of regulation. Our study investigates the means by which PPG neurons could be recruited by classical neuromodulators and hormonal peptides. We found that two gut hormones, leptin and cholecystokinin, differentially modulate PPG neurons. Cholecystokinin reduces or increases spike frequency, suggesting a heterogeneous signaling pathway in different PPG neurons, while leptin does not affect PPG neuronal firing. Acetylcholine modulates PPG neurons by increasing the spike frequency and eliciting bursts of action potentials, while serotonin does not affect PPG neuron excitability. The mechanisms behind this diverse modulation are not known, however, these results clearly indicate a complex interplay of metabolic signaling molecules and neuromodulators that may fine-tune neuronal microcircuits.
Collapse
Affiliation(s)
- Zhenbo Huang
- Program in Neuroscience, Florida State University, Tallahassee, FL, United States
| | - Roberta Tatti
- Program in Neuroscience, Florida State University, Tallahassee, FL, United States
| | - Ashley M Loeven
- Cell and Molecular Biology Program, Department of Biological Science, Florida State University, Tallahassee, FL, United States
| | - Daniel R Landi Conde
- Program in Neuroscience, Florida State University, Tallahassee, FL, United States
| | - Debra Ann Fadool
- Program in Neuroscience, Florida State University, Tallahassee, FL, United States.,Cell and Molecular Biology Program, Department of Biological Science, Florida State University, Tallahassee, FL, United States.,Institute of Molecular Biophysics, Florida State University, Tallahassee, FL, United States
| |
Collapse
|
95
|
Acute and Chronic Pain from Facial Skin and Oral Mucosa: Unique Neurobiology and Challenging Treatment. Int J Mol Sci 2021; 22:ijms22115810. [PMID: 34071720 PMCID: PMC8198570 DOI: 10.3390/ijms22115810] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/18/2021] [Accepted: 05/20/2021] [Indexed: 12/14/2022] Open
Abstract
The oral cavity is a portal into the digestive system, which exhibits unique sensory properties. Like facial skin, the oral mucosa needs to be exquisitely sensitive and selective, in order to detect harmful toxins versus edible food. Chemosensation and somatosensation by multiple receptors, including transient receptor potential channels, are well-developed to meet these needs. In contrast to facial skin, however, the oral mucosa rarely exhibits itch responses. Like the gut, the oral cavity performs mechanical and chemical digestion. Therefore, the oral mucosa needs to be insensitive, to some degree, in order to endure noxious irritation. Persistent pain from the oral mucosa is often due to ulcers, involving both tissue injury and infection. Trigeminal nerve injury and trigeminal neuralgia produce intractable pain in the orofacial skin and the oral mucosa, through mechanisms distinct from those seen in the spinal area, which is particularly difficult to predict or treat. The diagnosis and treatment of idiopathic chronic pain, such as atypical odontalgia (idiopathic painful trigeminal neuropathy or post-traumatic trigeminal neuropathy) and burning mouth syndrome, remain especially challenging. The central integration of gustatory inputs might modulate chronic oral and facial pain. A lack of pain in chronic inflammation inside the oral cavity, such as chronic periodontitis, involves the specialized functioning of oral bacteria. A more detailed understanding of the unique neurobiology of pain from the orofacial skin and the oral mucosa should help us develop novel methods for better treating persistent orofacial pain.
Collapse
|
96
|
Calcium imaging in population of dorsal root ganglion neurons unravels novel mechanisms of visceral pain sensitization and referred somatic hypersensitivity. Pain 2021; 162:1068-1081. [PMID: 33021564 DOI: 10.1097/j.pain.0000000000002096] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 09/23/2020] [Indexed: 12/25/2022]
Abstract
ABSTRACT Mechanisms of visceral pain sensitization and referred somatic hypersensitivity remain unclear. We conducted calcium imaging in Pirt-GCaMP6s mice to gauge responses of dorsal root ganglion (DRG) neurons to visceral and somatic stimulation in vivo. Intracolonic instillation of 2,4,6-trinitrobenzene sulfonic acid (TNBS) induced colonic inflammation and increased the percentage of L6 DRG neurons that responded to colorectal distension above that of controls at day 7. Colorectal distension did not activate L4 DRG neurons. TNBS-treated mice exhibited more Evans blue extravasation than did control mice and developed mechanical hypersensitivity in low-back skin and hind paws, which are innervated by L6 and L4 DRG neurons, respectively, suggesting that colonic inflammation induced mechanical hypersensitivity in both homosegmental and heterosegmental somatic regions. Importantly, the percentage of L4 DRG neurons activated by hind paw pinch and brush stimulation and calcium responses of L6 DRG neurons to low-back brush stimulation were higher at day 7 after TNBS than those in control mice. Visceral irritation from intracolonic capsaicin instillation also increased Evans blue extravasation in hind paws and low-back skin and acutely increased the percentage of L4 DRG neurons responding to hind paw pinch and the response of L6 DRG neurons to low-back brush stimulation. These findings suggest that TNBS-induced colitis and capsaicin-induced visceral irritation may sensitize L6 DRG neurons to colorectal and somatic inputs and also increase the excitability of L4 DRG neurons that do not receive colorectal inputs. These changes may represent a potential peripheral neuronal mechanism for visceral pain sensitization and referred somatic hypersensitivity.
Collapse
|
97
|
MacDonald DI, Sikandar S, Weiss J, Pyrski M, Luiz AP, Millet Q, Emery EC, Mancini F, Iannetti GD, Alles SRA, Arcangeletti M, Zhao J, Cox JJ, Brownstone RM, Zufall F, Wood JN. A central mechanism of analgesia in mice and humans lacking the sodium channel Na V1.7. Neuron 2021; 109:1497-1512.e6. [PMID: 33823138 PMCID: PMC8110947 DOI: 10.1016/j.neuron.2021.03.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 02/05/2020] [Accepted: 03/08/2021] [Indexed: 11/18/2022]
Abstract
Deletion of SCN9A encoding the voltage-gated sodium channel NaV1.7 in humans leads to profound pain insensitivity and anosmia. Conditional deletion of NaV1.7 in sensory neurons of mice also abolishes pain, suggesting that the locus of analgesia is the nociceptor. Here we demonstrate, using in vivo calcium imaging and extracellular recording, that NaV1.7 knockout mice have essentially normal nociceptor activity. However, synaptic transmission from nociceptor central terminals in the spinal cord is greatly reduced by an opioid-dependent mechanism. Analgesia is also reversed substantially by central but not peripheral application of opioid antagonists. In contrast, the lack of neurotransmitter release from olfactory sensory neurons is opioid independent. Male and female humans with NaV1.7-null mutations show naloxone-reversible analgesia. Thus, inhibition of neurotransmitter release is the principal mechanism of anosmia and analgesia in mouse and human Nav1.7-null mutants.
Collapse
Affiliation(s)
- Donald Iain MacDonald
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, Gower Street, London WC1E 6BT, UK.
| | - Shafaq Sikandar
- Centre for Experimental Medicine & Rheumatology, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Jan Weiss
- Center for Integrative Physiology and Molecular Medicine, Saarland University, 66421 Homburg, Germany
| | - Martina Pyrski
- Center for Integrative Physiology and Molecular Medicine, Saarland University, 66421 Homburg, Germany
| | - Ana P Luiz
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, Gower Street, London WC1E 6BT, UK
| | - Queensta Millet
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, Gower Street, London WC1E 6BT, UK
| | - Edward C Emery
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, Gower Street, London WC1E 6BT, UK
| | - Flavia Mancini
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London WC1E 6BT, UK
| | - Gian D Iannetti
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London WC1E 6BT, UK; Neuroscience and Behaviour Laboratory, Istituto Italiano di Tecnologia, Rome, Italy
| | - Sascha R A Alles
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, Gower Street, London WC1E 6BT, UK
| | - Manuel Arcangeletti
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, Gower Street, London WC1E 6BT, UK
| | - Jing Zhao
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, Gower Street, London WC1E 6BT, UK
| | - James J Cox
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, Gower Street, London WC1E 6BT, UK
| | | | - Frank Zufall
- Center for Integrative Physiology and Molecular Medicine, Saarland University, 66421 Homburg, Germany
| | - John N Wood
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, Gower Street, London WC1E 6BT, UK.
| |
Collapse
|
98
|
Xing Y, Steele HR, Hilley HB, Zhu Y, Lawson K, Niehoff T, Han L. Visualizing the Itch-Sensing Skin Arbors. J Invest Dermatol 2021; 141:1308-1316. [PMID: 33091423 PMCID: PMC8055730 DOI: 10.1016/j.jid.2020.08.030] [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: 06/19/2020] [Revised: 08/10/2020] [Accepted: 08/26/2020] [Indexed: 12/15/2022]
Abstract
Diverse sensory neurons exhibit distinct neuronal morphologies with a variety of axon terminal arborizations subserving their functions. Because of its clinical significance, the molecular and cellular mechanisms of itch are being intensely studied. However, a complete analysis of itch-sensing terminal arborization is missing. Using an MrgprC11CreERT2 transgenic mouse line, we labeled a small subset of itch-sensing neurons that express multiple itch-related molecules including MrgprA3, MrgprC11, histamine receptor H1, IL-31 receptor, 5-hydroxytryptamine receptor 1F, natriuretic precursor peptide B, and neuromedin B. By combining sparse genetic labeling and whole-mount placental alkaline phosphatase histochemistry, we found that itch-sensing skin arbors exhibit free endings with extensive axonal branching in the superficial epidermis and large receptive fields. These results revealed the unique morphological characteristics of itch-sensing neurons and provide intriguing insights into the basic mechanisms of itch transmission.
Collapse
Affiliation(s)
- Yanyan Xing
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Haley R Steele
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Henry B Hilley
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Yuyan Zhu
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Katy Lawson
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Taylor Niehoff
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Liang Han
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia, USA.
| |
Collapse
|
99
|
Abstract
Itch arising from glabrous skin (palms and soles) has attracted limited attention within the field due to the lack of methodology. This is despite glabrous itch arising from many medical conditions such as plantar and palmar psoriasis, dyshidrosis, and cholestasis. Therefore, we developed a mouse glabrous skin behavioral assay to investigate the contribution of three previously identified pruriceptive neurons in glabrous skin itch. Our results show that MrgprA3+ and MrgprD+ neurons, although key mediators for hairy skin itch, do not play important roles in glabrous skin itch, demonstrating a mechanistic difference in itch sensation between hairy and glabrous skin. We found that MrgprC11+ neurons are the major mediators for glabrous skin itch. Activation of MrgprC11+ neurons induced glabrous skin itch, while ablation of MrgprC11+ neurons reduced both acute and chronic glabrous skin itch. Our study provides insights into the mechanisms of itch and opens up new avenues for future glabrous skin itch research.
Collapse
|
100
|
Ishihara S, Obeidat AM, Wokosin DL, Ren D, Miller RJ, Malfait AM, Miller RE. The role of intra-articular neuronal CCR2 receptors in knee joint pain associated with experimental osteoarthritis in mice. Arthritis Res Ther 2021; 23:103. [PMID: 33827672 PMCID: PMC8025346 DOI: 10.1186/s13075-021-02486-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 03/22/2021] [Indexed: 12/12/2022] Open
Abstract
Background C–C chemokine receptor 2 (CCR2) signaling plays a key role in pain associated with experimental murine osteoarthritis (OA) after destabilization of the medial meniscus (DMM). Here, we aimed to assess if CCR2 expressed by intra-articular sensory neurons contributes to knee hyperalgesia in the early stages of the model. Methods DMM surgery was performed in the right knee of 10-week-old male wild-type (WT), Ccr2 null, or Ccr2RFP C57BL/6 mice. Knee hyperalgesia was measured using a Pressure Application Measurement device. CCR2 receptor antagonist (CCR2RA) was injected systemically (i.p.) or intra-articularly (i.a.) at different times after DMM to test its ability to reverse knee hyperalgesia. In vivo Ca2+ imaging of the dorsal root ganglion (DRG) was performed to assess sensory neuron responses to CCL2 injected into the knee joint cavity. CCL2 protein in the knee was measured by ELISA. Ccr2RFP mice and immunohistochemical staining for the pan-neuronal marker, protein gene product 9.5 (PGP9.5), or the sensory neuron marker, calcitonin gene-related peptide (CGRP), were used to visualize the location of CCR2 on intra-articular afferents. Results WT, but not Ccr2 null, mice displayed knee hyperalgesia 2–16 weeks after DMM. CCR2RA administered i.p. alleviated established hyperalgesia in WT mice 4 and 8 weeks after surgery. Intra-articular injection of CCL2 excited sensory neurons in the L4-DRG, as determined by in vivo calcium imaging; responses to CCL2 increased in mice 20 weeks after DMM. CCL2, but not vehicle, injected i.a. rapidly caused transient knee hyperalgesia in naïve WT, but not Ccr2 null, mice. Intra-articular CCR2RA injection also alleviated established hyperalgesia in WT mice 4 and 7 weeks after surgery. CCL2 protein was elevated in the knees of both WT and Ccr2 null mice 4 weeks after surgery. Co-expression of CCR2 and PGP9.5 as well as CCR2 and CGRP was observed in the lateral synovium of naïve mice; co-expression was also observed in the medial compartment of knees 8 weeks after DMM. Conclusions The findings suggest that CCL2-CCR2 signaling locally in the joint contributes to knee hyperalgesia in experimental OA, and it is in part mediated through direct stimulation of CCR2 expressed by intra-articular sensory afferents. Supplementary Information The online version contains supplementary material available at 10.1186/s13075-021-02486-y.
Collapse
Affiliation(s)
- Shingo Ishihara
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, 1735 W Harrison St, Room 714, Chicago, IL, 60612, USA
| | - Alia M Obeidat
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, 1735 W Harrison St, Room 714, Chicago, IL, 60612, USA
| | - David L Wokosin
- Department of Physiology, Northwestern University, Chicago, IL, 60611, USA
| | - Dongjun Ren
- Department of Pharmacology, Northwestern University, Chicago, IL, 60611, USA
| | - Richard J Miller
- Department of Pharmacology, Northwestern University, Chicago, IL, 60611, USA
| | - Anne-Marie Malfait
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, 1735 W Harrison St, Room 714, Chicago, IL, 60612, USA
| | - Rachel E Miller
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, 1735 W Harrison St, Room 714, Chicago, IL, 60612, USA.
| |
Collapse
|