1
|
Van Remoortel S, Hussein H, Boeckxstaens G. Mast cell modulation: A novel therapeutic strategy for abdominal pain in irritable bowel syndrome. Cell Rep Med 2024; 5:101780. [PMID: 39378882 PMCID: PMC11513802 DOI: 10.1016/j.xcrm.2024.101780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 07/10/2024] [Accepted: 09/17/2024] [Indexed: 10/10/2024]
Abstract
Irritable bowel syndrome (IBS) is one of the most prevalent gastrointestinal disorders characterized by recurrent abdominal pain and an altered defecation pattern. Chronic abdominal pain represents the hallmark IBS symptom and is reported to have the most bothersome impact on the patient's quality of life. Unfortunately, effective therapeutic strategies reducing abdominal pain are lacking, mainly attributed to a limited understanding of the contributing mechanisms. In the past few years, exciting new insights have pointed out that altered communication between gut immune cells and pain-sensing nerves acts as a hallmark driver of IBS-related abdominal pain. In this review, we aim to summarize our current knowledge on altered neuro-immune crosstalk as the main driver of altered pain signaling, with a specific focus on altered mast cell functioning herein, and highlight the relevance of targeting mast cell-mediated mechanisms as a novel therapeutic strategy for chronic abdominal pain in IBS patients.
Collapse
Affiliation(s)
- Samuel Van Remoortel
- Translational Research Centre for Gastrointestinal Disorders, KU Leuven, Leuven, Belgium
| | - Hind Hussein
- Translational Research Centre for Gastrointestinal Disorders, KU Leuven, Leuven, Belgium
| | - Guy Boeckxstaens
- Translational Research Centre for Gastrointestinal Disorders, KU Leuven, Leuven, Belgium.
| |
Collapse
|
2
|
Hussein H, Van Remoortel S, Boeckxstaens GE. Irritable bowel syndrome: When food is a pain in the gut. Immunol Rev 2024; 326:102-116. [PMID: 39037230 DOI: 10.1111/imr.13374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/23/2024]
Abstract
Irritable bowel syndrome (IBS) is a chronic gastrointestinal condition associated with altered bowel habits and recurrent abdominal pain, often triggered by food intake. Current treatments focus on improving stool pattern, but effective treatments for pain in IBS are still lacking due to our limited understanding of pathophysiological mechanisms. Visceral hypersensitivity (VHS), or abnormal visceral pain perception, underlies abdominal pain development in IBS, and mast cell activation has been shown to play an important role in the development of VHS. Our work recently revealed that abdominal pain in response to food intake is induced by the sensitization of colonic pain-sensing neurons by histamine produced by activated mast cells following a local IgE response to food. In this review, we summarize the current knowledge on abdominal pain and VHS pathophysiology in IBS, we outline the work leading to the discovery of the role of histamine in abdominal pain, and we introduce antihistamines as a novel treatment option to manage chronic abdominal pain in patients with IBS.
Collapse
Affiliation(s)
- Hind Hussein
- Center for Intestinal Neuro-Immune Interactions, Translational Research in Gastrointestinal Disorders (TARGID), Department of Chronic Diseases, Metabolism, and Ageing (CHROMETA), KU Leuven, Leuven, Belgium
| | - Samuel Van Remoortel
- Center for Intestinal Neuro-Immune Interactions, Translational Research in Gastrointestinal Disorders (TARGID), Department of Chronic Diseases, Metabolism, and Ageing (CHROMETA), KU Leuven, Leuven, Belgium
| | - Guy E Boeckxstaens
- Center for Intestinal Neuro-Immune Interactions, Translational Research in Gastrointestinal Disorders (TARGID), Department of Chronic Diseases, Metabolism, and Ageing (CHROMETA), KU Leuven, Leuven, Belgium
| |
Collapse
|
3
|
Li L, Yao H, Mo R, Xu L, Chen P, Chen Y, Hu JJ, Xie W, Song XJ. Blocking proteinase-activated receptor 2 signaling relieves pain, suppresses nerve sprouting, improves tissue repair, and enhances analgesic effect of B vitamins in rats with Achilles tendon injury. Pain 2024; 165:2055-2067. [PMID: 38598349 DOI: 10.1097/j.pain.0000000000003229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Accepted: 02/02/2024] [Indexed: 04/12/2024]
Abstract
ABSTRACT Tendon injury produces intractable pain and disability in movement, but the medications for analgesia and restoring functional integrity of tendon are still limited. In this study, we report that proteinase-activated receptor 2 (PAR2) activation in dorsal root ganglion (DRG) neurons contributes to chronic pain and tendon histopathological changes produced by Achilles tendon partial transection injury (TTI). Tendon partial transection injury increases the expression of PAR2 protein in both somata of DRG neurons and their peripheral terminals within the injured Achilles tendon. Activation of PAR2 promotes the primary sensory neuron plasticity by activating downstream cAMP-PKA pathway, phosphorylation of PKC, CaMKII, and CREB. Blocking PAR2 signaling by PAR2 small-interference RNA or antagonistic peptide PIP delays the onset of TTI-induced pain, reverses the ongoing pain, as well as inhibits sensory nerve sprouting, and promotes structural remodeling of the injured tendon. Vitamin B complex (VBC), containing thiamine (B1), pyridoxine (B6), and cyanocobalamin (B12), is effective to ameliorate TTI-induced pain, inhibit ectopic nerve sprouting, and accelerate tendon repair, through suppressing PAR2 activation. These findings reveal a critical role of PAR2 signaling in the development of chronic pain and histopathological alterations of injured tendon following Achilles tendon injury. This study suggests that the pharmaceuticals targeting PAR2, such as VBC, may be an effective approach for the treatment of tendon injury-induced pain and promoting tendon repair.
Collapse
Affiliation(s)
- Lihui Li
- Department of Medical Neuroscience, School of Medicine, Southern University of Science and Technology, Shenzhen, China
- SUSTech Center for Pain Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Hongyu Yao
- Department of Medical Neuroscience, School of Medicine, Southern University of Science and Technology, Shenzhen, China
- SUSTech Center for Pain Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Rufan Mo
- SUSTech Center for Pain Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Lihong Xu
- Department of Medical Neuroscience, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Peng Chen
- Department of Medical Neuroscience, School of Medicine, Southern University of Science and Technology, Shenzhen, China
- SUSTech Center for Pain Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Yuchen Chen
- Department of Medical Neuroscience, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Jiang-Jian Hu
- Department of Medical Neuroscience, School of Medicine, Southern University of Science and Technology, Shenzhen, China
- SUSTech Center for Pain Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Wei Xie
- The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, The School of Life Science and Technology, Southeast University, Nanjing, China
| | - Xue-Jun Song
- Department of Medical Neuroscience, School of Medicine, Southern University of Science and Technology, Shenzhen, China
- SUSTech Center for Pain Medicine, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
4
|
Zheng H, Kim M, Kim C, Kim Y, Cho PS, Lim JY, Lee H, Yun HI, Choi J, Hwang SW. GnRH peripherally modulates nociceptor functions, exacerbating mechanical pain. Front Mol Neurosci 2024; 17:1160435. [PMID: 38783903 PMCID: PMC11111891 DOI: 10.3389/fnmol.2024.1160435] [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: 02/07/2023] [Accepted: 04/22/2024] [Indexed: 05/25/2024] Open
Abstract
The function of peripheral nociceptors, the neurons that relay pain signals to the brain, are frequently tuned by local and systemic modulator substances. In this context, neurohormonal effects are emerging as an important modulatory mechanism, but many aspects remain to be elucidated. Here we report that gonadotropin-releasing hormone (GnRH), a brain-specific neurohormone, can aggravate pain by acting on nociceptors in mice. GnRH and GnRHR, the receptor for GnRH, are expressed in a nociceptor subpopulation. Administration of GnRH and its analogue, localized for selectively affecting the peripheral neurons, deteriorated mechanical pain, which was reproducible in neuropathic conditions. Nociceptor function was promoted by GnRH treatment in vitro, which appears to involve specific sensory transient receptor potential ion channels. These data suggest that peripheral GnRH can positively modulate nociceptor activities in its receptor-specific manner, contributing to pain exacerbation. Our study indicates that GnRH plays an important role in neurohormonal pain modulation via a peripheral mechanism.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Sun Wook Hwang
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
5
|
Ying LN, Sun Y, Cui LY, Zhang ZY, Li RF, Zhang J. Recent advances in the knowledge of the mechanism of reflux hypersensitivity. Scand J Gastroenterol 2024; 59:518-523. [PMID: 38343278 DOI: 10.1080/00365521.2024.2310177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 01/20/2024] [Indexed: 04/26/2024]
Abstract
Reflux hypersensitivity (RH) is a subtype of gastroesophageal reflux disease. The Rome IV criteria separated RH from the original nonerosive reflux disease subgroup and classified it as a new functional oesophageal disease. Recently, the pathogenesis of RH has become the focus of research. According to the latest research reports, upregulation of acid-sensitive receptors, distribution of calcitonin gene-related peptide-positive nerve fibres, and psychiatric comorbidity have key roles in the pathogenesis of RH. This work reviews the latest findings regarding RH mechanisms.
Collapse
Affiliation(s)
- Li Na Ying
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, P.R. China
| | - Yan Sun
- Department of Gastroenterology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, P.R. China
| | - Li Yang Cui
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, P.R. China
| | - Zhen Yu Zhang
- Graduate School of Bengbu Medical University, Bengbu, Anhui, P.R. China
| | - Rui Fang Li
- People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, P.R. China
| | - Jun Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, P.R. China
| |
Collapse
|
6
|
Achanta S, Chintagari NR, Balakrishna S, Liu B, Jordt SE. Pharmacologic Inhibition of Transient Receptor Potential Ion Channel Ankyrin 1 Counteracts 2-Chlorobenzalmalononitrile Tear Gas Agent-Induced Cutaneous Injuries. J Pharmacol Exp Ther 2024; 388:613-623. [PMID: 38050077 PMCID: PMC10801748 DOI: 10.1124/jpet.123.001666] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 10/27/2023] [Accepted: 10/31/2023] [Indexed: 12/06/2023] Open
Abstract
Deployment of the tear gas agent 2-chlorobenzalmalononitrile (CS) for riot control has significantly increased in recent years. The effects of CS have been believed to be transient and benign. However, CS induces severe pain, blepharospasm, lachrymation, airway obstruction, and skin blisters. Frequent injuries and hospitalizations have been reported after exposure. We have identified the sensory neuronal ion channel, transient receptor potential ankyrin 1 (TRPA1), as a key CS target resulting in acute irritation and pain and also as a mediator of neurogenic inflammation. Here, we examined the effects of pharmacologic TRPA1 inhibition on CS-induced cutaneous injury. We modeled CS-induced cutaneous injury by applying 10 μl CS agent [200 mM in dimethyl sulfoxide (DMSO)] to each side of the right ears of 8- to 9-week-old C57BL/6 male mice, whereas left ears were applied with solvent only (DMSO). The TRPA1 inhibitor HC-030031 or A-967079 was administered after CS exposure. CS exposure induced strong tissue swelling, plasma extravasation, and a dramatic increase in inflammatory cytokine levels in the mouse ear skin. We also showed that the effects of CS were not transient but caused persistent skin injuries. These injury parameters were reduced with TRPA1 inhibitor treatment. Further, we tested the pharmacologic activity of advanced TRPA1 antagonists in vitro. Our findings showed that TRPA1 is a crucial mediator of CS-induced nociception and tissue injury and that TRPA1 inhibitors are effective countermeasures that reduce key injury parameters when administered after exposure. Additional therapeutic efficacy studies with advanced TRPA1 antagonists and decontamination strategies are warranted. SIGNIFICANCE STATEMENT: 2-Chlorobenzalmalononitrile (CS) tear gas agent has been deployed as a crowd dispersion chemical agent in recent times. Exposure to CS tear gas agents has been believed to cause transient acute toxic effects that are minimal at most. Here we found that CS tear gas exposure causes both acute and persistent skin injuries and that treatment with transient receptor potential ion channel ankyrin 1 (TRPA1) antagonists ameliorated skin injuries.
Collapse
Affiliation(s)
- Satyanarayana Achanta
- Center for Translational Pain Medicine, Department of Anesthesiology (S.A., B.L., S.-E.J.) and Department of Pharmacology and Cancer Biology (S.-E.J.), Duke University School of Medicine, Durham, North Carolina; Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut (N.R.C., S.B.); and Integrated Toxicology and Environmental Health Program (ITEHP), Nicholas School of the Environment, Duke University, Durham, North Carolina (S.-E.J.)
| | - Narendranath Reddy Chintagari
- Center for Translational Pain Medicine, Department of Anesthesiology (S.A., B.L., S.-E.J.) and Department of Pharmacology and Cancer Biology (S.-E.J.), Duke University School of Medicine, Durham, North Carolina; Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut (N.R.C., S.B.); and Integrated Toxicology and Environmental Health Program (ITEHP), Nicholas School of the Environment, Duke University, Durham, North Carolina (S.-E.J.)
| | - Shrilatha Balakrishna
- Center for Translational Pain Medicine, Department of Anesthesiology (S.A., B.L., S.-E.J.) and Department of Pharmacology and Cancer Biology (S.-E.J.), Duke University School of Medicine, Durham, North Carolina; Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut (N.R.C., S.B.); and Integrated Toxicology and Environmental Health Program (ITEHP), Nicholas School of the Environment, Duke University, Durham, North Carolina (S.-E.J.)
| | - Boyi Liu
- Center for Translational Pain Medicine, Department of Anesthesiology (S.A., B.L., S.-E.J.) and Department of Pharmacology and Cancer Biology (S.-E.J.), Duke University School of Medicine, Durham, North Carolina; Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut (N.R.C., S.B.); and Integrated Toxicology and Environmental Health Program (ITEHP), Nicholas School of the Environment, Duke University, Durham, North Carolina (S.-E.J.)
| | - Sven-Eric Jordt
- Center for Translational Pain Medicine, Department of Anesthesiology (S.A., B.L., S.-E.J.) and Department of Pharmacology and Cancer Biology (S.-E.J.), Duke University School of Medicine, Durham, North Carolina; Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut (N.R.C., S.B.); and Integrated Toxicology and Environmental Health Program (ITEHP), Nicholas School of the Environment, Duke University, Durham, North Carolina (S.-E.J.)
| |
Collapse
|
7
|
Vlachova V, Barvik I, Zimova L. Human Transient Receptor Potential Ankyrin 1 Channel: Structure, Function, and Physiology. Subcell Biochem 2024; 104:207-244. [PMID: 38963489 DOI: 10.1007/978-3-031-58843-3_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/05/2024]
Abstract
The transient receptor potential ion channel TRPA1 is a Ca2+-permeable nonselective cation channel widely expressed in sensory neurons, but also in many nonneuronal tissues typically possessing barrier functions, such as the skin, joint synoviocytes, cornea, and the respiratory and intestinal tracts. Here, the primary role of TRPA1 is to detect potential danger stimuli that may threaten the tissue homeostasis and the health of the organism. The ability to directly recognize signals of different modalities, including chemical irritants, extreme temperatures, or osmotic changes resides in the characteristic properties of the ion channel protein complex. Recent advances in cryo-electron microscopy have provided an important framework for understanding the molecular basis of TRPA1 function and have suggested novel directions in the search for its pharmacological regulation. This chapter summarizes the current knowledge of human TRPA1 from a structural and functional perspective and discusses the complex allosteric mechanisms of activation and modulation that play important roles under physiological or pathophysiological conditions. In this context, major challenges for future research on TRPA1 are outlined.
Collapse
Affiliation(s)
- Viktorie Vlachova
- Department of Cellular Neurophysiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic.
| | - Ivan Barvik
- Division of Biomolecular Physics, Institute of Physics, Faculty of Mathematics and Physics, Charles University, Prague, Czech Republic.
| | - Lucie Zimova
- Department of Cellular Neurophysiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic.
| |
Collapse
|
8
|
Patil MJ, Kim SH, Bahia PK, Nair SS, Darcey TS, Fiallo J, Zhu XX, Frisina RD, Hadley SH, Taylor-Clark TE. A Novel Flp Reporter Mouse Shows That TRPA1 Expression Is Largely Limited to Sensory Neuron Subsets. eNeuro 2023; 10:ENEURO.0350-23.2023. [PMID: 37989590 PMCID: PMC10698635 DOI: 10.1523/eneuro.0350-23.2023] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 11/09/2023] [Accepted: 11/11/2023] [Indexed: 11/23/2023] Open
Abstract
Transient receptor potential ankyrin 1 (TRPA1) is a polymodal cation channel that is activated by electrophilic irritants, oxidative stress, cold temperature, and GPCR signaling. TRPA1 expression has been primarily identified in subsets of nociceptive sensory afferents and is considered a target for future analgesics. Nevertheless, TRPA1 has been implicated in other cell types including keratinocytes, epithelium, enterochromaffin cells, endothelium, astrocytes, and CNS neurons. Here, we developed a knock-in mouse that expresses the recombinase FlpO in TRPA1-expressing cells. We crossed the TRPA1Flp mouse with the R26ai65f mouse that expresses tdTomato in a Flp-sensitive manner. We found tdTomato expression correlated well with TRPA1 mRNA expression and sensitivity to TRPA1 agonists in subsets of TRPV1 (transient receptor potential vanilloid receptor type 1)-expressing neurons in the vagal ganglia and dorsal root ganglia (DRGs), although tdTomato expression efficiency was limited in DRG. We observed tdTomato-expressing afferent fibers centrally (in the medulla and spinal cord) and peripherally in the esophagus, gut, airways, bladder, and skin. Furthermore, chemogenetic activation of TRPA1-expressing nerves in the paw evoked flinching behavior. tdTomato expression was very limited in other cell types. We found tdTomato in subepithelial cells in the gut mucosa but not in enterochromaffin cells. tdTomato was also observed in supporting cells within the cochlea, but not in hair cells. Lastly, tdTomato was occasionally observed in neurons in the somatomotor cortex and the piriform area, but not in astrocytes or vascular endothelium. Thus, this novel mouse strain may be useful for mapping and manipulating TRPA1-expressing cells and deciphering the role of TRPA1 in physiological and pathophysiological processes.
Collapse
Affiliation(s)
- Mayur J Patil
- Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida 33612
| | - Seol-Hee Kim
- Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida 33612
| | - Parmvir K Bahia
- Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida 33612
| | - Sanjay S Nair
- Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida 33612
| | - Teresa S Darcey
- Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida 33612
| | - Jailene Fiallo
- Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida 33612
| | - Xiao Xia Zhu
- Medical Engineering, College of Engineering, University of South Florida, Tampa, Florida 33620
| | - Robert D Frisina
- Medical Engineering, College of Engineering, University of South Florida, Tampa, Florida 33620
| | - Stephen H Hadley
- Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida 33612
| | - Thomas E Taylor-Clark
- Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida 33612
| |
Collapse
|
9
|
Akasaka R, Furukawa A, Hayashi Y, Hitomi S, Koyama R, Oshima E, Tamura M, Yonemoto M, Hojo Y, Takahashi R, Shibuta I, Iwata K, Yonehara Y, Shinoda M. PAR2-dependent phosphorylation of TRPV4 at the trigeminal nerve terminals contributes to tongue cancer pain. J Oral Biosci 2023; 65:356-364. [PMID: 37838226 DOI: 10.1016/j.job.2023.10.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/10/2023] [Accepted: 10/10/2023] [Indexed: 10/16/2023]
Abstract
OBJECTIVE This study aimed to clarify the interactions between the tongue and primary afferent fibers in tongue cancer pain. METHODS A pharmacological analysis was conducted to evaluate mechanical hypersensitivity of the tongues of rats with squamous cell carcinoma (SCC). Changes in trigeminal ganglion (TG) neurons projecting to the tongue were analyzed using immunohistochemistry and western blotting. RESULTS SCC inoculation of the tongue caused persistent mechanical sensitization and tumor formation. Trypsin expression was significantly upregulated in cancer lesions. Continuous trypsin inhibition or protease-activated receptor 2 (PAR2) antagonism in the tongue significantly inhibited SCC-induced mechanical sensitization. No changes were observed in PAR2 and transient receptor potential vanilloid 4 (TRPV4) levels in the TG or the number of PAR2-and TRPV4-expressing TG neurons after SCC inoculation. In contrast, the relative amount of phosphorylated TRPV4 in the TG was significantly increased after SCC inoculation and abrogated by PAR2 antagonism in the tongue. TRPV4 antagonism in the tongue significantly ameliorated the mechanical sensitization caused by SCC inoculation. CONCLUSIONS Our findings indicate that tumor-derived trypsin sensitizes primary afferent fibers by PAR2 stimulation and subsequent TRPV4 phosphorylation, resulting in severe tongue pain.
Collapse
Affiliation(s)
- Ryuta Akasaka
- Department of Oral and Maxillofacial Surgery II, Nihon University School of Dentistry, Tokyo, 101-8310, Japan
| | - Akihiko Furukawa
- Department of Oral and Maxillofacial Surgery II, Nihon University School of Dentistry, Tokyo, 101-8310, Japan
| | - Yoshinori Hayashi
- Department of Physiology, Nihon University School of Dentistry, Tokyo, 101-8310, Japan.
| | - Suzuro Hitomi
- Department of Physiology, Nihon University School of Dentistry, Tokyo, 101-8310, Japan
| | - Ryo Koyama
- Department of Oral and Maxillofacial Surgery II, Nihon University School of Dentistry, Tokyo, 101-8310, Japan
| | - Eri Oshima
- Department of Physiology, Nihon University School of Dentistry, Tokyo, 101-8310, Japan
| | - Miki Tamura
- Department of Physiology, Nihon University School of Dentistry, Tokyo, 101-8310, Japan
| | - Mamiko Yonemoto
- Department of Physiology, Nihon University School of Dentistry, Tokyo, 101-8310, Japan
| | - Yasushi Hojo
- Department of Physiology, Nihon University School of Dentistry, Tokyo, 101-8310, Japan
| | - Ryosuke Takahashi
- Department of Physiology, Nihon University School of Dentistry, Tokyo, 101-8310, Japan
| | - Ikuko Shibuta
- Department of Physiology, Nihon University School of Dentistry, Tokyo, 101-8310, Japan
| | - Koichi Iwata
- Department of Physiology, Nihon University School of Dentistry, Tokyo, 101-8310, Japan
| | - Yoshiyuki Yonehara
- Department of Oral and Maxillofacial Surgery II, Nihon University School of Dentistry, Tokyo, 101-8310, Japan
| | - Masamichi Shinoda
- Department of Physiology, Nihon University School of Dentistry, Tokyo, 101-8310, Japan
| |
Collapse
|
10
|
Luostarinen S, Hämäläinen M, Pemmari A, Moilanen E. The regulation of TRPA1 expression and function by Th1 and Th2-type inflammation in human A549 lung epithelial cells. Inflamm Res 2023:10.1007/s00011-023-01750-y. [PMID: 37386145 DOI: 10.1007/s00011-023-01750-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/14/2023] [Accepted: 03/02/2023] [Indexed: 07/01/2023] Open
Abstract
BACKGROUND Transient Receptor Potential Ankyrin 1 (TRPA1) is a cation channel that mediates pain, itch, cough, and neurogenic inflammation in response to pungent compounds such as acrolein in cigarette smoke. TRPA1 is also activated by endogenous factors and promotes inflammation in asthma models. We have recently shown that TRPA1 is upregulated by inflammatory cytokines in A549 human lung epithelial cells. Here, we explored the effects of Th1 and Th2-type inflammation on TRPA1. METHODS AND RESULTS TRPA1 expression and function was studied in A549 human lung epithelial cells. To induce inflammation, the cells were exposed to a combination of cytokines TNF-α and IL-1β; and to model Th1 or Th2-type responses, IFN-γ or IL-4/IL-13 was added, respectively. TRPA1 expression (measured by RT-PCR and Western blot) and function (assessed by Fluo-3AM intracellular calcium measurement) was enhanced under the influence of TNF-α + IL-1β. IFN-γ further enhanced TRPA1 expression and function, whereas IL-4 and IL-13 suppressed them. The effects of IFN-γ and IL-4 on TRPA1 expression were reversed by the Janus kinase (JAK) inhibitors baricitinib and tofacitinib, and those of IL-4 also by the STAT6 inhibitor AS1517499. The glucocorticoid dexamethasone downregulated TRPA1 expression, whereas the PDE4 inhibitor rolipram had no effect. Under all conditions, TRPA1 blockade was found to reduce the production of LCN2 and CXCL6. CONCLUSIONS TRPA1 expression and function in lung epithelial cells was upregulated under inflammatory conditions. IFN-γ further increased TRPA1 expression while IL-4 and IL-13 suppressed that in a JAK-STAT6 dependent manner which is novel. TRPA1 also modulated the expression of genes relevant to innate immunity and lung disease. We propose that the paradigm of Th1 and Th2 inflammation is a major determinant of TRPA1 expression and function, which should be considered when targeting TRPA1 for pharmacotherapy in inflammatory (lung) disease.
Collapse
Affiliation(s)
- Samu Luostarinen
- The Immunopharmacology Research Group, Faculty of Medicine and Health Technology, Tampere University and Tampere University Hospital, Tampere, Finland
| | - Mari Hämäläinen
- The Immunopharmacology Research Group, Faculty of Medicine and Health Technology, Tampere University and Tampere University Hospital, Tampere, Finland
| | - Antti Pemmari
- The Immunopharmacology Research Group, Faculty of Medicine and Health Technology, Tampere University and Tampere University Hospital, Tampere, Finland
| | - Eeva Moilanen
- The Immunopharmacology Research Group, Faculty of Medicine and Health Technology, Tampere University and Tampere University Hospital, Tampere, Finland.
| |
Collapse
|
11
|
Cohen CF, Roh J, Lee SH, Park CK, Berta T. Targeting Nociceptive Neurons and Transient Receptor Potential Channels for the Treatment of Migraine. Int J Mol Sci 2023; 24:ijms24097897. [PMID: 37175602 PMCID: PMC10177956 DOI: 10.3390/ijms24097897] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 04/22/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
Migraine is a neurovascular disorder that affects approximately 12% of the global population. While its exact causes are still being studied, researchers believe that nociceptive neurons in the trigeminal ganglia play a key role in the pain signals of migraine. These nociceptive neurons innervate the intracranial meninges and convey pain signals from the meninges to the thalamus. Targeting nociceptive neurons is considered promising due to their accessibility and distinct molecular profile, which includes the expression of several transient receptor potential (TRP) channels. These channels have been linked to various pain conditions, including migraine. This review discusses the role and mechanisms of nociceptive neurons in migraine, the challenges of current anti-migraine drugs, and the evidence for well-studied and emerging TRP channels, particularly TRPC4, as novel targets for migraine prevention and treatment.
Collapse
Affiliation(s)
- Cinder Faith Cohen
- Pain Research Center, Department of Anesthesiology, Medical Center, University of Cincinnati, Cincinnati, OH 45219, USA
- Neuroscience Graduate Program, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Jueun Roh
- Pain Research Center, Department of Anesthesiology, Medical Center, University of Cincinnati, Cincinnati, OH 45219, USA
- Department of Physiology, Gachon Pain Center, College of Medicine, Gachon University, Incheon 21936, Republic of Korea
| | - Sang Hoon Lee
- Pain Research Center, Department of Anesthesiology, Medical Center, University of Cincinnati, Cincinnati, OH 45219, USA
- Neuroscience Graduate Program, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Chul-Kyu Park
- Department of Physiology, Gachon Pain Center, College of Medicine, Gachon University, Incheon 21936, Republic of Korea
| | - Temugin Berta
- Pain Research Center, Department of Anesthesiology, Medical Center, University of Cincinnati, Cincinnati, OH 45219, USA
| |
Collapse
|
12
|
Abstract
The ability to detect stimuli from the environment plays a pivotal role in our survival. The molecules that allow the detection of such signals include ion channels, which are proteins expressed in different cells and organs. Among these ion channels, the transient receptor potential (TRP) family responds to the presence of diverse chemicals, temperature, and osmotic changes, among others. This family of ion channels includes the TRPV or vanilloid subfamily whose members serve several physiological functions. Although these proteins have been studied intensively for the last two decades, owing to their structural and functional complexities, a number of controversies regarding their function still remain. Here, we discuss some salient features of their regulation in light of these controversies and outline some of the efforts pushing the field forward.
Collapse
Affiliation(s)
- Tamara Rosenbaum
- Department of Cognitive Neuroscience, Neuroscience Division, Institute for Cellular Physiology, National Autonomous University of Mexico, Coyoacán, México;
| | - León D Islas
- Department of Physiology, School of Medicine, National Autonomous University of Mexico, Coyoacán, México
| |
Collapse
|
13
|
Sánchez JC, Muñoz LV, Galindo-Márquez ML, Valencia-Vásquez A, García AM. Paclitaxel Regulates TRPA1 Function and Expression Through PKA and PKC. Neurochem Res 2023; 48:295-304. [PMID: 36098890 PMCID: PMC9823074 DOI: 10.1007/s11064-022-03748-0] [Citation(s) in RCA: 7] [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: 03/11/2022] [Revised: 08/05/2022] [Accepted: 08/30/2022] [Indexed: 01/11/2023]
Abstract
Paclitaxel (PTX) is a frequently used anticancer drug that causes peripheral neuropathy. Transient receptor potential ankyrin 1 (TRPA1), a plasma membrane calcium channel, has been associated with PTX toxicity and with other chemotherapy agents such as oxaliplatin and vincristine. However, the effect of PTX on the functional expression and calcium currents of TRPA1 has not been determined. The present study shows the effect of PTX on TRPA1 activity in a neuronal cell line (SH-SY5Y). The effect of PTX on the expression of TRPA1 was assessed through quantitative PCR and Western blot analyses to determine the relative mRNA and protein expression levels. To assess the effect on calcium flux and currents, cells were exposed to PTX; simultaneously, a specific agonist and antagonist of TRPA1 were added to evaluate the differential response in exposed versus control cells. To assess the effect of PKA, PKC and PI3K on PTX-induced TRPA1 increased activity, selective inhibitors were added to these previous experiments. PTX increased the mRNA and protein expression of TRPA1 as well as the TRPA1-mediated Ca2+ currents and intracellular Ca2+ concentrations. This effect was dependent on AITC (a selective specific agonist) and was abolished with HC-030031 (a selective specific antagonist). The inhibition of PKA and PKC reduced the effect of PTX on the functional expression of TRPA1, whereas the inhibition of PI3K had no effects. PTX-induced neuropathy involves TRPA1 activity through an increase in functional expression and is regulated by PKA and PKC signaling. These findings support the role of the TRPA1 channel in the mechanisms altered by PTX, which can be involved in the process that lead to chemotherapy-induced neuropathy.
Collapse
Affiliation(s)
- Julio C Sánchez
- Faculty of Health Sciences, Universidad Tecnológica de Pereira, AA 97, La Julita, 660003, Pereira, Colombia.
| | - Laura V Muñoz
- Faculty of Health Sciences, Universidad Tecnológica de Pereira, AA 97, La Julita, 660003, Pereira, Colombia
| | | | - Aníbal Valencia-Vásquez
- Faculty of Health Sciences, Universidad Tecnológica de Pereira, AA 97, La Julita, 660003, Pereira, Colombia
| | - Andrés M García
- Faculty of Health Sciences, Universidad Tecnológica de Pereira, AA 97, La Julita, 660003, Pereira, Colombia
| |
Collapse
|
14
|
Rosenbaum T, Morales-Lázaro SL. Regulation of ThermoTRP Channels by PIP2 and Cholesterol. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1422:245-277. [PMID: 36988884 DOI: 10.1007/978-3-031-21547-6_9] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
Abstract
Transient receptor potential (TRP) ion channels are proteins that are expressed by diverse tissues and that play pivotal functions in physiology. These channels are polymodal and are activated by several stimuli. Among TRPs, some members of this family of channels respond to changes in ambient temperature and are known as thermoTRPs. These proteins respond to heat or cold in the noxious range and some of them to temperatures considered innocuous, as well as to mechanical, osmotic, and/or chemical stimuli. In addition to this already complex ability to respond to different signals, the activity of these ion channels can be fine-tuned by lipids. Two lipids well known to modulate ion channel activity are phosphatidylinositol 4,5-bisphosphate (PIP2) and cholesterol. These lipids can either influence the function of these proteins through direct interaction by binding to a site in the structure of the ion channel or through indirect mechanisms, which can include modifying membrane properties, such as curvature and rigidity, by regulating their expression or by modulating the actions of other molecules or signaling pathways that affect the physiology of ion channels. Here, we summarize the key aspects of the regulation of thermoTRP channels by PIP2 and cholesterol.
Collapse
Affiliation(s)
- Tamara Rosenbaum
- Departamento de Neurociencia Cognitiva, División Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, Mexico.
| | - Sara L Morales-Lázaro
- Departamento de Neurociencia Cognitiva, División Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| |
Collapse
|
15
|
Yao K, Dou B, Zhang Y, Chen Z, Li Y, Fan Z, Ma Y, Du S, Wang J, Xu Z, Liu Y, Lin X, Wang S, Guo Y. Inflammation-the role of TRPA1 channel. Front Physiol 2023; 14:1093925. [PMID: 36875034 PMCID: PMC9977828 DOI: 10.3389/fphys.2023.1093925] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 02/08/2023] [Indexed: 02/18/2023] Open
Abstract
Recently, increasing numbers of studies have demonstrated that transient receptor potential ankyrin 1 (TRPA1) can be used as a potential target for the treatment of inflammatory diseases. TRPA1 is expressed in both neuronal and non-neuronal cells and is involved in diverse physiological activities, such as stabilizing of cell membrane potential, maintaining cellular humoral balance, and regulating intercellular signal transduction. TRPA1 is a multi-modal cell membrane receptor that can sense different stimuli, and generate action potential signals after activation via osmotic pressure, temperature, and inflammatory factors. In this study, we introduced the latest research progress on TRPA1 in inflammatory diseases from three different aspects. First, the inflammatory factors released after inflammation interacts with TRPA1 to promote inflammatory response; second, TRPA1 regulates the function of immune cells such as macrophages and T cells, In addition, it has anti-inflammatory and antioxidant effects in some inflammatory diseases. Third, we have summarized the application of antagonists and agonists targeting TRPA1 in the treatment of some inflammatory diseases.
Collapse
Affiliation(s)
- Kaifang Yao
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Baomin Dou
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yue Zhang
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhihan Chen
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yanwei Li
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zezhi Fan
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yajing Ma
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Simin Du
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jiangshan Wang
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhifang Xu
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yangyang Liu
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Xiaowei Lin
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Shenjun Wang
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yi Guo
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China.,School of Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
16
|
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
|
17
|
Khan S, Patra PH, Somerfield H, Benya-Aphikul H, Upadhya M, Zhang X. IQGAP1 promotes chronic pain by regulating the trafficking and sensitization of TRPA1 channels. Brain 2022:6881565. [PMID: 36477832 DOI: 10.1093/brain/awac462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 11/14/2022] [Accepted: 11/24/2022] [Indexed: 12/12/2022] Open
Abstract
TRPA1 channels have been implicated in mechanical and cold hypersensitivity in chronic pain. But how TRPA1 mediates this process is unclear. Here we show that IQ-motif containing GTPase activating protein 1 (IQGAP1) is responsible using a combination of biochemical, molecular, Ca2+ imaging and behavioural approaches. TRPA1 and IQGAP1 bind to each other and are highly colocalised in sensory DRG neurons in mice. The expression of IQGAP1 but not TRPA1 is increased in chronic inflammatory and neuropathic pain. However, TRPA1 undergoes increased trafficking to the membrane of DRG neurons catalysed by the small GTPase Cdc42 associated with IQGAP1, leading to functional sensitization of the channel. Activation of PKA is also sufficient to evoke TRPA1 trafficking and sensitization. All these responses are, however, completely prevented in the absence of IQGAP1. Concordantly, deletion of IQGAP1 markedly reduces mechanical and cold hypersensitivity in chronic inflammatory and neuropathic pain in mice. IQGAP1 thus promotes chronic pain by coupling the trafficking and signalling machineries to TRPA1 channels.
Collapse
Affiliation(s)
- Shakil Khan
- School of Health & Life Sciences, Aston University, Birmingham B4 7ET, UK
| | - Pabitra H Patra
- School of Health & Life Sciences, Aston University, Birmingham B4 7ET, UK
| | - Hannah Somerfield
- School of Health & Life Sciences, Aston University, Birmingham B4 7ET, UK
| | | | - Manoj Upadhya
- School of Health & Life Sciences, Aston University, Birmingham B4 7ET, UK
| | - Xuming Zhang
- School of Health & Life Sciences, Aston University, Birmingham B4 7ET, UK
- School of Life Sciences, University of Warwick, Coventry CV4 7AL, UK
| |
Collapse
|
18
|
Ślęczkowska M, Almomani R, Marchi M, Salvi E, de Greef BTA, Sopacua M, Hoeijmakers JGJ, Lindsey P, Waxman SG, Lauria G, Faber CG, Smeets HJM, Gerrits MM. Peripheral Ion Channel Genes Screening in Painful Small Fiber Neuropathy. Int J Mol Sci 2022; 23:ijms232214095. [PMID: 36430572 PMCID: PMC9696564 DOI: 10.3390/ijms232214095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/09/2022] [Accepted: 11/10/2022] [Indexed: 11/18/2022] Open
Abstract
Neuropathic pain is a characteristic feature of small fiber neuropathy (SFN), which in 18% of the cases is caused by genetic variants in voltage-gated sodium ion channels. In this study, we assessed the role of fifteen other ion channels in neuropathic pain. Patients with SFN (n = 414) were analyzed for ANO1, ANO3, HCN1, KCNA2, KCNA4, KCNK18, KCNN1, KCNQ3, KCNQ5, KCNS1, TRPA1, TRPM8, TRPV1, TRPV3 and TRPV4 variants by single-molecule molecular inversion probes-next-generation sequencing. These patients did not have genetic variants in SCN3A, SCN7A-SCN11A and SCN1B-SCN4B. In twenty patients (20/414, 4.8%), a potentially pathogenic heterozygous variant was identified in an ion-channel gene (ICG). Variants were present in seven genes, for two patients (0.5%) in ANO3, one (0.2%) in KCNK18, two (0.5%) in KCNQ3, seven (1.7%) in TRPA1, three (0.7%) in TRPM8, three (0.7%) in TRPV1 and two (0.5%) in TRPV3. Variants in the TRP genes were the most frequent (n = 15, 3.6%), partly in patients with high mean maximal pain scores VAS = 9.65 ± 0.7 (n = 4). Patients with ICG variants reported more severe pain compared to patients without such variants (VAS = 9.36 ± 0.72 vs. VAS = 7.47 ± 2.37). This cohort study identified ICG variants in neuropathic pain in SFN, complementing previous findings of ICG variants in diabetic neuropathy. These data show that ICG variants are central in neuropathic pain of different etiologies and provides promising gene candidates for future research.
Collapse
Affiliation(s)
- Milena Ślęczkowska
- Department of Toxicogenomics, Maastricht University, 6229 ER Maastricht, The Netherlands
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Centre+, 6229 ER Maastricht, The Netherlands
| | - Rowida Almomani
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Centre+, 6229 ER Maastricht, The Netherlands
- Department of Medical Laboratory Sciences, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Margherita Marchi
- Neuroalgology Unit, IRCCS Foundation “Carlo Besta” Neurological Institute, 20133 Milan, Italy
| | - Erika Salvi
- Neuroalgology Unit, IRCCS Foundation “Carlo Besta” Neurological Institute, 20133 Milan, Italy
| | - Bianca T A de Greef
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Centre+, 6229 ER Maastricht, The Netherlands
| | - Maurice Sopacua
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Centre+, 6229 ER Maastricht, The Netherlands
| | - Janneke G J Hoeijmakers
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Centre+, 6229 ER Maastricht, The Netherlands
| | - Patrick Lindsey
- Department of Toxicogenomics, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Stephen G Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Giuseppe Lauria
- Neuroalgology Unit, IRCCS Foundation “Carlo Besta” Neurological Institute, 20133 Milan, Italy
| | - Catharina G Faber
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Centre+, 6229 ER Maastricht, The Netherlands
- Correspondence:
| | - Hubert J M Smeets
- Department of Toxicogenomics, Maastricht University, 6229 ER Maastricht, The Netherlands
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Centre+, 6229 ER Maastricht, The Netherlands
| | - Monique M Gerrits
- Department of Clinical Genetics, Maastricht University Medical Centre+, 6229 HX Maastricht, The Netherlands
| |
Collapse
|
19
|
Afzal R, Shim WS. Activation of serotonin receptor 2 by glucosylsphingosine can be enhanced by TRPA1 but not TRPV1: Implication of a novel glucosylsphingosine-mediated itch pathway. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2022; 1864:184014. [PMID: 35908608 DOI: 10.1016/j.bbamem.2022.184014] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 07/13/2022] [Accepted: 07/22/2022] [Indexed: 06/15/2023]
Abstract
Glucosylsphingosine (GS) is an endogenous sphingolipid that specifically accumulates in the skin of patients with atopic dermatitis (AD). Notably, it was recently found that GS can induce itch sensation by activating serotonin receptor 2A and TRPV4 ion channels. However, it is still uncertain whether other molecules are involved in GS-induced itch sensation. Therefore, by using the calcium imaging technique, we investigated whether serotonin receptor 2 - specifically 2A and 2B - can interact with TRPV1 and TRPA1, because these are representative ion channels in the transmission of itch. As a result, it was found that GS did not activate TRPV1 or TRPA1 per se. Moreover, cells expressing both serotonin receptor 2 and TRPV1 did not show any changes in calcium responses. However, enhanced calcium responses were observed in cells expressing serotonin receptor 2 and TRPA1, suggesting a possible interaction between these two molecules. Similar synergistic effects were also observed in cells expressing serotonin receptor 2 and TRPA1, but not TRPV1. Furthermore, a phospholipase C inhibitor (U73122) and a store-operated calcium entry blocker (SKF96365) significantly reduced GS-induced responses in cells expressing both serotonin receptor 2 and TRPA1, but not with pre-treatment with a Gβγ-complex blocker (gallein). Therefore, we propose a putative novel pathway for GS-induced itch sensation, such that serotonin receptor 2 could be coupled to TRPA1 but not TRPV1 in sensory neurons.
Collapse
Affiliation(s)
- Ramsha Afzal
- Department of Ophthalmology, Incheon St. Mary Hospital, College of Medicine, The Catholic University of Korea, 58 Dongsu-ro, Bupyeong-gu, Incheon 403-720, Republic of Korea
| | - Won-Sik Shim
- College of Pharmacy, Gachon University, Hambangmoe-ro 191, Yeonsu-gu, Incheon 21936, Republic of Korea; Gachon Institute of Pharmaceutical Sciences, Hambangmoe-ro 191, Yeonsu-gu, Incheon 21936, Republic of Korea.
| |
Collapse
|
20
|
Zhuo X, Wu Y, Fu X, Liang X, Xiang Y, Li J, Mao C, Jiang Y. The Yin‐Yang roles of protease‐activated receptors in inflammatory signalling and diseases. FEBS J 2022; 289:4000-4020. [DOI: 10.1111/febs.16406] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 01/26/2022] [Accepted: 02/15/2022] [Indexed: 12/15/2022]
Affiliation(s)
- Xin Zhuo
- School of Life Science and Engineering Southwest Jiaotong University Chengdu China
| | - Yue Wu
- School of Life Science and Engineering Southwest Jiaotong University Chengdu China
| | - Xiujuan Fu
- School of Life Science and Engineering Southwest Jiaotong University Chengdu China
| | - Xiaoyu Liang
- School of Life Science and Engineering Southwest Jiaotong University Chengdu China
| | - Yuxin Xiang
- School of Life Science and Engineering Southwest Jiaotong University Chengdu China
| | - Jianbin Li
- School of Life Science and Engineering Southwest Jiaotong University Chengdu China
| | - Canquan Mao
- School of Life Science and Engineering Southwest Jiaotong University Chengdu China
| | - Yuhong Jiang
- School of Life Science and Engineering Southwest Jiaotong University Chengdu China
| |
Collapse
|
21
|
Gao Y, Mei C, Chen P, Chen X. The contribution of neuro-immune crosstalk to pain in the peripheral nervous system and the spinal cord. Int Immunopharmacol 2022; 107:108700. [DOI: 10.1016/j.intimp.2022.108700] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/23/2022] [Accepted: 03/10/2022] [Indexed: 12/16/2022]
|
22
|
Iannone LF, Nassini R, Patacchini R, Geppetti P, De Logu F. Neuronal and non-neuronal TRPA1 as therapeutic targets for pain and headache relief. Temperature (Austin) 2022; 10:50-66. [PMID: 37187829 PMCID: PMC10177743 DOI: 10.1080/23328940.2022.2075218] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 05/02/2022] [Accepted: 05/04/2022] [Indexed: 10/18/2022] Open
Abstract
The transient receptor potential ankyrin 1 (TRPA1), a member of the TRP superfamily of channels, has a major role in different types of pain. TRPA1 is primarily localized to a subpopulation of primary sensory neurons of the trigeminal, vagal, and dorsal root ganglia. This subset of nociceptors produces and releases the neuropeptide substance P (SP) and calcitonin gene-related peptide (CGRP), which mediate neurogenic inflammation. TRPA1 is characterized by unique sensitivity for an unprecedented number of reactive byproducts of oxidative, nitrative, and carbonylic stress and to be activated by several chemically heterogenous, exogenous, and endogenous compounds. Recent preclinical evidence has revealed that expression of TRPA1 is not limited to neurons, but its functional role has been reported in central and peripheral glial cells. In particular, Schwann cell TRPA1 was recently implicated in sustaining mechanical and thermal (cold) hypersensitivity in mouse models of macrophage-dependent and macrophage-independent inflammatory, neuropathic, cancer, and migraine pain. Some analgesics and herbal medicines/natural products widely used for the acute treatment of pain and headache have shown some inhibitory action at TRPA1. A series of high affinity and selective TRPA1 antagonists have been developed and are currently being tested in phase I and phase II clinical trials for different diseases with a prominent pain component. Abbreviations: 4-HNE, 4-hydroxynonenal; ADH-2, alcohol dehydrogenase-2; AITC, allyl isothiocyanate; ANKTD, ankyrin-like protein with transmembrane domains protein 1; B2 receptor, bradykinin 2 receptor; CIPN, chemotherapeutic-induced peripheral neuropathy; CGRP, calcitonin gene related peptide; CRISPR, clustered regularly interspaced short palindromic repeats; CNS, central nervous system; COOH, carboxylic terminal; CpG, C-phosphate-G; DRG, dorsal root ganglia; EP, prostaglandins; GPCR, G-protein-coupled receptors; GTN, glyceryl trinitrate; MAPK, mitogen-activated protein kinase; M-CSF, macrophage-colony stimulating factor; NAPQI, N-Acetyl parabenzoquinone-imine; NGF, nerve growth factor; NH2, amino terminal; NKA, neurokinin A; NO, nitric oxide; NRS, numerical rating scale; PAR2, protease-activated receptor 2; PMA, periorbital mechanical allodynia; PLC, phospholipase C; PKC, protein kinase C; pSNL, partial sciatic nerve ligation; RCS, reactive carbonyl species; ROS, reactive oxygen species; RNS, nitrogen oxygen species; SP, substance P; TG, trigeminal ganglion; THC, Δ9-tetrahydrocannabinol; TrkA, neurotrophic receptor tyrosine kinase A; TRP, transient receptor potential; TRPC, TRP canonical; TRPM, TRP melastatin; TRPP, TRP polycystin; TRPM, TRP mucolipin; TRPA, TRP ankyrin; TRPV, TRP vanilloid; VG, vagal ganglion.
Collapse
Affiliation(s)
- Luigi F. Iannone
- 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
| | - Romina Nassini
- Section of Clinical Pharmacology and Oncology, Department of Health Sciences, University of Florence, Florence, Italy
| | - Riccardo Patacchini
- Corporate Drug Development, Chiesi Farmaceutici S.p.A, Nuovo Centro Ricerche, Parma, 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 Logu
- Section of Clinical Pharmacology and Oncology, Department of Health Sciences, University of Florence, Florence, Italy
| |
Collapse
|
23
|
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
|
24
|
TRPA1 promotes melanosome phagocytosis in keratinocytes via PAR-2/CYLD axis. J Dermatol Sci 2022; 106:181-188. [DOI: 10.1016/j.jdermsci.2022.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 05/06/2022] [Accepted: 05/15/2022] [Indexed: 11/23/2022]
|
25
|
Balcziak LK, Russo AF. Dural Immune Cells, CGRP, and Migraine. Front Neurol 2022; 13:874193. [PMID: 35432179 PMCID: PMC9009415 DOI: 10.3389/fneur.2022.874193] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 03/16/2022] [Indexed: 11/26/2022] Open
Abstract
Migraine is the most common neurological disorder in the world, affecting 12% of the population. Migraine involves the central nervous system, trigeminal nerves and meninges. Recent advances have shown that targeting calcitonin gene-related peptide (CGRP) through either antibodies or small molecule receptor antagonists is effective at reducing episodic and chronic migraine episodes, but these therapeutics are not effective in all patients. This suggests that migraine does not have a singular molecular cause but is likely due to dysregulated physiology of multiple mechanisms. An often-overlooked part of migraine is the potential involvement of the immune system. Clinical studies have shown that migraine patients may have dysregulation in their immune system, with abnormal plasma cytokine levels either during the attack or at baseline. In addition, those who are immunocompromised appear to be at a higher risk of migraine-like disorders. A recent study showed that migraine caused changes to transcription of immune genes in the blood, even following treatment with sumatriptan. The dura mater is densely packed with macrophages, mast and dendritic cells, and they have been found to associate with meningeal blood vessels and trigeminal afferent endings. Recent work in mice shows activation and morphological changes of these cells in rodents following the migraine trigger cortical spreading depression. Importantly, each of these immune cell types can respond directly to CGRP. Since immune cells make up a large portion of the dura, have functional responses to CGRP, and interact with trigeminal afferents, CGRP actions on the dural immune system are likely to play key roles in migraine.
Collapse
Affiliation(s)
- Louis K. Balcziak
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, United States
- Neuroscience Graduate Program, University of Iowa, Iowa City, IA, United States
- *Correspondence: Louis K. Balcziak
| | - Andrew F. Russo
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, United States
- Department of Neurology, University of Iowa, Iowa City, IA, United States
- Center for the Prevention and Treatment of Visual Loss, Veterans Administration Health Center, Iowa City, IA, United States
| |
Collapse
|
26
|
Steinhoff M, Ahmad F, Pandey A, Datsi A, AlHammadi A, Al-Khawaga S, Al-Malki A, Meng J, Alam M, Buddenkotte J. Neuro-immune communication regulating pruritus in atopic dermatitis. J Allergy Clin Immunol 2022; 149:1875-1898. [PMID: 35337846 DOI: 10.1016/j.jaci.2022.03.010] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 02/13/2022] [Accepted: 03/10/2022] [Indexed: 11/26/2022]
Abstract
Atopic dermatitis (AD) is a common, chronic-relapsing inflammatory skin disease with significant disease burden. Genetic and environmental trigger factors contribute to AD, activating two of our largest organs, the nervous and immune system. Dysregulation of neuro-immune circuits plays a key role in the pathophysiology of AD causing inflammation, pruritus, pain, and barrier dysfunction. Sensory nerves can be activated by environmental or endogenous trigger factors transmitting itch stimuli to the brain. Upon stimulation, sensory nerve endings also release neuromediators into the skin contributing again to inflammation, barrier dysfunction and itch. Additionally, dysfunctional peripheral and central neuronal structures contribute to neuroinflammation, sensitization, nerve elongation, neuropathic itch, thus chronification and therapy-resistance. Consequently, neuro-immune circuits in skin and central nervous system may be targets to treat pruritus in AD. Cytokines, chemokines, proteases, lipids, opioids, ions excite/sensitize sensory nerve endings not only induce itch but further aggravate/perpetuate inflammation, skin barrier disruption, and pruritus. Thus, targeted therapies for neuro-immune circuits as well as pathway inhibitors (e.g., kinase inhibitors) may be beneficial to control pruritus in AD either in systemic and/or topical form. Understanding neuro-immune circuits and neuronal signaling will optimize our approach to control all pathological mechanisms in AD, inflammation, barrier dysfunction and pruritus.
Collapse
Affiliation(s)
- Martin Steinhoff
- Department of Dermatology and Venereology, Hamad Medical Corporation, Doha, Qatar; Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar; Department of Dermatology, Weill Cornell Medicine-Qatar, Doha, Qatar; Qatar University, College of Medicine, Doha, Qatar; Department of Dermatology, Weill Cornell Medicine, New York, USA.
| | - Fareed Ahmad
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Atul Pandey
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Angeliki Datsi
- Institute for Transplantational Diagnostics and Cell Therapeutics, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Ayda AlHammadi
- Department of Dermatology and Venereology, Hamad Medical Corporation, Doha, Qatar
| | - Sara Al-Khawaga
- Department of Dermatology and Venereology, Hamad Medical Corporation, Doha, Qatar
| | - Aysha Al-Malki
- Department of Dermatology and Venereology, Hamad Medical Corporation, Doha, Qatar
| | - Jianghui Meng
- National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland
| | - Majid Alam
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Joerg Buddenkotte
- Department of Dermatology and Venereology, Hamad Medical Corporation, Doha, Qatar; Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| |
Collapse
|
27
|
Nociception and hypersensitivity involve distinct neurons and molecular transducers in Drosophila. Proc Natl Acad Sci U S A 2022; 119:e2113645119. [PMID: 35294287 PMCID: PMC8944580 DOI: 10.1073/pnas.2113645119] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
SignificanceFunctional plasticity of the nociceptive circuit is a remarkable feature and is of clinical relevance. As an example, nociceptors lower their threshold upon tissue injury, a process known as allodynia that would facilitate healing by guarding the injured areas. However, long-lasting hypersensitivity could lead to chronic pain, a debilitating disease not effectively treated. Therefore, it is crucial to dissect the mechanisms underlying basal nociception and nociceptive hypersensitivity. In both vertebrate and invertebrate species, conserved transient receptor potential (Trp) channels are the primary transducers of noxious stimuli. Here, we provide a precedent that in Drosophila larvae, heat sensing in the nociception and hypersensitivity states is mediated by distinct heat-sensitive neurons and TrpA1 alternative isoforms.
Collapse
|
28
|
Aguilera-Lizarraga J, Hussein H, Boeckxstaens GE. Immune activation in irritable bowel syndrome: what is the evidence? Nat Rev Immunol 2022; 22:674-686. [PMID: 35296814 DOI: 10.1038/s41577-022-00700-9] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/18/2022] [Indexed: 12/15/2022]
Abstract
Irritable bowel syndrome (IBS) is a chronic functional gastrointestinal disorder that is characterized by abdominal pain and an altered defecation pattern. It affects between 5 and 20% of the general population and can seriously impact quality of life. The pathophysiology of IBS is rather complex and multifactorial including, for example, altered signalling by the gut-brain axis, dysbiosis, abnormal visceral pain signalling and intestinal immune activation. The latter has gained particular interest in recent years, with growing insight into the bidirectional communication between the nervous system and the immune system. In this Review, we detail the current evidence suggesting that immune activation contributes to the pathology seen in patients with IBS and discuss the potential mechanisms involved. Moreover, we describe how immune mediators, particularly those released by mast cells, can directly activate or sensitize pain-transmitting nerves, leading to increased pain signalling and abdominal pain. Finally, we discuss the potential of interventions targeting immune activation as a new therapeutic strategy for patients suffering from IBS.
Collapse
Affiliation(s)
- Javier Aguilera-Lizarraga
- Laboratory for Intestinal Neuroimmune Interactions, Translational Research Centre for Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Hind Hussein
- Laboratory for Intestinal Neuroimmune Interactions, Translational Research Centre for Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Guy E Boeckxstaens
- Laboratory for Intestinal Neuroimmune Interactions, Translational Research Centre for Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium.
| |
Collapse
|
29
|
Riccio D, Andersen HH, Arendt-Nielsen L. Mild Skin Heating Evokes Warmth Hyperknesis Selectively for Histaminergic and Serotoninergic Itch in Humans. Acta Derm Venereol 2022; 102:adv00649. [PMID: 35083491 PMCID: PMC9558757 DOI: 10.2340/actadv.v102.173] [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] [Indexed: 11/16/2022] Open
Abstract
Chronic itch can severely affect quality of life. Patients report that their chronic itch can be exacerbated by exposure to warm conditions (“warmth hyperknesis”). The aim of this mechanistic study was to investigate the effect of mild heating of the skin in humans on various experimental models of itch. A total of 18 healthy subjects were recruited to the study. Itch was provoked by histamine, serotonin, or cowhage in 3 different sessions. The provoked area was heated with an infrared lamp, and the skin temperature was either not altered, or was increased by 4°C or 7°C. Subsequent to induction of itch, the itch intensity was recorded for 10 min while the skin was heated continuously throughout the entire period of itch induction. Heating the skin resulted in a significant increase in itch intensity when provoked by histamine or serotonin. It is possible that thermoception and pruriception interact and selectively produce a higher itch intensity in histaminergic and serotoninergic itch.
Collapse
Affiliation(s)
| | | | - Lars Arendt-Nielsen
- Faculty of Medicine, Aalborg University, Fredrik Bajers Vej, D3-212, DK-9220 Aalborg East, Denmark.
| |
Collapse
|
30
|
Upregulation of TRPM3 in bladder afferents is involved in chronic pain in CYP-induced cystitis. Pain 2022; 163:2200-2212. [DOI: 10.1097/j.pain.0000000000002616] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 01/05/2022] [Indexed: 11/26/2022]
|
31
|
Romanova J, Rydlovskaya A, Mochalov S, Proskurina O, Gorokh Y, Nebolsin V. The Effect of Anti-Chemokine Oral Drug XC8 on Cough Triggered by The Agonists of TRPA1 But Not TRPV1 Channels in Guinea Pigs. Pulm Ther 2022; 8:105-122. [PMID: 35133638 PMCID: PMC8824739 DOI: 10.1007/s41030-022-00183-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 01/14/2022] [Indexed: 01/09/2023] Open
Abstract
Introduction Chronic cough heavily affects patients’ quality of life, and there are no effective licensed therapies available. Cough is a complication of severe acute respiratory syndrome-related coronavirus 2 (SARS-CoV-2) infection, asthma, and other diseases. Patients with various diseases have a different profile of tussive responses to diverse cough triggers, thereby suggesting sundry mechanisms of neuronal dysfunctions. Previously, we demonstrated that the small molecule drug XC8 shows a clinical anti-asthmatic effect. The objective of the present study was to investigate the effect of XC8 on cough. Methods We studied the antitussive effect of XC8 on cough induced by agonists activating human transient receptor potential (TRP) cation channels TRPA1 or TRPV1 in guinea pigs. We checked the agonistic/antagonistic activity of XC8 on the human cation channels TRPA1, TRPV1, TRPM8, P2X purinoceptor 2 (P2X2), and human acid sensing ion channel 3 (hASIC3) in Fluorescent Imaging Plate Reader (FLIPR) assay. Results XC8 demonstrated clear antitussive activity and dose-dependently inhibited cough in guinea pigs induced by citric acid alone (up to 67.1%) or in combination with IFN-γ (up to 76.4%). XC8 suppressed cough reflexes induced by the repeated inhalation of citric acid (up to 80%) or by cinnamaldehyde (up to 60%). No activity of XC8 against cough evoked by capsaicin was revealed. No direct agonistic/antagonistic activity of XC8 on human TRPA1, TRPV1, TRPM8, P2X2, or hASIC3 was detected. Conclusions XC8 acts against cough evoked by the activation of TRPA1 (citric acid/cinnamaldehyde) but not TRPV1 (capsaicin) channels. XC8 inhibits the cough reflex and suppresses the cough potentiation by IFN-γ. XC8 might be of significant therapeutic value for patients suffering from chronic cough associated with inflammation.
Collapse
Affiliation(s)
- Julia Romanova
- Pharmenterprises LLC, 42 Bolshoj Blvd., Building 1, office 771, 772, Skolkovo Innovation Centre, Moscow, 121205, Russian Federation.
| | - Anastasia Rydlovskaya
- Pharmenterprises LLC, 42 Bolshoj Blvd., Building 1, office 771, 772, Skolkovo Innovation Centre, Moscow, 121205, Russian Federation
| | - Stepan Mochalov
- Pharmenterprises LLC, 42 Bolshoj Blvd., Building 1, office 771, 772, Skolkovo Innovation Centre, Moscow, 121205, Russian Federation
| | - Oxana Proskurina
- Pharmenterprises LLC, 42 Bolshoj Blvd., Building 1, office 771, 772, Skolkovo Innovation Centre, Moscow, 121205, Russian Federation
| | - Yulia Gorokh
- Pharmenterprises LLC, 42 Bolshoj Blvd., Building 1, office 771, 772, Skolkovo Innovation Centre, Moscow, 121205, Russian Federation
| | - Vladimir Nebolsin
- Pharmenterprises LLC, 42 Bolshoj Blvd., Building 1, office 771, 772, Skolkovo Innovation Centre, Moscow, 121205, Russian Federation
| |
Collapse
|
32
|
The Emerging Pro-Algesic Profile of Transient Receptor Potential Vanilloid Type 4. Rev Physiol Biochem Pharmacol 2022; 186:57-93. [PMID: 36378366 DOI: 10.1007/112_2022_75] [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: 11/16/2022]
Abstract
Transient receptor potential vanilloid type 4 (TRPV4) channels are Ca2+-permeable non-selective cation channels which mediate a wide range of physiological functions and are activated and modulated by a diverse array of stimuli. One of this ion channel's least discussed functions is in relation to the generation and maintenance of certain pain sensations. However, in the two decades which have elapsed since the identification of this ion channel, considerable data has emerged concerning its function in mediating pain sensations. TRPV4 is a mediator of mechanical hyperalgesia in the various contexts in which a mechanical stimulus, comprising trauma (at the macro-level) or discrete extracellular pressure or stress (at the micro-level), results in pain. TRPV4 is also recognised as constituting an essential component in mediating inflammatory pain. It also plays a role in relation to many forms of neuropathic-type pain, where it functions in mediating mechanical allodynia and hyperalgesia.Here, we review the role of TRPV4 in mediating pain sensations.
Collapse
|
33
|
Duitama M, Moreno Y, Santander SP, Casas Z, Sutachan JJ, Torres YP, Albarracín SL. TRP Channels as Molecular Targets to Relieve Cancer Pain. Biomolecules 2021; 12:1. [PMID: 35053150 PMCID: PMC8774023 DOI: 10.3390/biom12010001] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 11/17/2021] [Accepted: 11/23/2021] [Indexed: 12/13/2022] Open
Abstract
Transient receptor potential (TRP) channels are critical receptors in the transduction of nociceptive stimuli. The microenvironment of diverse types of cancer releases substances, including growth factors, neurotransmitters, and inflammatory mediators, which modulate the activity of TRPs through the regulation of intracellular signaling pathways. The modulation of TRP channels is associated with the peripheral sensitization observed in patients with cancer, which results in mild noxious sensory stimuli being perceived as hyperalgesia and allodynia. Secondary metabolites derived from plant extracts can induce the activation, blocking, and desensitization of TRP channels. Thus, these compounds could act as potential therapeutic agents, as their antinociceptive properties could be beneficial in relieving cancer-derived pain. In this review, we will summarize the role of TRPV1 and TRPA1 in pain associated with cancer and discuss molecules that have been reported to modulate these channels, focusing particularly on the mechanisms of channel activation associated with molecules released in the tumor microenvironment.
Collapse
Affiliation(s)
- Milena Duitama
- Departamento de Nutrición y Bioquímica, Pontificia Universidad Javeriana, Bogotá 110231, Colombia; (M.D.); (Z.C.); (J.J.S.)
| | - Yurany Moreno
- Department of Lymphoma & Myeloma, MD Anderson Cancer Center, The University of Texas, Houston, TX 77030, USA;
| | - Sandra Paola Santander
- Phytoimmunomodulation Research Group, Juan N. Corpas University Foundation, Bogotá 111111, Colombia;
| | - Zulma Casas
- Departamento de Nutrición y Bioquímica, Pontificia Universidad Javeriana, Bogotá 110231, Colombia; (M.D.); (Z.C.); (J.J.S.)
| | - Jhon Jairo Sutachan
- Departamento de Nutrición y Bioquímica, Pontificia Universidad Javeriana, Bogotá 110231, Colombia; (M.D.); (Z.C.); (J.J.S.)
| | - Yolima P. Torres
- Departamento de Nutrición y Bioquímica, Pontificia Universidad Javeriana, Bogotá 110231, Colombia; (M.D.); (Z.C.); (J.J.S.)
| | - Sonia L. Albarracín
- Departamento de Nutrición y Bioquímica, Pontificia Universidad Javeriana, Bogotá 110231, Colombia; (M.D.); (Z.C.); (J.J.S.)
| |
Collapse
|
34
|
Comes N, Gasull X, Callejo G. Proton Sensing on the Ocular Surface: Implications in Eye Pain. Front Pharmacol 2021; 12:773871. [PMID: 34899333 PMCID: PMC8652213 DOI: 10.3389/fphar.2021.773871] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 11/09/2021] [Indexed: 01/15/2023] Open
Abstract
Protons reaching the eyeball from exogenous acidic substances or released from damaged cells during inflammation, immune cells, after tissue injury or during chronic ophthalmic conditions, activate or modulate ion channels present in sensory nerve fibers that innervate the ocular anterior surface. Their identification as well as their role during disease is critical for the understanding of sensory ocular pathophysiology. They are likely to mediate some of the discomfort sensations accompanying several ophthalmic formulations and may represent novel targets for the development of new therapeutics for ocular pathologies. Among the ion channels expressed in trigeminal nociceptors innervating the anterior surface of the eye (cornea and conjunctiva) and annex ocular structures (eyelids), members of the TRP and ASIC families play a critical role in ocular acidic pain. Low pH (pH 6) activates TRPV1, a polymodal ion channel also activated by heat, capsaicin and hyperosmolar conditions. ASIC1, ASIC3 and heteromeric ASIC1/ASIC3 channels present in ocular nerve terminals are activated at pH 7.2–6.5, inducing pain by moderate acidifications of the ocular surface. These channels, together with TRPA1, are involved in acute ocular pain, as well as in painful sensations during allergic keratoconjunctivitis or other ophthalmic conditions, as blocking or reducing channel expression ameliorates ocular pain. TRPV1, TRPA1 and other ion channels are also present in corneal and conjunctival cells, promoting inflammation of the ocular surface after injury. In addition to the above-mentioned ion channels, members of the K2P and P2X ion channel families are also expressed in trigeminal neurons, however, their role in ocular pain remains unclear to date. In this report, these and other ion channels and receptors involved in acid sensing during ocular pathologies and pain are reviewed.
Collapse
Affiliation(s)
- Núria Comes
- Neurophysiology Laboratory, Department of Biomedicine, Medical School, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Xavier Gasull
- Neurophysiology Laboratory, Department of Biomedicine, Medical School, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Gerard Callejo
- Neurophysiology Laboratory, Department of Biomedicine, Medical School, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| |
Collapse
|
35
|
Trypsin induces an aversive response in zebrafish by PAR2 activation in keratinocytes. PLoS One 2021; 16:e0257774. [PMID: 34624042 PMCID: PMC8500423 DOI: 10.1371/journal.pone.0257774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 09/09/2021] [Indexed: 11/19/2022] Open
Abstract
Previously we have shown that trypsin, a protein typically involved in digestion, is released from gills of both fresh and saltwater fishes into surrounding water under stress or injury. We have also shown that each species produces trypsin with different specific activities. In this report, using zebrafish as a model, we identified that trypsin induces an aversive response in zebrafish larvae and adult zebrafish. Since Protease-Activated Receptor 2 (PAR2) responds to trypsin, we tested whether the aversive response is dependent on the activation of PAR2 located on the zebrafish skin cells. Zebrafish larvae treated separately with neomycin and zinc sulfate also showed aversive response indicating neuromast, and olfactory cells are not involved in this aversion. Cultured keratinocytes from zebrafish showed a response to trypsin. Zebrafish larvae subjected to knockdown of par2a also exhibited reduced escape response. Similarly, par2a-deficient mutant larvae displayed no response to trypsin. Since it has been shown that stress activates PAR2 and sends signals to the brain as shown by the increased c-fos expression, we tested c-fos expression in adult zebrafish brains after trypsin treatment of adults and found enhanced c-fos expression by qRT-PCR. Taken together, our results show that the trypsin activates PAR2 on keratinocytes signaling the brain, and this pathway of trypsin-induced escape response will provide a unique communication mechanism in zebrafish. Furthermore, since PAR2 activation also occurs in pain/pruritus sensing, this model might be useful in elucidating components of signaling pathways in pain/pruritus.
Collapse
|
36
|
Macrophage as a Peripheral Pain Regulator. Cells 2021; 10:cells10081881. [PMID: 34440650 PMCID: PMC8392675 DOI: 10.3390/cells10081881] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/22/2021] [Accepted: 07/22/2021] [Indexed: 12/30/2022] Open
Abstract
A neuroimmune crosstalk is involved in somatic and visceral pathological pain including inflammatory and neuropathic components. Apart from microglia essential for spinal and supraspinal pain processing, the interaction of bone marrow-derived infiltrating macrophages and/or tissue-resident macrophages with the primary afferent neurons regulates pain signals in the peripheral tissue. Recent studies have uncovered previously unknown characteristics of tissue-resident macrophages, such as their origins and association with regulation of pain signals. Peripheral nerve macrophages and intestinal resident macrophages, in addition to adult monocyte-derived infiltrating macrophages, secrete a variety of mediators, such as tumor necrosis factor-α, interleukin (IL)-1β, IL-6, high mobility group box 1 and bone morphogenic protein 2 (BMP2), that regulate the excitability of the primary afferents. Neuron-derived mediators including neuropeptides, ATP and macrophage-colony stimulating factor regulate the activity or polarization of diverse macrophages. Thus, macrophages have multitasks in homeostatic conditions and participate in somatic and visceral pathological pain by interacting with neurons.
Collapse
|
37
|
Luostarinen S, Hämäläinen M, Hatano N, Muraki K, Moilanen E. The inflammatory regulation of TRPA1 expression in human A549 lung epithelial cells. Pulm Pharmacol Ther 2021; 70:102059. [PMID: 34302984 DOI: 10.1016/j.pupt.2021.102059] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 06/06/2021] [Accepted: 07/17/2021] [Indexed: 10/20/2022]
Abstract
Transient receptor potential ankyrin-1 (TRPA1) is an ion channel mediating pain and cough signals in sensory neurons. We and others have shown that TRPA1 is also expressed in some non-neuronal cells and supports inflammatory responses. To address the pathogenesis and to uncover potential targets for pharmacotherapy in inflammatory lung diseases, we set out to study the expression of TRPA1 in human A549 lung epithelial cells under inflammatory conditions. TRPA1 expression was determined by RT-qPCR and Western blotting at a mRNA and protein level, respectively and its function was studied by Fluo 3-AM intracellular Ca2+ measurement in A549 lung epithelial cells. TRPA1 promoter activity was assessed by reporter gene assay. TRPA1 expression was very low in A549 cells in the absence of inflammatory stimuli. Tumor necrosis factor-α (TNF-α) significantly increased TRPA1 expression and a synergy was found between TNF-α, interleukin-1β (IL-1β) and interferon-γ (IFN-γ). Reporter gene experiments indicate that the combination of TNF-α and IL-1β increases TRPA1 promoter activity while the effect of IFN-γ seems to be non-transcriptional. Interestingly, the glucocorticoid dexamethasone downregulated TRPA1 expression in A549 cells by reducing TRPA1 mRNA stability in a transcription-dependent manner. Furthermore, pharmacological blockade of TRPA1 reduced the production of the pro-inflammatory cytokine IL-8. In conclusion, TRPA1 was found to be expressed and functional in human A549 lung epithelial cells under inflammatory conditions. The anti-inflammatory steroid dexamethasone reduced TRPA1 expression through post-transcriptional mechanisms. The results reveal TRPA1 as a potential mediator and drug target in inflammatory lung conditions.
Collapse
Affiliation(s)
- Samu Luostarinen
- The Immunopharmacology Research Group, Faculty of Medicine and Health Technology, Tampere University and Tampere University Hospital, Tampere, Finland
| | - Mari Hämäläinen
- The Immunopharmacology Research Group, Faculty of Medicine and Health Technology, Tampere University and Tampere University Hospital, Tampere, Finland
| | - Noriyuki Hatano
- Laboratory of Cellular Pharmacology, School of Pharmacy, Aichi-Gakuin University, Nagoya, Japan
| | - Katsuhiko Muraki
- Laboratory of Cellular Pharmacology, School of Pharmacy, Aichi-Gakuin University, Nagoya, Japan
| | - Eeva Moilanen
- The Immunopharmacology Research Group, Faculty of Medicine and Health Technology, Tampere University and Tampere University Hospital, Tampere, Finland.
| |
Collapse
|
38
|
Common and discrete mechanisms underlying chronic pain and itch: peripheral and central sensitization. Pflugers Arch 2021; 473:1603-1615. [PMID: 34245379 DOI: 10.1007/s00424-021-02599-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 05/26/2021] [Accepted: 06/22/2021] [Indexed: 12/30/2022]
Abstract
Normally, an obvious antagonism exists between pain and itch. In normal conditions, painful stimuli suppress itch sensation, whereas pain killers often generate itch. Although pain and itch are mediated by separate pathways under normal conditions, most chemicals are not highly specific to one sensation in chronic pathologic conditions. Notably, in patients with neuropathic pain, histamine primarily induces pain rather than itch, while in patients with atopic dermatitis, bradykinin triggers itch rather than pain. Accordingly, repetitive scratching even enhances itch sensation in chronic itch conditions. Physicians often prescribe pain relievers to patients with chronic itch, suggesting common mechanisms underlying chronic pain and itch, especially peripheral and central sensitization. Rather than separating itch and pain, studies should investigate chronic itch and pain including neuropathic and inflammatory conditions. Here, we reviewed chronic sensitization leading to chronic pain and itch at both peripheral and central levels. Studies investigating the connection between pain and itch facilitate the development of new therapeutics against both chronic dysesthesias based on the underlying pathophysiology.
Collapse
|
39
|
Pierre O, Fouchard M, Le Goux N, Buscaglia P, Leschiera R, Lewis RJ, Mignen O, Fluhr JW, Misery L, Le Garrec R. Pacific-Ciguatoxin-2 and Brevetoxin-1 Induce the Sensitization of Sensory Receptors Mediating Pain and Pruritus in Sensory Neurons. Mar Drugs 2021; 19:387. [PMID: 34356812 PMCID: PMC8306505 DOI: 10.3390/md19070387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/25/2021] [Accepted: 06/26/2021] [Indexed: 11/24/2022] Open
Abstract
Ciguatera fish poisoning (CFP) and neurotoxic shellfish poisoning syndromes are induced by the consumption of seafood contaminated by ciguatoxins and brevetoxins. Both toxins cause sensory symptoms such as paresthesia, cold dysesthesia and painful disorders. An intense pruritus, which may become chronic, occurs also in CFP. No curative treatment is available and the pathophysiology is not fully elucidated. Here we conducted single-cell calcium video-imaging experiments in sensory neurons from newborn rats to study in vitro the ability of Pacific-ciguatoxin-2 (P-CTX-2) and brevetoxin-1 (PbTx-1) to sensitize receptors and ion channels, (i.e., to increase the percentage of responding cells and/or the response amplitude to their pharmacological agonists). In addition, we studied the neurotrophin release in sensory neurons co-cultured with keratinocytes after exposure to P-CTX-2. Our results show that P-CTX-2 induced the sensitization of TRPA1, TRPV4, PAR2, MrgprC, MrgprA and TTX-r NaV channels in sensory neurons. P-CTX-2 increased the release of nerve growth factor and brain-derived neurotrophic factor in the co-culture supernatant, suggesting that those neurotrophins could contribute to the sensitization of the aforementioned receptors and channels. Our results suggest the potential role of sensitization of sensory receptors/ion channels in the induction or persistence of sensory disturbances in CFP syndrome.
Collapse
Affiliation(s)
- Ophélie Pierre
- Laboratoire Interactions Epithéliums-Neurones (LIEN), University of Brest, EA4685, F-29200 Brest, France; (M.F.); (R.L.); (J.W.F.); (L.M.); (R.L.G.)
| | - Maxime Fouchard
- Laboratoire Interactions Epithéliums-Neurones (LIEN), University of Brest, EA4685, F-29200 Brest, France; (M.F.); (R.L.); (J.W.F.); (L.M.); (R.L.G.)
- Department of Dermatology, University Hospital of Brest, F-29200 Brest, France
| | - Nelig Le Goux
- Lymphocytes B et Autoimmunité, Faculty of Medicine and Health Sciences, University of Brest, Inserm, UMR1227, F-29200 Brest, France; (N.L.G.); (P.B.); (O.M.)
| | - Paul Buscaglia
- Lymphocytes B et Autoimmunité, Faculty of Medicine and Health Sciences, University of Brest, Inserm, UMR1227, F-29200 Brest, France; (N.L.G.); (P.B.); (O.M.)
- Department of Molecular Physiology and Biophysics, Fraternal Order of Eagle Diabetes Research Center, Iowa Neuroscience Institute, Pappajohn Biomedical Institute, University of Iowa, Iowa City, IA 52242, USA
| | - Raphaël Leschiera
- Laboratoire Interactions Epithéliums-Neurones (LIEN), University of Brest, EA4685, F-29200 Brest, France; (M.F.); (R.L.); (J.W.F.); (L.M.); (R.L.G.)
| | - Richard J. Lewis
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia;
| | - Olivier Mignen
- Lymphocytes B et Autoimmunité, Faculty of Medicine and Health Sciences, University of Brest, Inserm, UMR1227, F-29200 Brest, France; (N.L.G.); (P.B.); (O.M.)
| | - Joachim W. Fluhr
- Laboratoire Interactions Epithéliums-Neurones (LIEN), University of Brest, EA4685, F-29200 Brest, France; (M.F.); (R.L.); (J.W.F.); (L.M.); (R.L.G.)
- Department of Dermatology, University Hospital of Brest, F-29200 Brest, France
- Department of Dermatology and Allergology, Universitaetsmedizin Charité Berlin, D-10117 Berlin, Germany
| | - Laurent Misery
- Laboratoire Interactions Epithéliums-Neurones (LIEN), University of Brest, EA4685, F-29200 Brest, France; (M.F.); (R.L.); (J.W.F.); (L.M.); (R.L.G.)
- Department of Dermatology, University Hospital of Brest, F-29200 Brest, France
| | - Raphaële Le Garrec
- Laboratoire Interactions Epithéliums-Neurones (LIEN), University of Brest, EA4685, F-29200 Brest, France; (M.F.); (R.L.); (J.W.F.); (L.M.); (R.L.G.)
| |
Collapse
|
40
|
Meng J, Li Y, Fischer MJM, Steinhoff M, Chen W, Wang J. Th2 Modulation of Transient Receptor Potential Channels: An Unmet Therapeutic Intervention for Atopic Dermatitis. Front Immunol 2021; 12:696784. [PMID: 34276687 PMCID: PMC8278285 DOI: 10.3389/fimmu.2021.696784] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 06/15/2021] [Indexed: 12/12/2022] Open
Abstract
Atopic dermatitis (AD) is a multifaceted, chronic relapsing inflammatory skin disease that affects people of all ages. It is characterized by chronic eczema, constant pruritus, and severe discomfort. AD often progresses from mild annoyance to intractable pruritic inflammatory lesions associated with exacerbated skin sensitivity. The T helper-2 (Th2) response is mainly linked to the acute and subacute phase, whereas Th1 response has been associated in addition with the chronic phase. IL-17, IL-22, TSLP, and IL-31 also play a role in AD. Transient receptor potential (TRP) cation channels play a significant role in neuroinflammation, itch and pain, indicating neuroimmune circuits in AD. However, the Th2-driven cutaneous sensitization of TRP channels is underappreciated. Emerging findings suggest that critical Th2-related cytokines cause potentiation of TRP channels, thereby exaggerating inflammation and itch sensation. Evidence involves the following: (i) IL-13 enhances TRPV1 and TRPA1 transcription levels; (ii) IL-31 sensitizes TRPV1 via transcriptional and channel modulation, and indirectly modulates TRPV3 in keratinocytes; (iii) The Th2-cytokine TSLP increases TRPA1 synthesis in sensory neurons. These changes could be further enhanced by other Th2 cytokines, including IL-4, IL-25, and IL-33, which are inducers for IL-13, IL-31, or TSLP in skin. Taken together, this review highlights that Th2 cytokines potentiate TRP channels through diverse mechanisms under different inflammatory and pruritic conditions, and link this effect to distinct signaling cascades in AD. This review strengthens the notion that interrupting Th2-driven modulation of TRP channels will inhibit transition from acute to chronic AD, thereby aiding the development of effective therapeutics and treatment optimization.
Collapse
Affiliation(s)
- Jianghui Meng
- School of Life Sciences, Henan University, Kaifeng, China.,National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland
| | - Yanqing Li
- School of Life Sciences, Henan University, Kaifeng, China
| | - Michael J M Fischer
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Martin Steinhoff
- Department of Dermatology and Venereology, Hamad Medical Corporation, Doha, Qatar.,Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar.,Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar.,Department of Dermatology, Weill Cornell Medicine-Qatar, Doha, Qatar.,Qatar University, College of Medicine, Doha, Qatar.,Department of Dermatology, Weill Cornell Medicine, New York, NY, United States
| | - Weiwei Chen
- School of Life Sciences, Henan University, Kaifeng, China
| | - Jiafu Wang
- School of Life Sciences, Henan University, Kaifeng, China.,School of Biotechnology, Faculty of Science and Health, Dublin City University, Dublin, Ireland
| |
Collapse
|
41
|
Mai L, Liu Q, Huang F, He H, Fan W. Involvement of Mast Cells in the Pathophysiology of Pain. Front Cell Neurosci 2021; 15:665066. [PMID: 34177465 PMCID: PMC8222580 DOI: 10.3389/fncel.2021.665066] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 05/07/2021] [Indexed: 12/11/2022] Open
Abstract
Mast cells (MCs) are immune cells and are widely distributed throughout the body. MCs are not only classically viewed as effector cells of some allergic diseases but also participate in host defense, innate and acquired immunity, homeostatic responses, and immunoregulation. Mounting evidence indicates that activation of MCs releasing numerous vasoactive and inflammatory mediators has effects on the nervous system and has been involved in different pain conditions. Here, we review the latest advances made about the implication of MCs in pain. Possible cellular and molecular mechanisms regarding the crosstalk between MC and the nervous system in the initiation and maintenance of pain are also discussed.
Collapse
Affiliation(s)
- Lijia Mai
- Department of Anesthesiology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, China
| | - Qing Liu
- Department of Anesthesiology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, China
| | - Fang Huang
- Guangdong Provincial Key Laboratory of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, China
| | - Hongwen He
- Guangdong Provincial Key Laboratory of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, China
| | - Wenguo Fan
- Department of Anesthesiology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
42
|
Eller OC, Yang X, Fuentes IM, Pierce AN, Jones BM, Brake AD, Wang R, Dussor G, Christianson JA. Voluntary Wheel Running Partially Attenuates Early Life Stress-Induced Neuroimmune Measures in the Dura and Evoked Migraine-Like Behaviors in Female Mice. Front Physiol 2021; 12:665732. [PMID: 34122137 PMCID: PMC8194283 DOI: 10.3389/fphys.2021.665732] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/29/2021] [Indexed: 12/13/2022] Open
Abstract
Migraine is a complex neurological disorder that affects three times more women than men and can be triggered by endogenous and exogenous factors. Stress is a common migraine trigger and exposure to early life stress increases the likelihood of developing chronic pain disorders later in life. Here, we used our neonatal maternal separation (NMS) model of early life stress to investigate whether female NMS mice have an increased susceptibility to evoked migraine-like behaviors and the potential therapeutic effect of voluntary wheel running. NMS was performed for 3 h/day during the first 3 weeks of life and initial observations were made at 12 weeks of age after voluntary wheel running (Exercise, -Ex) or sedentary behavior (-Sed) for 4 weeks. Mast cell degranulation rates were significantly higher in dura mater from NMS-Sed mice, compared to either naïve-Sed or NMS-Ex mice. Protease activated receptor 2 (PAR2) protein levels in the dura were significantly increased in NMS mice and a significant interaction of NMS and exercise was observed for transient receptor potential ankyrin 1 (TRPA1) protein levels in the dura. Behavioral assessments were performed on adult (>8 weeks of age) naïve and NMS mice that received free access to a running wheel beginning at 4 weeks of age. Facial grimace, paw mechanical withdrawal threshold, and light aversion were measured following direct application of inflammatory soup (IS) onto the dura or intraperitoneal (IP) nitroglycerin (NTG) injection. Dural IS resulted in a significant decrease in forepaw withdrawal threshold in all groups of mice, while exercise significantly increased grimace score across all groups. NTG significantly increased grimace score, particularly in exercised mice. A significant effect of NMS and a significant interaction effect of exercise and NMS were observed on hindpaw sensitivity following NTG injection. Significant light aversion was observed in NMS mice, regardless of exercise, following NTG. Finally, exercise significantly reduced calcitonin gene-related peptide (CGRP) protein level in the dura of NMS and naïve mice. Taken together, these findings suggest that while voluntary wheel running improved some measures in NMS mice that have been associated with increased migraine susceptibility, behavioral outcomes were not impacted or even worsened by exercise.
Collapse
Affiliation(s)
- Olivia C. Eller
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Xiaofang Yang
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Isabella M. Fuentes
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Angela N. Pierce
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, United States
- Department of Physiology, Kansas City University of Medicine and Biosciences, Joplin, MO, United States
| | - Brittni M. Jones
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Aaron D. Brake
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Ruipeng Wang
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Gregory Dussor
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Dallas, TX, United States
| | - Julie A. Christianson
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, United States
- Department of Anesthesiology, University of Kansas Medical Center, Kansas City, KS, United States
| |
Collapse
|
43
|
McMillan H, Lundy FT, Dunne OM, Al-Natour B, Jeanneau C, About I, Curtis TM, El Karim I. Endogenous Mas-related G-protein-coupled receptor X1 activates and sensitizes TRPA1 in a human model of peripheral nerves. FASEB J 2021; 35:e21492. [PMID: 33788969 DOI: 10.1096/fj.202001667rr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 02/16/2021] [Accepted: 02/16/2021] [Indexed: 12/21/2022]
Abstract
Mas-related G-protein-coupled receptor X1 (MrgprX1) is a human-specific Mrgpr and its expression is restricted to primary sensory neurons. However, its role in nociception and pain signaling pathways is largely unknown. This study aims to investigate a role for MrgprX1 in nociception via interaction with the pain receptor, Transient Receptor Potential Ankyrin 1 (TRPA1), using in-vitro and in-vivo human neuronal models. MrgprX1 protein expression in human trigeminal nociceptors was investigated by the immunolabeling of the dental pulp and cultured peripheral neuronal equivalent (PNE) cells. MrgprX1 receptor signaling was monitored by Fura-2-based Ca2+ imaging using PNEs and membrane potential responses were measured using FluoVoltTM . Immunofluorescent staining revealed MrgprX1 expression in-vivo in dental afferents, which was more intense in inflamed compared to healthy dental pulps. Endogenous MrgprX1 protein expression was confirmed in the in-vitro human PNE model. MrgprX1 receptor signaling and the mechanisms through which it couples to TRPA1 were studied by Ca2+ imaging. Results showed that MrgprX1 activates TRPA1 and induces membrane depolarization in a TRPA1 dependent manner. In addition, MrgprX1 sensitizes TRPA1 to agonist stimulation via Protein Kinase C (PKC). The activation and sensitization of TRPA1 by MrgprX1 in a model of human nerves suggests an important role for this receptor in the modulation of nociception.
Collapse
Affiliation(s)
- Hayley McMillan
- School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, UK
| | - Fionnuala T Lundy
- School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, UK
| | - Orla M Dunne
- School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, UK
| | - Banan Al-Natour
- School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, UK
- Department of Oral Medicine and Oral Surgery, Faculty of Dentistry, Jordan University of Science and Technology, Irbid, Jordan
| | | | - Imad About
- Aix Marseille Univ, CNRS, ISM, Inst Movement Sci, Marseille, France
| | - Tim M Curtis
- School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, UK
| | - Ikhlas El Karim
- School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, UK
| |
Collapse
|
44
|
Takahashi S, Kurogi M, Saitoh O. The diversity in sensitivity of TRPA1 and TRPV1 of various animals to polyphenols. Biomed Res 2021; 42:43-51. [PMID: 33840685 DOI: 10.2220/biomedres.42.43] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The perception of tastes is sensed by the receptors that stimulate sensory cells. We previously reported that TRPA1 and TRPV1 channels expressed in the oral cavity of mammals, are activated by the auto-oxidized product of epigallocatechin gallate (oxiEGCG), a major astringent catechin in green tea. Here, we investigated and compared the sensitivity of TRPA1 and TRPV1 from various animals to astringent polyphenols. We selected three polyphenols, oxiEGCG, tannic acid and myricetin. HEK293T cells expressing TRPA1 or TRPV1 from mammal, bird, reptile, amphibian, and fish, were analyzed for their activation by the Ca2+-imaging. We found the apparent diversity in the polyphenol-sensitivity among various animals. Mammalian TRPs showed relatively higher sensitivity to polyphenols, and especially, human TRPA1 and TRPV1 could be activated by all of three polyphenols at 20 μM. Reptile TRP channels, however, were insensitive to any polyphenols examined. Moreover, the polyphenol-sensitivity of zebrafish TRPA1 and TRPV1 was quite different from that of medaka TRP channels. Since many polyphenols are present in plants and the sensing of polyphenols using TRP channels in the oral cavity might cause astringent taste, the observed diversity of the polyphenol-sensitivity of TRP channels might be involved in the divergence in the food habit of various animals.
Collapse
Affiliation(s)
- Sayuri Takahashi
- Department of Bio-Science, Faculty of Bio-Science, Nagahama Institute of Bio-Science and Technology
| | - Mako Kurogi
- Department of Bio-Science, Faculty of Bio-Science, Nagahama Institute of Bio-Science and Technology
| | - Osamu Saitoh
- Department of Bio-Science, Faculty of Bio-Science, Nagahama Institute of Bio-Science and Technology
| |
Collapse
|
45
|
Luostarinen S, Hämäläinen M, Moilanen E. Transient Receptor Potential Ankyrin 1 (TRPA1)-An Inflammation-Induced Factor in Human HaCaT Keratinocytes. Int J Mol Sci 2021; 22:ijms22073322. [PMID: 33805042 PMCID: PMC8037497 DOI: 10.3390/ijms22073322] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/10/2021] [Accepted: 03/17/2021] [Indexed: 12/13/2022] Open
Abstract
Transient receptor potential ankyrin 1 (TRPA1) is an ion channel mainly studied in sensory neurons where it mediates itch, pain and neurogenic inflammation. Recently, some nonneuronal cells have also been shown to express TRPA1 to support inflammatory responses. To address the role of TRPA1 in skin inflammation, we aimed to investigate TRPA1 expression in keratinocytes. HaCaT cells (a model of human keratinocytes) and skin biopses from wild-type and TRPA1 deficient mice were used in the studies. TRPA1 expression in nonstimulated keratinocytes was very low but significantly inducible by the proinflammatory cytokine tumor necrosis factor (TNF) in an nuclear factor kappa B (NF-κB), and mitogen-activated protein (MAP) kinase (p38 and c-Jun N-terminal kinase, JNK)-dependent manner. Interestingly, drugs widely used to treat skin inflammation, the calcineurin inhibitors tacrolimus and cyclosporine and the glucocorticoid dexamethasone, significantly decreased TRPA1 expression. Furthermore, pharmacological inhibition and genetic deletion of TRPA1 reduced the synthesis of TNF-induced monocyte chemoattractant protein 1 (MCP-1) in keratinocytes and mouse skin biopsies. In conclusion, these findings point to an inflammatory role for TRPA1 in keratinocytes and present TRPA1 as a potential drug target in inflammatory skin diseases.
Collapse
|
46
|
Oultram JMJ, Pegler JL, Bowser TA, Ney LJ, Eamens AL, Grof CPL. Cannabis sativa: Interdisciplinary Strategies and Avenues for Medical and Commercial Progression Outside of CBD and THC. Biomedicines 2021; 9:biomedicines9030234. [PMID: 33652704 PMCID: PMC7996784 DOI: 10.3390/biomedicines9030234] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 02/16/2021] [Accepted: 02/23/2021] [Indexed: 12/11/2022] Open
Abstract
Cannabis sativa (Cannabis) is one of the world’s most well-known, yet maligned plant species. However, significant recent research is starting to unveil the potential of Cannabis to produce secondary compounds that may offer a suite of medical benefits, elevating this unique plant species from its illicit narcotic status into a genuine biopharmaceutical. This review summarises the lengthy history of Cannabis and details the molecular pathways that underpin the production of key secondary metabolites that may confer medical efficacy. We also provide an up-to-date summary of the molecular targets and potential of the relatively unknown minor compounds offered by the Cannabis plant. Furthermore, we detail the recent advances in plant science, as well as synthetic biology, and the pharmacology surrounding Cannabis. Given the relative infancy of Cannabis research, we go on to highlight the parallels to previous research conducted in another medically relevant and versatile plant, Papaver somniferum (opium poppy), as an indicator of the possible future direction of Cannabis plant biology. Overall, this review highlights the future directions of cannabis research outside of the medical biology aspects of its well-characterised constituents and explores additional avenues for the potential improvement of the medical potential of the Cannabis plant.
Collapse
Affiliation(s)
- Jackson M. J. Oultram
- Centre for Plant Science, University of Newcastle, University Drive, Callaghan, NSW 2308, Australia; (J.M.J.O.); (J.L.P.); (A.L.E.)
| | - Joseph L. Pegler
- Centre for Plant Science, University of Newcastle, University Drive, Callaghan, NSW 2308, Australia; (J.M.J.O.); (J.L.P.); (A.L.E.)
| | - Timothy A. Bowser
- CannaPacific Pty Ltd., 109 Ocean Street, Dudley, NSW 2290, Australia;
| | - Luke J. Ney
- School of Psychological Sciences, University of Tasmania, Hobart, TAS 7005, Australia;
| | - Andrew L. Eamens
- Centre for Plant Science, University of Newcastle, University Drive, Callaghan, NSW 2308, Australia; (J.M.J.O.); (J.L.P.); (A.L.E.)
| | - Christopher P. L. Grof
- Centre for Plant Science, University of Newcastle, University Drive, Callaghan, NSW 2308, Australia; (J.M.J.O.); (J.L.P.); (A.L.E.)
- CannaPacific Pty Ltd., 109 Ocean Street, Dudley, NSW 2290, Australia;
- Correspondence: ; Tel.: +612-4921-5858
| |
Collapse
|
47
|
van den Berg MPM, Nijboer-Brinksma S, Bos IST, van den Berge M, Lamb D, van Faassen M, Kema IP, Gosens R, Kistemaker LEM. The novel TRPA1 antagonist BI01305834 inhibits ovalbumin-induced bronchoconstriction in guinea pigs. Respir Res 2021; 22:48. [PMID: 33557843 PMCID: PMC7871391 DOI: 10.1186/s12931-021-01638-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 01/25/2021] [Indexed: 01/05/2023] Open
Abstract
Background Asthma is a chronic respiratory disease in which the nervous system plays a central role. Sensory nerve activation, amongst others via Transient Receptor Potential Ankyrin 1 (TRPA1) channels, contributes to asthma characteristics including cough, bronchoconstriction, mucus secretion, airway hyperresponsiveness (AHR) and inflammation. In the current study, we evaluated the efficacy of the novel TRPA1 antagonist BI01305834 against AHR and inflammation in guinea-pig models of asthma. Methods First, a pilot study was performed in a guinea-pig model of allergic asthma to find the optimal dose of BI01305834. Next, the effect of BI01305834 on (1) AHR to inhaled histamine after the early and late asthmatic reaction (EAR and LAR), (2) magnitude of EAR and LAR and (3) airway inflammation was assessed. Precision-cut lung slices and trachea strips were used to investigate the bronchoprotective and bronchodilating-effect of BI01305834. Statistical evaluation of differences of in vivo data was performed using a Mann–Whitney U test or One-way nonparametric Kruskal–Wallis ANOVA, for ex vivo data One- or Two-way ANOVA was used, all with Dunnett’s post-hoc test where appropriate. Results A dose of 1 mg/kg BI01305834 was selected based on AHR and exposure data in blood samples from the pilot study. In the subsequent study, 1 mg/kg BI01305834 inhibited AHR after the EAR, and the development of EAR and LAR elicited by ovalbumin in ovalbumin-sensitized guinea pigs. BI01305834 did not inhibit allergen-induced total and differential cells in the lavage fluid and interleukin-13 gene expression in lung homogenates. Furthermore, BI01305834 was able to inhibit allergen and histamine-induced airway narrowing in guinea-pig lung slices, without affecting histamine release, and reverse allergen-induced bronchoconstriction in guinea-pig trachea strips. Conclusions TRPA1 inhibition protects against AHR and the EAR and LAR in vivo and allergen and histamine-induced airway narrowing ex vivo, and reverses allergen-induced bronchoconstriction independently of inflammation. This effect was partially dependent upon histamine, suggesting a neuronal and possible non-neuronal role for TRPA1 in allergen-induced bronchoconstriction.
Collapse
Affiliation(s)
- Mariska P M van den Berg
- Department of Molecular Pharmacology, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands.,Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Susan Nijboer-Brinksma
- Department of Molecular Pharmacology, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands.,Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - I Sophie T Bos
- Department of Molecular Pharmacology, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands.,Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Maarten van den Berge
- Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Department of Pulmonary Diseases, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - David Lamb
- Immunology + Respiratory, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Martijn van Faassen
- Department of Laboratory Medicine, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Ido P Kema
- Department of Laboratory Medicine, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Reinoud Gosens
- Department of Molecular Pharmacology, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands.,Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Loes E M Kistemaker
- Department of Molecular Pharmacology, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands. .,Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| |
Collapse
|
48
|
Yamaki S, Chau A, Gonzales L, McKemy DD. Nociceptive afferent phenotyping reveals that transient receptor potential ankyrin 1 promotes cold pain through neurogenic inflammation upstream of the neurotrophic factor receptor GFRα3 and the menthol receptor transient receptor potential melastatin 8. Pain 2021; 162:609-618. [PMID: 32826761 PMCID: PMC8312403 DOI: 10.1097/j.pain.0000000000002043] [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: 06/03/2020] [Accepted: 07/17/2020] [Indexed: 12/14/2022]
Abstract
ABSTRACT The proper detection and behavioral response to painfully cold temperatures is critical for avoiding potentially harmful tissue damage. Cold allodynia and hyperalgesia, pain associated with innocuous cooling and exaggerated pain with noxious cold, respectively, are common in patients with chronic pain. In peripheral somatosensory afferents, the ion channels transient receptor potential melastatin 8 (TRPM8) and transient receptor potential ankyrin 1 (TRPA1) are candidate receptors for innocuous and noxious cold temperatures, respectively. However, the role of TRPA1 as a cold sensor has remained controversial, and recent evidence suggests that TRPM8 channels and afferents mediate the detection of both pleasant and painful cold. To determine the role of TRPA1 afferents in cold-induced mouse behaviors in vivo, we used functional phenotyping by targeted nerve conduction block with the cell-impermeant lidocaine derivative QX-314. Surprisingly, we find that injection of QX-314 with TRPA1 agonists reduces cold-induced behaviors in mice, but does so in a TRPM8-dependent manner. Moreover, this effect is sexually dimorphic and requires the glial cell line-derived neurotrophic factor receptor GFRα3, as does cold hypersensitivity produced by the activation of TRPA1 channels. Taken together, these results suggest that under conditions of neurogenic inflammation, TRPA1 works upstream of GFRα3 and TRPM8 to produce cold hypersensitivity, providing novel insights into the role of TRPA1 channels in cold pain.
Collapse
Affiliation(s)
- Shanni Yamaki
- Molecular Biology Graduate Program; Department of Biological Sciences, University of Southern California, 3641 Watt Way / HNB 201, Los Angeles, CA 90089 U.S.A
- Neurobiology Section; Department of Biological Sciences, University of Southern California, 3641 Watt Way / HNB 201, Los Angeles, CA 90089 U.S.A
| | - Amanda Chau
- Neurobiology Section; Department of Biological Sciences, University of Southern California, 3641 Watt Way / HNB 201, Los Angeles, CA 90089 U.S.A
| | - Luigi Gonzales
- Neurobiology Section; Department of Biological Sciences, University of Southern California, 3641 Watt Way / HNB 201, Los Angeles, CA 90089 U.S.A
| | - David D. McKemy
- Molecular Biology Graduate Program; Department of Biological Sciences, University of Southern California, 3641 Watt Way / HNB 201, Los Angeles, CA 90089 U.S.A
- Neurobiology Section; Department of Biological Sciences, University of Southern California, 3641 Watt Way / HNB 201, Los Angeles, CA 90089 U.S.A
| |
Collapse
|
49
|
Spinal PAR2 Activation Contributes to Hypersensitivity Induced by Peripheral Inflammation in Rats. Int J Mol Sci 2021; 22:ijms22030991. [PMID: 33498178 PMCID: PMC7863954 DOI: 10.3390/ijms22030991] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/16/2021] [Accepted: 01/17/2021] [Indexed: 11/17/2022] Open
Abstract
The mechanisms of inflammatory pain need to be identified in order to find new superior treatments. Protease-activated receptors 2 (PAR2) and transient receptor potential vanilloid 1 (TRPV1) are highly co-expressed in dorsal root ganglion neurons and implicated in pain development. Here, we examined the role of spinal PAR2 in hyperalgesia and the modulation of synaptic transmission in carrageenan-induced peripheral inflammation, using intrathecal (i.t.) treatment in the behavioral experiments and recordings of spontaneous, miniature and dorsal root stimulation-evoked excitatory postsynaptic currents (sEPSCs, mEPSCs and eEPSCs) in spinal cord slices. Intrathecal PAR2-activating peptide (AP) administration aggravated the carrageenan-induced thermal hyperalgesia, and this was prevented by a TRPV1 antagonist (SB 366791) and staurosporine i.t. pretreatment. Additionally, the frequency of the mEPSC and sEPSC and the amplitude of the eEPSC recorded from the superficial dorsal horn neurons were enhanced after acute PAR2 AP application, while prevented with SB 366791 or staurosporine pretreatment. PAR2 antagonist application reduced the thermal hyperalgesia and decreased the frequency of mEPSC and sEPSC and the amplitude of eEPSC. Our findings highlight the contribution of spinal PAR2 activation to carrageenan-induced hyperalgesia and the importance of dorsal horn PAR2 and TRPV1 receptor interactions in the modulation of nociceptive synaptic transmission.
Collapse
|
50
|
Calmodulin Supports TRPA1 Channel Association with Opioid Receptors and Glutamate NMDA Receptors in the Nervous Tissue. Int J Mol Sci 2020; 22:ijms22010229. [PMID: 33379368 PMCID: PMC7795679 DOI: 10.3390/ijms22010229] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 12/21/2020] [Accepted: 12/24/2020] [Indexed: 12/24/2022] Open
Abstract
Transient receptor potential ankyrin member 1 (TRPA1) belongs to the family of thermo TRP cation channels that detect harmful temperatures, acids and numerous chemical pollutants. TRPA1 is expressed in nervous tissue, where it participates in the genesis of nociceptive signals in response to noxious stimuli and mediates mechanical hyperalgesia and allodynia associated with different neuropathies. The glutamate N-methyl-d-aspartate receptor (NMDAR), which plays a relevant role in allodynia to mechanical stimuli, is connected via histidine triad nucleotide-binding protein 1 (HINT1) and type 1 sigma receptor (σ1R) to mu-opioid receptors (MORs), which mediate the most potent pain relief. Notably, neuropathic pain causes a reduction in MOR antinociceptive efficacy, which can be reversed by blocking spinal NMDARs and TRPA1 channels. Thus, we studied whether TRPA1 channels form complexes with MORs and NMDARs that may be implicated in the aforementioned nociceptive signals. Our data suggest that TRPA1 channels functionally associate with MORs, delta opioid receptors and NMDARs in the dorsal root ganglia, the spinal cord and brain areas. These associations were altered in response to pharmacological interventions and the induction of inflammatory and also neuropathic pain. The MOR-TRPA1 and NMDAR-TRPA1 associations do not require HINT1 or σ1R but appear to be mediated by calcium-activated calmodulin. Thus, TRPA1 channels may associate with NMDARs to promote ascending acute and chronic pain signals and to control MOR antinociception.
Collapse
|