1
|
Gandini MA, Zamponi GW. Navigating the Controversies: Role of TRPM Channels in Pain States. Int J Mol Sci 2024; 25:10284. [PMID: 39408620 PMCID: PMC11476983 DOI: 10.3390/ijms251910284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/17/2024] [Accepted: 09/22/2024] [Indexed: 10/20/2024] Open
Abstract
Chronic pain is a debilitating condition that affects up to 1.5 billion people worldwide and bears a tremendous socioeconomic burden. The success of pain medicine relies on our understanding of the type of pain experienced by patients and the mechanisms that give rise to it. Ion channels are among the key targets for pharmacological intervention in chronic pain conditions. Therefore, it is important to understand how changes in channel properties, trafficking, and molecular interactions contribute to pain sensation. In this review, we discuss studies that have demonstrated the involvement of transient receptor potential M2, M3, and M8 channels in pain generation and transduction, as well as the controversies surrounding these findings.
Collapse
Affiliation(s)
- Maria A. Gandini
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada;
- Department of Clinical Neurosciences, Alberta Children’s Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Gerald W. Zamponi
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada;
- Department of Clinical Neurosciences, Alberta Children’s Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| |
Collapse
|
2
|
Hastings LE, Frye EV, Carlson ER, Chuong V, Matthews AN, Koob GF, Vendruscolo LF, Marchette RCN. Cold nociception as a measure of hyperalgesia during spontaneous heroin withdrawal in mice. Pharmacol Biochem Behav 2024; 235:173694. [PMID: 38128767 PMCID: PMC10842911 DOI: 10.1016/j.pbb.2023.173694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 12/13/2023] [Accepted: 12/14/2023] [Indexed: 12/23/2023]
Abstract
Opioids are powerful analgesic drugs that are used clinically to treat pain. However, chronic opioid use causes compensatory neuroadaptations that result in greater pain sensitivity during withdrawal, known as opioid withdrawal-induced hyperalgesia (OWIH). Cold nociception tests are commonly used in humans, but preclinical studies often use mechanical and heat stimuli to measure OWIH. Thus, further characterization of cold nociception stimuli is needed in preclinical models. We assessed three cold nociception tests-thermal gradient ring (5-30 °C, 5-50 °C, 15-40 °C, and 25-50 °C), dynamic cold plate (4 °C to -1 °C at -1 °C/min, -1 °C to 4 °C at +1 °C/min), and stable cold plate (10 °C, 6 °C, and 2 °C)-to measure hyperalgesia in a mouse protocol of heroin dependence. On the thermal gradient ring, mice in the heroin withdrawal group preferred warmer temperatures, and the results depended on the ring's temperature range. On the dynamic cold plate, heroin withdrawal increased the number of nociceptive responses, with a temperature ramp from 4 °C to -1 °C yielding the largest response. On the stable cold plate, heroin withdrawal increased the number of nociceptive responses, and a plate temperature of 2 °C yielded the most significant increase in responses. Among the three tests, the stable cold plate elicited the most robust change in behavior between heroin-dependent and nondependent mice and had the highest throughput. To pharmacologically characterize the stable cold plate test, we used μ-opioid and non-opioid receptor-targeting drugs that have been previously shown to reverse OWIH in mechanical and heat nociception assays. The full μ-opioid receptor agonist methadone and μ-opioid receptor partial agonist buprenorphine decreased OWIH, whereas the preferential μ-opioid receptor antagonist naltrexone increased OWIH. Two N-methyl-d-aspartate receptor antagonists (ketamine, MK-801), a corticotropin-releasing factor 1 receptor antagonist (R121919), a β2-adrenergic receptor antagonist (butoxamine), an α2-adrenergic receptor agonist (lofexidine), and a 5-hydroxytryptamine-3 receptor antagonist (ondansetron) had no effect on OWIH. These data demonstrate that the stable cold plate at 2 °C yields a robust, reliable, and concise measure of OWIH that is sensitive to opioid agonists.
Collapse
Affiliation(s)
- Lyndsay E Hastings
- Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
| | - Emma V Frye
- Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
| | - Erika R Carlson
- Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
| | - Vicky Chuong
- Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA; Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Intitute on Drug Abuse, Intramural Research Program, and National Institute on Alcohol Abuse and Alcoholism, Division of Intramural Clinical and Biological Research, Baltimore, MD, USA
| | - Aniah N Matthews
- Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
| | - George F Koob
- Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA
| | - Leandro F Vendruscolo
- Stress and Addiction Neuroscience Unit, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Intramural Research Program, and National Institute on Alcohol Abuse and Alcoholism, Division of Intramural Clinical and Biological Research, Baltimore, MD, USA
| | - Renata C N Marchette
- Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, USA.
| |
Collapse
|
3
|
Ruan Y, Jin X, Ji H, Zhu C, Yang Y, Zhou Y, Yu G, Wang C, Tang Z. Water extract of Notopterygium incisum alleviates cold allodynia in neuropathic pain by regulation of TRPA1. JOURNAL OF ETHNOPHARMACOLOGY 2023; 305:116065. [PMID: 36587876 DOI: 10.1016/j.jep.2022.116065] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 06/17/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Neuropathic pain can be debilitating and drastically affects the quality of life of those patients suffering from this condition. The Chinese herb Notopterygium incisum Ting ex H.T. Chang has long been used to disperse "cold". One under examined clinical feature of neuropathic pain is sensitivity to cold. Patients with neuropathic pain or arthritis usually describe a worsening of symptoms during the winter. AIMS OF THIS STUDY We proposed to test the hypothesis that Notopterygium incisum has a positive effect on the cold sensitivity found in neuropathic pain. MATERIALS AND METHODS In this study, we established chronic constriction injury (CCI) and cisplatin induced neuropathic pain mice models. Behavioral experiments and physiological examination methods were employed to investigate the effect of water extract of Notopterygium incisum (WN) on cold pain. RESULTS We found WN reduced cold pain and allyl isothiocyanate (AITC, Transient Receptor Potential A1 (TRPA1 agonist)) induced pain. WN inhibited AITC induced calcium response in HEK 293 cells transfected with TRPA1 and dorsal root ganglion (DRG) neurons. Moreover, we found that oral administration of WN reduced cold allodynia and mechanical allodynia caused by (CCI) and cisplatin induced neuropathic pain. We also observed that oral administration of WN decreased responses to AITC in DRG neurons as well as expression of TRPA1 in the WN treated neuropathic pain model. CONCLUSIONS The present study provide evidence that Notopterygium incisum alleviates cold allodynia in CCI and cisplatin induced neuropathic pain mouse models. WN alleviated neuropathic pain induced cold allodynia via directly modulating TRPA1. Our findings identify WN as a promising candidate for treating neuropathic pain that highlights a new mechanism of Notopterygium incisum on 'disperse cold'.
Collapse
Affiliation(s)
- Yonglan Ruan
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Xiang Jin
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Haiwang Ji
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Chan Zhu
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Yan Yang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Yuan Zhou
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Guang Yu
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Changming Wang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Zongxiang Tang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
4
|
Li Z, Zhang H, Wang Y, Li Y, Li Q, Zhang L. The distinctive role of menthol in pain and analgesia: Mechanisms, practices, and advances. Front Mol Neurosci 2022; 15:1006908. [PMID: 36277488 PMCID: PMC9580369 DOI: 10.3389/fnmol.2022.1006908] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 09/13/2022] [Indexed: 11/13/2022] Open
Abstract
Menthol is an important flavoring additive that triggers a cooling sensation. Under physiological condition, low to moderate concentrations of menthol activate transient receptor potential cation channel subfamily M member 8 (TRPM8) in the primary nociceptors, such as dorsal root ganglion (DRG) and trigeminal ganglion, generating a cooling sensation, whereas menthol at higher concentration could induce cold allodynia, and cold hyperalgesia mediated by TRPM8 sensitization. In addition, the paradoxical irritating properties of high concentrations of menthol is associated with its activation of transient receptor potential cation channel subfamily A member 1 (TRPA1). Under pathological situation, menthol activates TRPM8 to attenuate mechanical allodynia and thermal hyperalgesia following nerve injury or chemical stimuli. Recent reports have recapitulated the requirement of central group II/III metabotropic glutamate receptors (mGluR) with endogenous κ-opioid signaling pathways for menthol analgesia. Additionally, blockage of sodium channels and calcium influx is a determinant step after menthol exposure, suggesting the possibility of menthol for pain management. In this review, we will also discuss and summarize the advances in menthol-related drugs for pathological pain treatment in clinical trials, especially in neuropathic pain, musculoskeletal pain, cancer pain and postoperative pain, with the aim to find the promising therapeutic candidates for the resolution of pain to better manage patients with pain in clinics.
Collapse
Affiliation(s)
- Ziping Li
- The Graduate School, Tianjin Medical University, Tianjin, China
| | - Haoyue Zhang
- The Graduate School, Tianjin Medical University, Tianjin, China
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yigang Wang
- The Graduate School, Tianjin Medical University, Tianjin, China
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yize Li
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Qing Li
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
- Qing Li,
| | - Linlin Zhang
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
- *Correspondence: Linlin Zhang,
| |
Collapse
|
5
|
Petitjean H, Héberlé E, Hilfiger L, Łapieś O, Rodrigue G, Charlet A. TRP channels and monoterpenes: Past and current leads on analgesic properties. Front Mol Neurosci 2022; 15:945450. [PMID: 35966017 PMCID: PMC9373873 DOI: 10.3389/fnmol.2022.945450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
The activation of the transient receptor potential (TRP) channels expressed by sensory neurons is essential to the transduction of thermal and mechanical sensory information. In the setting of chronic inflammatory conditions, the activation of the melastatin family member 8 (TRPM8), the TRP vanilloid 1 (TRPV1), and the TRP ankyrin 1 (TRPA1) is correlated with pain hypersensitivity reactions. Monoterpenes, among which pulegone and menthol, a major class of phytocompounds present in essential oils of medicinal plants, are known modulators of those TRP channels activity. In the present review, we correlate the monoterpene content of plants with their historical therapeutic properties. We then describe how monoterpenes exert their anti-inflammatory and antihyperalgesia effects through modulation of TRP channels activity. Finally, we discuss the importance and the potential of characterizing new plant extracts and reassessing studied plant extracts for the development of ethnopharmacology-based innovative treatments for chronic pain. This review suggests that monoterpene solutions, based on composition from traditional healing herbs, offer an interesting avenue for the development of new phytotherapeutic treatments to alleviate chronic inflammatory pain conditions.
Collapse
Affiliation(s)
| | | | - Louis Hilfiger
- Benephyt, Strasbourg, France
- Centre National de la Recherche Scientifique, University of Strasbourg, Institute of Cellular and Integrative Neuroscience, INCI UPR3212, Strasbourg, France
| | - Olga Łapieś
- Centre National de la Recherche Scientifique, University of Strasbourg, Institute of Cellular and Integrative Neuroscience, INCI UPR3212, Strasbourg, France
| | | | - Alexandre Charlet
- Centre National de la Recherche Scientifique, University of Strasbourg, Institute of Cellular and Integrative Neuroscience, INCI UPR3212, Strasbourg, France
| |
Collapse
|
6
|
Martín-Escura C, Medina-Peris A, Spear LA, de la Torre Martínez R, Olivos-Oré LA, Barahona MV, González-Rodríguez S, Fernández-Ballester G, Fernández-Carvajal A, Artalejo AR, Ferrer-Montiel A, González-Muñiz R. β-Lactam TRPM8 Antagonist RGM8-51 Displays Antinociceptive Activity in Different Animal Models. Int J Mol Sci 2022; 23:ijms23052692. [PMID: 35269831 PMCID: PMC8910920 DOI: 10.3390/ijms23052692] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/18/2022] [Accepted: 02/22/2022] [Indexed: 02/05/2023] Open
Abstract
Transient receptor potential melastatin subtype 8 (TRPM8) is a cation channel extensively expressed in sensory neurons and implicated in different painful states. However, the effectiveness of TRPM8 modulators for pain relief is still a matter of discussion, since structurally diverse modulators lead to different results, depending on the animal pain model. In this work, we described the antinociceptive activity of a β–lactam derivative, RGM8-51, showing good TRPM8 antagonist activity, and selectivity against related thermoTRP channels and other pain-mediating receptors. In primary cultures of rat dorsal root ganglion (DRG) neurons, RGM8-51 potently reduced menthol-evoked neuronal firing without affecting the major ion conductances responsible for action potential generation. This compound has in vivo antinociceptive activity in response to cold, in a mouse model of oxaliplatin-induced peripheral neuropathy. In addition, it reduces cold, mechanical and heat hypersensitivity in a rat model of neuropathic pain arising after chronic constriction of the sciatic nerve. Furthermore, RGM8-51 exhibits mechanical hypersensitivity-relieving activity, in a mouse model of NTG-induced hyperesthesia. Taken together, these preclinical results substantiate that this TRPM8 antagonist is a promising pharmacological tool to study TRPM8-related diseases.
Collapse
Affiliation(s)
- Cristina Martín-Escura
- Instituto de Química Médica (IQM-CSIC), 28006 Madrid, Spain; (C.M.-E.); (L.A.S.)
- Alodia Farmacéutica SL, 28108 Alcobendas, Spain
| | - Alicia Medina-Peris
- IDiBE, Universidad Miguel Hernández, 03202 Elche, Spain; (A.M.-P.); (R.d.l.T.M.); (S.G.-R.); (G.F.-B.); (A.F.-M.)
| | - Luke A. Spear
- Instituto de Química Médica (IQM-CSIC), 28006 Madrid, Spain; (C.M.-E.); (L.A.S.)
| | - Roberto de la Torre Martínez
- IDiBE, Universidad Miguel Hernández, 03202 Elche, Spain; (A.M.-P.); (R.d.l.T.M.); (S.G.-R.); (G.F.-B.); (A.F.-M.)
| | - Luis A. Olivos-Oré
- Departamento de Farmacología y Toxicología, Facultad de Veterinaria, Universidad Complutense de Madrid, 28040 Madrid, Spain; (L.A.O.-O.); (M.V.B.); (A.R.A.)
- Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - María Victoria Barahona
- Departamento de Farmacología y Toxicología, Facultad de Veterinaria, Universidad Complutense de Madrid, 28040 Madrid, Spain; (L.A.O.-O.); (M.V.B.); (A.R.A.)
- Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Sara González-Rodríguez
- IDiBE, Universidad Miguel Hernández, 03202 Elche, Spain; (A.M.-P.); (R.d.l.T.M.); (S.G.-R.); (G.F.-B.); (A.F.-M.)
| | - Gregorio Fernández-Ballester
- IDiBE, Universidad Miguel Hernández, 03202 Elche, Spain; (A.M.-P.); (R.d.l.T.M.); (S.G.-R.); (G.F.-B.); (A.F.-M.)
| | - Asia Fernández-Carvajal
- IDiBE, Universidad Miguel Hernández, 03202 Elche, Spain; (A.M.-P.); (R.d.l.T.M.); (S.G.-R.); (G.F.-B.); (A.F.-M.)
- Correspondence: (A.F.-C.); (R.G.-M.); Tel.: +00-34-258-74-34 (R.G.-M.)
| | - Antonio R. Artalejo
- Departamento de Farmacología y Toxicología, Facultad de Veterinaria, Universidad Complutense de Madrid, 28040 Madrid, Spain; (L.A.O.-O.); (M.V.B.); (A.R.A.)
- Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Antonio Ferrer-Montiel
- IDiBE, Universidad Miguel Hernández, 03202 Elche, Spain; (A.M.-P.); (R.d.l.T.M.); (S.G.-R.); (G.F.-B.); (A.F.-M.)
| | - Rosario González-Muñiz
- Instituto de Química Médica (IQM-CSIC), 28006 Madrid, Spain; (C.M.-E.); (L.A.S.)
- Correspondence: (A.F.-C.); (R.G.-M.); Tel.: +00-34-258-74-34 (R.G.-M.)
| |
Collapse
|
7
|
Su L, Bai X, Niu T, Zhuang X, Dong B, Li Y, Yu Y, Wang G. P2Y1 Purinergic Receptor Contributes to Remifentanil-Induced Cold Hyperalgesia via Transient Receptor Potential Melastatin 8-Dependent Regulation of N-methyl-d-aspartate Receptor Phosphorylation in Dorsal Root Ganglion. Anesth Analg 2021; 133:794-810. [PMID: 34166321 DOI: 10.1213/ane.0000000000005617] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Remifentanil can induce postinfusion cold hyperalgesia. N-methyl-d-aspartate receptor (NMDAR) activation and upregulation of transient receptor potential melastatin 8 (TRPM8) membrane trafficking in dorsal root ganglion (DRG) are critical to cold hyperalgesia derived from neuropathic pain, and TRPM8 activation causes NMDAR-dependent cold response. Contribution of P2Y1 purinergic receptor (P2Y1R) activation in DRG to cold pain hypersensitivity and NMDAR activation induced by P2Y1R upregulation in neurons are also unraveled. This study explores whether P2Y1R contributes to remifentanil-induced cold hyperalgesia via TRPM8-dependent regulation of NMDAR phosphorylation in DRG. METHODS Rats with remifentanil-induced cold hyperalgesia were injected with TRPM8 antagonist or P2Y1R antagonist at 10 minutes before remifentanil infusion. Cold hyperalgesia (paw lift number and withdrawal duration on cold plate) was measured at -24, 2, 6, 24, and 48 hours following remifentanil infusion. After the last behavioral test, P2Y1R expression, TRPM8 expression and membrane trafficking, and NMDAR subunit (NR1 and NR2B) expression and phosphorylation in DRG were detected by western blot, and colocalization of P2Y1R with TRPM8 was determined by double-labeling immunofluorescence. Two-way repeated measures analysis of variance (ANOVA) or 2 × 2 factorial design ANOVA with repeated measures was used to analyze behavioral data of cold hyperalgesia. One-way ANOVA followed by Bonferroni post hoc comparisons was used to analyze the data in western blot and immunofluorescence. RESULTS Remifentanil infusion (1 μg·kg-1·min-1 for 60 minutes) induced cold hyperalgesia (hyperalgesia versus control, paw lift number and withdrawal duration on cold plate at 2-48 hours, P < .0001) with upregulated NR1 (hyperalgesia versus naive, 48 hours, mean ± standard deviation [SD], 114.00% ± 12.48% vs 41.75% ± 5.20%, P < .005) and NR2B subunits expression (104.13% ± 8.37% vs 24.63% ± 4.87%, P < .005), NR1 phosphorylation at Ser896 (91.88% ± 7.08% vs 52.00% ± 7.31%, P < .005) and NR2B phosphorylation at Tyr1472 (115.75% ± 8.68% vs 59.75% ± 7.78%, P < .005), TRPM8 expression (115.38% ± 9.27% vs 40.50% ± 4.07%, P < .005) and membrane trafficking (112.88% ± 5.62% vs 48.88% ± 6.49%, P < .005), and P2Y1R expression (128.25% ± 14.86% vs 45.13% ± 7.97%, P < .005) in DRG. Both TRPM8 and P2Y1R antagonists attenuated remifentanil-induced cold hyperalgesia and downregulated increased NR1 and NR2B expression and phosphorylation induced by remifentanil (remifentanil + RQ-00203078 versus remifentanil + saline, NR1 phosphorylation, 69.38% ± 3.66% vs 92.13% ± 4.85%; NR2B phosphorylation, 72.25% ± 6.43% vs 111.75% ± 11.00%, P < .0001). NMDAR activation abolished inhibition of TRPM8 and P2Y1R antagonists on remifentanil-induced cold hyperalgesia. P2Y1R antagonist inhibited remifentanil-evoked elevations in TRPM8 expression and membrane trafficking and P2Y1R-TRPM8 coexpression (remifentanil + 2'-deoxy-N6-methyl adenosine 3',5'-diphosphate [MRS2179] versus remifentanil + saline, coexpression, 8.33% ± 1.33% vs 22.19% ± 2.15%, P < .0001). CONCLUSIONS Attenuation of remifentanil-induced cold hyperalgesia by P2Y1R inhibition is attributed to downregulations in NMDAR expression and phosphorylation via diminishing TRPM8 expression and membrane trafficking in DRG.
Collapse
Affiliation(s)
- Lin Su
- From the Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
- Tianjin Research Institute of Anesthesiology, Tianjin, People's Republic of China
| | - Xiaoqing Bai
- Department of Science and Education, Tianjin Beichen Hospital, Tianjin, People's Republic of China
| | - Tongxiang Niu
- Tianjin Research Institute of Anesthesiology, Tianjin, People's Republic of China
- Department of Anesthesiology, Tianjin Jinnan Hospital, Tianjin, People's Republic of China
| | - Xinqi Zhuang
- Tianjin Research Institute of Anesthesiology, Tianjin, People's Republic of China
- Department of Anesthesiology, Tianjin Medical University Second Hospital, Tianjin, People's Republic of China
| | - Beibei Dong
- From the Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
- Tianjin Research Institute of Anesthesiology, Tianjin, People's Republic of China
| | - Yize Li
- From the Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
- Tianjin Research Institute of Anesthesiology, Tianjin, People's Republic of China
| | - Yonghao Yu
- From the Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
- Tianjin Research Institute of Anesthesiology, Tianjin, People's Republic of China
| | - Guolin Wang
- From the Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
- Tianjin Research Institute of Anesthesiology, Tianjin, People's Republic of China
| |
Collapse
|
8
|
Izquierdo C, Martín-Martínez M, Gómez-Monterrey I, González-Muñiz R. TRPM8 Channels: Advances in Structural Studies and Pharmacological Modulation. Int J Mol Sci 2021; 22:ijms22168502. [PMID: 34445208 PMCID: PMC8395166 DOI: 10.3390/ijms22168502] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/23/2021] [Accepted: 07/27/2021] [Indexed: 12/13/2022] Open
Abstract
The transient receptor potential melastatin subtype 8 (TRPM8) is a cold sensor in humans, activated by low temperatures (>10, <28 °C), but also a polymodal ion channel, stimulated by voltage, pressure, cooling compounds (menthol, icilin), and hyperosmolarity. An increased number of experimental results indicate the implication of TRPM8 channels in cold thermal transduction and pain detection, transmission, and maintenance in different tissues and organs. These channels also have a repercussion on different kinds of life-threatening tumors and other pathologies, which include urinary and respiratory tract dysfunctions, dry eye disease, and obesity. This compendium firstly covers newly described papers on the expression of TRPM8 channels and their correlation with pathological states. An overview on the structural knowledge, after cryo-electron microscopy success in solving different TRPM8 structures, as well as some insights obtained from mutagenesis studies, will follow. Most recently described families of TRPM8 modulators are also covered, along with a section of molecules that have reached clinical trials. To finalize, authors provide an outline of the potential prospects in the TRPM8 field.
Collapse
Affiliation(s)
- Carolina Izquierdo
- Departamento de Biomiméticos, Instituto de Química Médica, Juan de la Cierva 3, 28006 Madrid, Spain; (C.I.); (M.M.-M.)
- Programa de Doctorado en Química Orgánica, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Mercedes Martín-Martínez
- Departamento de Biomiméticos, Instituto de Química Médica, Juan de la Cierva 3, 28006 Madrid, Spain; (C.I.); (M.M.-M.)
| | - Isabel Gómez-Monterrey
- Dipartimento di Farmacia, Università degli Studi di Napoli “Federico II”, Via D. Montesano 49, 80131 Naples, Italy
- Correspondence: (I.G.-M.); (R.G.-M.)
| | - Rosario González-Muñiz
- Departamento de Biomiméticos, Instituto de Química Médica, Juan de la Cierva 3, 28006 Madrid, Spain; (C.I.); (M.M.-M.)
- Correspondence: (I.G.-M.); (R.G.-M.)
| |
Collapse
|
9
|
Liu L, Xu XB, Qu ZY, Zhao LP, Zhang CS, Li ZJ, Lyu TL, Wang XF, Jing XH, Li B. Determining 5HT 7R's Involvement in Modifying the Antihyperalgesic Effects of Electroacupuncture on Rats With Recurrent Migraine. Front Neurosci 2021; 15:668616. [PMID: 34163324 PMCID: PMC8215279 DOI: 10.3389/fnins.2021.668616] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 04/20/2021] [Indexed: 01/14/2023] Open
Abstract
Electroacupuncture (EA) is widely used in clinical practice to relieve migraine pain. 5-HT7 receptor (5-HT7R) has been reported to play an excitatory role in neuronal systems and regulate hyperalgesic pain and neurogenic inflammation. 5-HT7R could influence phosphorylation of protein kinase A (PKA)- or extracellular signal-regulated kinase1 / 2 (ERK1 / 2)-mediated signaling pathways, which mediate sensitization of nociceptive neurons via interacting with cyclic adenosine monophosphate (cAMP). In this study, we evaluated the role of 5-HT7R in the antihyperalgesic effects of EA and the underlying mechanism through regulation of PKA and ERK1 / 2 in trigeminal ganglion (TG) and trigeminal nucleus caudalis (TNC). Hyperalgesia was induced in rats with dural injection of inflammatory soup (IS) to cause meningeal neurogenic inflammatory pain. Electroacupuncture was applied for 15 min every other day before IS injection. Von Frey filaments, tail-flick, hot-plate, and cold-plated tests were used to evaluate the mechanical and thermal hyperalgesia. Neuronal hyperexcitability in TNC was studied by an electrophysiological technique. The 5-HT7R antagonist (SB269970) or 5-HT7R agonist (AS19) was administered intrathecally before each IS application at 2-day intervals during the 7-day injection protocol. The changes in 5-HT7R and 5-HT7R-associated signaling pathway were examined by real-time polymerase chain reaction (RT-PCR), Western blot, immunofluorescence, and enzyme-linked immunosorbent assay (ELISA) analyses. When compared with IS group, mechanical and thermal pain thresholds of the IS + EA group were significantly increased. Furthermore, EA prevented the enhancement of both spontaneous activity and evoked responses of second-order trigeminovascular neurons in TNC. Remarkable decreases in 5-HT7R mRNA expression and protein levels were detected in the IS + EA group. More importantly, 5-HT7R agonist AS19 impaired the antihyperalgesic effects of EA on p-PKA and p-ERK1 / 2. Injecting 5-HT7R antagonist SB-269970 into the intrathecal space of IS rats mimicked the effects of EA antihyperalgesia and inhibited p-PKA and p-ERK1 / 2. Our findings indicate that 5-HT7R mediates the antihyperalgesic effects of EA on IS-induced migraine pain by regulating PKA and ERK1 / 2 in TG and TNC.
Collapse
Affiliation(s)
- Lu Liu
- Department of Acupuncture and Moxibustion, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Key Laboratory of Acupuncture Neuromodulation, Beijing, China
| | - Xiao-Bai Xu
- Department of Acupuncture and Moxibustion, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Key Laboratory of Acupuncture Neuromodulation, Beijing, China
| | - Zheng-Yang Qu
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Luo-Peng Zhao
- Department of Acupuncture and Moxibustion, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Key Laboratory of Acupuncture Neuromodulation, Beijing, China.,Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing, China
| | - Claire-Shuiqing Zhang
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| | - Zhi-Juan Li
- Department of Acupuncture and Moxibustion, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Key Laboratory of Acupuncture Neuromodulation, Beijing, China
| | - Tian-Li Lyu
- Department of Acupuncture and Moxibustion, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Key Laboratory of Acupuncture Neuromodulation, Beijing, China
| | - Xue-Fei Wang
- Department of Acupuncture and Moxibustion, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Key Laboratory of Acupuncture Neuromodulation, Beijing, China
| | - Xiang-Hong Jing
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Bin Li
- Department of Acupuncture and Moxibustion, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Key Laboratory of Acupuncture Neuromodulation, Beijing, China
| |
Collapse
|
10
|
Nazıroğlu M, Öz A, Yıldızhan K. Selenium and Neurological Diseases: Focus on Peripheral Pain and TRP Channels. Curr Neuropharmacol 2021; 18:501-517. [PMID: 31903884 PMCID: PMC7457405 DOI: 10.2174/1570159x18666200106152631] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 11/26/2019] [Accepted: 01/04/2020] [Indexed: 12/18/2022] Open
Abstract
Pain is a complex physiological process that includes many components. Growing evidence supports the idea that oxidative stress and Ca2+ signaling pathways participate in pain detection by neurons. The main source of endogenous reactive oxygen species (ROS) is mitochondrial dysfunction induced by membrane depolarization, which is in turn caused by Ca2+ influx into the cytosol of neurons. ROS are controlled by antioxidants, including selenium. Selenium plays an important role in the nervous system, including the brain, where it acts as a cofactor for glutathione peroxidase and is incorporated into selenoproteins involved in antioxidant defenses. It has neuroprotective effects through modulation of excessive ROS production, inflammation, and Ca2+ overload in several diseases, including inflammatory pain, hypersensitivity, allodynia, diabetic neuropathic pain, and nociceptive pain. Ca2+ entry across membranes is mediated by different channels, including transient receptor potential (TRP) channels, some of which (e.g., TRPA1, TRPM2, TRPV1, and TRPV4) can be activated by oxidative stress and have a role in the induction of peripheral pain. The results of recent studies indicate the modulator roles of selenium in peripheral pain through inhibition of TRP channels in the dorsal root ganglia of experimental animals. This review summarizes the protective role of selenium in TRP channel regulation, Ca2+ signaling, apoptosis, and mitochondrial oxidative stress in peripheral pain induction.
Collapse
Affiliation(s)
- Mustafa Nazıroğlu
- Neuroscience Research Center, Suleyman Demirel University, Isparta, Turkey.,Department of Biophysics, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey.,Drug Discovery Unit, BSN Health, Analysis and Innovation Ltd. Inc. Teknokent, Isparta, Turkey
| | - Ahmi Öz
- Department of Biophysics, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
| | - Kenan Yıldızhan
- Department of Biophysics, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
| |
Collapse
|
11
|
Pratt SJP, Lee RM, Chang KT, Hernández-Ochoa EO, Annis DA, Ory EC, Thompson KN, Bailey PC, Mathias TJ, Ju JA, Vitolo MI, Schneider MF, Stains JP, Ward CW, Martin SS. Mechanoactivation of NOX2-generated ROS elicits persistent TRPM8 Ca 2+ signals that are inhibited by oncogenic KRas. Proc Natl Acad Sci U S A 2020; 117:26008-26019. [PMID: 33020304 PMCID: PMC7584994 DOI: 10.1073/pnas.2009495117] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Changes in the mechanical microenvironment and mechanical signals are observed during tumor progression, malignant transformation, and metastasis. In this context, understanding the molecular details of mechanotransduction signaling may provide unique therapeutic targets. Here, we report that normal breast epithelial cells are mechanically sensitive, responding to transient mechanical stimuli through a two-part calcium signaling mechanism. We observed an immediate, robust rise in intracellular calcium (within seconds) followed by a persistent extracellular calcium influx (up to 30 min). This persistent calcium was sustained via microtubule-dependent mechanoactivation of NADPH oxidase 2 (NOX2)-generated reactive oxygen species (ROS), which acted on transient receptor potential cation channel subfamily M member 8 (TRPM8) channels to prolong calcium signaling. In contrast, the introduction of a constitutively active oncogenic KRas mutation inhibited the magnitude of initial calcium signaling and severely blunted persistent calcium influx. The identification that oncogenic KRas suppresses mechanically-induced calcium at the level of ROS provides a mechanism for how KRas could alter cell responses to tumor microenvironment mechanics and may reveal chemotherapeutic targets for cancer. Moreover, we find that expression changes in both NOX2 and TRPM8 mRNA predict poor clinical outcome in estrogen receptor (ER)-negative breast cancer patients, a population with limited available treatment options. The clinical and mechanistic data demonstrating disruption of this mechanically-activated calcium pathway in breast cancer patients and by KRas activation reveal signaling alterations that could influence cancer cell responses to the tumor mechanical microenvironment and impact patient survival.
Collapse
Affiliation(s)
- Stephen J P Pratt
- Program in Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, MD 21201;
- Department of Pharmacology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Marlene and Stewart Greenebaum National Cancer Institute Comprehensive Cancer Center, School of Medicine, University of Maryland, Baltimore, MD 21201
| | - Rachel M Lee
- Department of Pharmacology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Marlene and Stewart Greenebaum National Cancer Institute Comprehensive Cancer Center, School of Medicine, University of Maryland, Baltimore, MD 21201
| | - Katarina T Chang
- Department of Pharmacology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Marlene and Stewart Greenebaum National Cancer Institute Comprehensive Cancer Center, School of Medicine, University of Maryland, Baltimore, MD 21201
| | - Erick O Hernández-Ochoa
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, MD 21201
| | - David A Annis
- Department of Pharmacology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Marlene and Stewart Greenebaum National Cancer Institute Comprehensive Cancer Center, School of Medicine, University of Maryland, Baltimore, MD 21201
| | - Eleanor C Ory
- Department of Pharmacology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Marlene and Stewart Greenebaum National Cancer Institute Comprehensive Cancer Center, School of Medicine, University of Maryland, Baltimore, MD 21201
| | - Keyata N Thompson
- Department of Pharmacology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Marlene and Stewart Greenebaum National Cancer Institute Comprehensive Cancer Center, School of Medicine, University of Maryland, Baltimore, MD 21201
| | - Patrick C Bailey
- Program in Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Department of Pharmacology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Marlene and Stewart Greenebaum National Cancer Institute Comprehensive Cancer Center, School of Medicine, University of Maryland, Baltimore, MD 21201
| | - Trevor J Mathias
- Department of Pharmacology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Marlene and Stewart Greenebaum National Cancer Institute Comprehensive Cancer Center, School of Medicine, University of Maryland, Baltimore, MD 21201
| | - Julia A Ju
- Department of Pharmacology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Marlene and Stewart Greenebaum National Cancer Institute Comprehensive Cancer Center, School of Medicine, University of Maryland, Baltimore, MD 21201
| | - Michele I Vitolo
- Department of Pharmacology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Marlene and Stewart Greenebaum National Cancer Institute Comprehensive Cancer Center, School of Medicine, University of Maryland, Baltimore, MD 21201
| | - Martin F Schneider
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, MD 21201
| | - Joseph P Stains
- Department of Orthopaedics, School of Medicine, University of Maryland, Baltimore, MD 21201
| | - Christopher W Ward
- Department of Orthopaedics, School of Medicine, University of Maryland, Baltimore, MD 21201
- School of Nursing, University of Maryland, Baltimore, MD 21201
| | - Stuart S Martin
- Department of Pharmacology, School of Medicine, University of Maryland, Baltimore, MD 21201;
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Marlene and Stewart Greenebaum National Cancer Institute Comprehensive Cancer Center, School of Medicine, University of Maryland, Baltimore, MD 21201
| |
Collapse
|
12
|
Increased Transient Receptor Potential Melastatin 8 Expression in the Development of Bladder Pain in Patients With Interstitial Cystitis/Bladder Pain Syndrome. Urology 2020; 146:301.e1-301.e6. [PMID: 33045289 DOI: 10.1016/j.urology.2020.09.037] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 09/25/2020] [Accepted: 09/27/2020] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To explore the role of transient receptor potential melastatin 8 (TRPM8) in the occurrence and development of bladder pain in interstitial cystitis/bladder pain syndrome (IC/BPS) patients. The differences in the content and location distribution of TRPM8 in bladder were compared between IC/BPS and control group. METHODS All enrolled patients answered questionnaire such as O'leary-Sant symptom index, visual analog scale (VAS), quality of life (QOL), and pelvic pain and urinary urgency frequency (PUF) score, then bladder specimens were collected. Analyses such as qRT-PCR, western blot, and immunofluorescence were performed to determine the changes in TRPM8 content and expression in neurons and sensory nerves between the IC/BPS and control group, and the relationships between TRPM8 and various clinical scores were also analyzed. RESULTS There were significant differences in the O'leary-Sant score, PUF score, VAS, and QOL score between IC/BPS and the control group (P < .05). Compared with the control group, the expression levels of TRPM8 mRNA and protein were significantly increased in the IC/BPS bladder tissues (P < .01). Immunofluorescence examination also revealed that (1) the number of neurons and sensory nerves displayed a significant upward trend in the bladder tissue of IC/BPS patients (2) the expression levels of TRPM8 on neurons and sensory nerves also increased significantly in IC/BPS group. CONCLUSION In IC/BPS patients, TRPM8 content increased significantly and mainly expressed on increased neurons and sensory nerves in bladder tissue. These results may indicate a mechanism by which bladder pain is more easily to spread in IC/BPS patients, and may also indicate an important mechanism for pain sensitization in such patients.
Collapse
|
13
|
Wang W, Wang H, Zhao Z, Huang X, Xiong H, Mei Z. Thymol activates TRPM8-mediated Ca 2+ influx for its antipruritic effects and alleviates inflammatory response in Imiquimod-induced mice. Toxicol Appl Pharmacol 2020; 407:115247. [PMID: 32971067 DOI: 10.1016/j.taap.2020.115247] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 08/06/2020] [Accepted: 09/18/2020] [Indexed: 12/24/2022]
Abstract
Psoriasis is a highly prevalent chronic dermatitis, characterized by widespread skin inflammation and spontaneous itch. Given the adverse reactions and drug dependence of current treatment, new drugs for psoriasis therapy are urgently needed. This study aims to explore the anti-psoriatic effects of thymol in imiquimod (IMQ) induced mice, and elucidate the potential mechanisms for its therapeutic activities. Thymol reduced the scratching behavior in IMQ mice, and activated Ca2+ response in cervical DRG neurons via TRPM8 channel. Also, thymol alleviated psoriasis-like skin lesions, and attenuated the enhanced infiltration of dermal neutrophils, dendritic cells (DCs) and Th17 cells. In addition, it reversed the upregulated expression of pro-inflammatory cytokines in the skin (TNF-α, IL-22, IL-23, IL-17A, IL-17F, IL-17C, IL-6, IL-1β and IFN-γ) and serum (TNF-α, IL-6, IL-1β, IL-17A and IFN-γ). Our results indicated that thymol can effectively ameliorate pruritus and the symptoms of psoriasis-like inflammation induced by IMQ, which makes it a promising drug for the treatment of psoriasis.
Collapse
Affiliation(s)
- Wen Wang
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan 430074, China
| | - Hua Wang
- College of Life Sciences, South-Central University for Nationalities, Wuhan 430074, China
| | - Zhongqiu Zhao
- Washington University School of Medicine, St. Louis, MO 63110, United States; Barnes-Jewish Hospital, St. Louis, MO 63110, United States
| | - Xiaoqing Huang
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (Ministry of Education), School of Pharmaceutical Sciences, Wuhan University, Wuhan 430072, China
| | - Hairong Xiong
- College of Life Sciences, South-Central University for Nationalities, Wuhan 430074, China.
| | - Zhinan Mei
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan 430074, China.
| |
Collapse
|
14
|
Bumgarner JR, Walker WH, Liu JA, Walton JC, Nelson RJ. Dim Light at Night Exposure Induces Cold Hyperalgesia and Mechanical Allodynia in Male Mice. Neuroscience 2020; 434:111-119. [PMID: 32201267 PMCID: PMC7176554 DOI: 10.1016/j.neuroscience.2020.03.022] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 03/10/2020] [Accepted: 03/12/2020] [Indexed: 01/13/2023]
Abstract
The growing presence of artificial lighting across the globe presents a number of challenges to human and ecological health despite its societal benefits. Exposure to artificial light at night, a seemingly innocuous aspect of modern life, disrupts behavior and physiological functions. Specifically, light at night induces neuroinflammation, which is implicated in neuropathic and nociceptive pain states, including hyperalgesia and allodynia. Because of its influence on neuroinflammation, we investigated the effects of dim light at night exposure on pain responsiveness in male mice. In this study, mice exposed to four days of dim (5 lux) light at night exhibited cold hyperalgesia. Further, after 28 days of exposure, mice exhibited both cold hyperalgesia and mechanical allodynia. No heat/hot hyperalgesia was observed in this experiment. Altered nociception in mice exposed to dim light at night was concurrent with upregulated interleukin-6 and nerve growth factor mRNA expression in the medulla and elevated μ-opioid receptor mRNA expression in the periaqueductal gray region of the brain. The current results support the relationship between disrupted circadian rhythms and altered pain sensitivity. In summary, we observed that dim light at night induces cold hyperalgesia and mechanical allodynia, potentially through elevated neuroinflammation and dysregulation of the endogenous opioid system.
Collapse
Affiliation(s)
- Jacob R Bumgarner
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA.
| | - William H Walker
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
| | - Jennifer A Liu
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
| | - James C Walton
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
| | - Randy J Nelson
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
| |
Collapse
|
15
|
Iftinca M, Basso L, Flynn R, Kwok C, Roland C, Hassan A, Defaye M, Ramachandran R, Trang T, Altier C. Chronic morphine regulates TRPM8 channels via MOR-PKCβ signaling. Mol Brain 2020; 13:61. [PMID: 32290846 PMCID: PMC7155267 DOI: 10.1186/s13041-020-00599-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 04/02/2020] [Indexed: 02/05/2023] Open
Abstract
Postoperative shivering and cold hypersensitivity are major side effects of acute and chronic opioid treatments respectively. TRPM8 is a cold and menthol-sensitive channel found in a subset of dorsal root ganglion (DRG) nociceptors. Deletion or inhibition of the TRPM8 channel was found to prevent the cold hyperalgesia induced by chronic administration of morphine. Here, we examined the mechanisms by which morphine was able to promote cold hypersensitivity in DRG neurons and transfected HEK cells. Mice daily injected with morphine for 5 days developed cold hyperalgesia. Treatment with morphine did not alter the expressions of cold sensitive TREK-1, TRAAK and TRPM8 in DRGs. However, TRPM8-expressing DRG neurons isolated from morphine-treated mice exhibited hyperexcitability. Sustained morphine treatment in vitro sensitized TRPM8 responsiveness to cold or menthol and reduced activation-evoked desensitization of the channel. Blocking phospholipase C (PLC) as well as protein kinase C beta (PKCβ), but not protein kinase A (PKA) or Rho-associated protein kinase (ROCK), restored channel desensitization. Identification of two PKC phosphorylation consensus sites, S1040 and S1041, in the TRPM8 and their site-directed mutation were able to prevent the MOR-induced reduction in TRPM8 desensitization. Our results show that activation of MOR by morphine 1) promotes hyperexcitability of TRPM8-expressing neurons and 2) induces a PKCβ-mediated reduction of TRPM8 desensitization. This MOR-PKCβ dependent modulation of TRPM8 may underlie the onset of cold hyperalgesia caused by repeated administration of morphine. Our findings point to TRPM8 channel and PKCβ as important targets for opioid-induced cold hypersensitivity.
Collapse
Affiliation(s)
- Mircea Iftinca
- Department of Physiology and Pharmacology, Inflammation Research Network-Snyder Institute for Chronic Diseases and Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Dr NW, Calgary, Alberta, T2N 4N1, Canada
| | - Lilian Basso
- Department of Physiology and Pharmacology, Inflammation Research Network-Snyder Institute for Chronic Diseases and Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Dr NW, Calgary, Alberta, T2N 4N1, Canada
| | - Robyn Flynn
- Hotchkiss Brain Institute. Cumming School of Medicine. University of Calgary, Calgary, Alberta, T2N 4N1, Canada
| | - Charlie Kwok
- Hotchkiss Brain Institute. Cumming School of Medicine. University of Calgary, Calgary, Alberta, T2N 4N1, Canada
| | - Corinne Roland
- Department of Physiology and Pharmacology, Inflammation Research Network-Snyder Institute for Chronic Diseases and Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Dr NW, Calgary, Alberta, T2N 4N1, Canada
| | - Ahmed Hassan
- Department of Physiology and Pharmacology, Inflammation Research Network-Snyder Institute for Chronic Diseases and Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Dr NW, Calgary, Alberta, T2N 4N1, Canada
| | - Manon Defaye
- Department of Physiology and Pharmacology, Inflammation Research Network-Snyder Institute for Chronic Diseases and Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Dr NW, Calgary, Alberta, T2N 4N1, Canada
| | - Rithwik Ramachandran
- Department of Physiology and Pharmacology. Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada
| | - Tuan Trang
- Hotchkiss Brain Institute. Cumming School of Medicine. University of Calgary, Calgary, Alberta, T2N 4N1, Canada
| | - Christophe Altier
- Department of Physiology and Pharmacology, Inflammation Research Network-Snyder Institute for Chronic Diseases and Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Dr NW, Calgary, Alberta, T2N 4N1, Canada.
| |
Collapse
|
16
|
Wright B, Kronen PW, Lascelles D, Monteiro B, Murrell JC, Robertson S, Steagall PVM, Yamashita K. Ice therapy: cool, current and complicated. J Small Anim Pract 2020; 61:267-271. [PMID: 32201945 DOI: 10.1111/jsap.13130] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 02/13/2020] [Accepted: 02/16/2020] [Indexed: 12/01/2022]
Abstract
This is the fourth Capsule review article provided by the WSAVA Global Pain Council and which discusses the use of ice or cold therapy as a non-pharmacologic modality for pain control in small animal practice. The physiological effects of cold therapy on tissues, receptors and ion channels are discussed; as well as indications, recommendations for, and limitations of use.
Collapse
Affiliation(s)
- B Wright
- Mistralvet, 4450 Thompson Parkway, Johnstown, CO, 80534, USA
| | - P W Kronen
- Veterinary Anaesthesia Service - International Zuercherstrasse 39, Winterthur 8400, Switzerland.,Center for Applied Biotechnologyand Molecular Medicine - Winterthurer Strasse 190, Zürich, 8057, Switzerland
| | - D Lascelles
- North Carolina State University - Comparative Pain Research Laboratory and Surgery Section 4700 Hillsborough Street Raleigh, Raleigh, NC, 27606, USA
| | - B Monteiro
- University of Montreal - Biomedical Sciences 3200 rueSicote, Saint-Hyacinthe, Quebec J2S 2M2, Canada
| | - J C Murrell
- University of Bristol - Clinical Veterinary Science Langford House Langford, Bristol BS40 5DU, United Kingdom of Great Britain and Northern Ireland
| | | | | | - K Yamashita
- Rakuno Gakuen University - Small Animal Clinical Sciences 582 Bunkyodai-Midorimachi, Ebetsu, Hokkaodo, 069-8501, Japan
| |
Collapse
|
17
|
Abstract
This paper is the fortieth consecutive installment of the annual anthological review of research concerning the endogenous opioid system, summarizing articles published during 2017 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides and receptors as well as effects of opioid/opiate agonists and antagonists. The review is subdivided into the following specific topics: molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors (1), the roles of these opioid peptides and receptors in pain and analgesia in animals (2) and humans (3), opioid-sensitive and opioid-insensitive effects of nonopioid analgesics (4), opioid peptide and receptor involvement in tolerance and dependence (5), stress and social status (6), learning and memory (7), eating and drinking (8), drug abuse and alcohol (9), sexual activity and hormones, pregnancy, development and endocrinology (10), mental illness and mood (11), seizures and neurologic disorders (12), electrical-related activity and neurophysiology (13), general activity and locomotion (14), gastrointestinal, renal and hepatic functions (15), cardiovascular responses (16), respiration and thermoregulation (17), and immunological responses (18).
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, CUNY, 65-30 Kissena Blvd., Flushing, NY, 11367, United States.
| |
Collapse
|
18
|
MacDonald DI, Wood JN, Emery EC. Molecular mechanisms of cold pain. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2020; 7:100044. [PMID: 32090187 PMCID: PMC7025288 DOI: 10.1016/j.ynpai.2020.100044] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 01/23/2020] [Accepted: 01/24/2020] [Indexed: 12/17/2022]
Abstract
The sensation of cooling is essential for survival. Extreme cold is a noxious stimulus that drives protective behaviour and that we thus perceive as pain. However, chronic pain patients suffering from cold allodynia paradoxically experience innocuous cooling as excruciating pain. Peripheral sensory neurons that detect decreasing temperature express numerous cold-sensitive and voltage-gated ion channels that govern their response to cooling in health and disease. In this review, we discuss how these ion channels control the sense of cooling and cold pain under physiological conditions, before focusing on the molecular mechanisms by which ion channels can trigger pathological cold pain. With the ever-rising number of patients burdened by chronic pain, we end by highlighting the pressing need to define the cells and molecules involved in cold allodynia and so identify new, rational drug targets for the analgesic treatment of cold pain.
Collapse
|
19
|
Li F, Yang W, Jiang H, Guo C, Huang AJW, Hu H, Liu Q. TRPV1 activity and substance P release are required for corneal cold nociception. Nat Commun 2019; 10:5678. [PMID: 31831729 PMCID: PMC6908618 DOI: 10.1038/s41467-019-13536-0] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 10/30/2019] [Indexed: 01/07/2023] Open
Abstract
As a protective mechanism, the cornea is sensitive to noxious stimuli. Here, we show that in mice, a high proportion of corneal TRPM8+ cold-sensing fibers express the heat-sensitive TRPV1 channel. Despite its insensitivity to cold, TRPV1 enhances membrane potential changes and electrical firing of TRPM8+ neurons in response to cold stimulation. This elevated neuronal excitability leads to augmented ocular cold nociception in mice. In a model of dry eye disease, the expression of TRPV1 in TRPM8+ cold-sensing fibers is increased, and results in severe cold allodynia. Overexpression of TRPV1 in TRPM8+ sensory neurons leads to cold allodynia in both corneal and non-corneal tissues without affecting their thermal sensitivity. TRPV1-dependent neuronal sensitization facilitates the release of the neuropeptide substance P from TRPM8+ cold-sensing neurons to signal nociception in response to cold. Our study identifies a mechanism underlying corneal cold nociception and suggests a potential target for the treatment of ocular pain. The eye shows protective responses to noxious stimuli including cold. Here, the authors show that TRPV1, found co-expressed on TRPM8 + fibres in the cornea, is necessary for cold nociception in the eye.
Collapse
Affiliation(s)
- Fengxian Li
- Department of Anesthesiology, Center for the Study of Itch and Sensory Disorders, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA.,Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Guangdong, China
| | - Weishan Yang
- Department of Anesthesiology, Center for the Study of Itch and Sensory Disorders, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Haowu Jiang
- Department of Anesthesiology, Center for the Study of Itch and Sensory Disorders, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Changxiong Guo
- Department of Anesthesiology, Center for the Study of Itch and Sensory Disorders, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Andrew J W Huang
- Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Hongzhen Hu
- Department of Anesthesiology, Center for the Study of Itch and Sensory Disorders, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Qin Liu
- Department of Anesthesiology, Center for the Study of Itch and Sensory Disorders, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA. .,Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
20
|
Basiri F, Rad A, Mahdian D, Molavi M, Amin B. Effects of glucosamine against morphine-induced antinociceptive tolerance and dependence in mice. J Biomed Sci 2019; 26:21. [PMID: 30782159 PMCID: PMC6380027 DOI: 10.1186/s12929-019-0513-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Accepted: 02/11/2019] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND The most important limitations of morphine in pain therapy are its tolerance and dependence. In this study, we evaluated the protective effect of glucosamine against morphine-induced tolerance and dependence in mice. METHODS Mice received twice daily morphine (20 mg/kg, s.c.) alone, or along with orally administered glucosamine (500, 1000 and 2000 mg/kg), for 9 continuous days. To assess antinociceptive effect of morphine, percentage of maximal possible effect (%MPE) of animals exposed to thermal stimulus was measured in the hot plate test, 30 min after morphine administration. Test was performed on days 1, 3, 5, 7 and 9. The effect of glucosamine on the naloxone (5 mg/kg, i.p.)-precipitated morphine withdrawal, was also evaluated. Changes in brain gene expression levels of induced nitric oxide synthase (iNOS), enzyme responsible for nitric oxide generation, as well as pro-inflammatory mediator, tumor necrosis alpha (TNF-α) were measured in morphine tolerated animals, as well as after withdrawal by real-time polymerase chain reaction (RT-PCR). Protein content of TNF-α was evaluated via ELISA assay. RESULTS Tolerance to antinociceptive effect of morphine was developed after 7 days of morphine treatment. The concurrent administration of glucosamine (500, 1000 and 2000 mg/kg) with morphine, significantly inhibited tolerance development, on days 7 and 9. In addition, glucosamine ameliorated the naloxone-precipitated opioid withdrawal symptoms (tremor, jumping, teeth chattering, grooming). However, diarrhea was significantly improved only with the dose of 500 mg/kg. Increased mRNA expression of iNOS as well as TNF-α mRNA expression and protein, after both morphine tolerance and withdrawal, were considerably reduced by glucosamine (1000 mg/kg) in the morphine withdrawal animals. CONCLUSION These data support the utility of glucosamine in attenuating both tolerance to nociceptive effects of morphine as well as withdrawal-induced behavioral profile. Anti-oxidant and anti-inflammatory effects are responsible, at least in part, for the protective effects of this drug.
Collapse
Affiliation(s)
- Faezeh Basiri
- Student Research Committee, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Abolfazl Rad
- Cellular and Molecular Research Center, Department of Physiology and Pharmacology, Faculty of Medicine, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Davood Mahdian
- Cellular and Molecular Research Center, Department of Physiology and Pharmacology, Faculty of Medicine, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Mehdi Molavi
- Departement of Internal Medicine, Faculty of Medicine, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Bahareh Amin
- Cellular and Molecular Research Center, Department of Physiology and Pharmacology, Faculty of Medicine, Sabzevar University of Medical Sciences, Sabzevar, Iran.
| |
Collapse
|
21
|
Opioid-induced hyperalgesia in clinical anesthesia practice: what has remained from theoretical concepts and experimental studies? Curr Opin Anaesthesiol 2018; 30:458-465. [PMID: 28590258 DOI: 10.1097/aco.0000000000000485] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW This article reviews the phenomenon of opioid-induced hyperalgesia (OIH) and its implications for clinical anesthesia. The goal of this review is to give an update on perioperative prevention and treatment strategies, based on findings in preclinical and clinical research. RECENT FINDINGS Several systems have been suggested to be involved in the pathophysiology of OIH with a focus on the glutaminergic system. Very recently preclinical data revealed that peripheral μ-opioid receptors (MORs) are key players in the development of OIH and acute opioid tolerance (AOT). Peripheral MOR antagonists could, thus, become a new prevention/treatment option of OIH in the perioperative setting. Although the impact of OIH on postoperative pain seems to be moderate, recent evidence suggests that increased hyperalgesia following opioid treatment correlates with the risk of developing persistent pain after surgery. In clinical practice, distinction among OIH, AOT and acute opioid withdrawal remains difficult, especially because a specific quantitative sensory test to diagnose OIH has not been validated yet. SUMMARY Since the immediate postoperative period is not ideal to initiate long-term treatment for OIH, the best strategy is to prevent its occurrence. A multimodal approach, including choice of opioid, dose limitations and addition of nonopioid analgesics, is recommended.
Collapse
|
22
|
Kerui G, Jasmin L. Dual effects of brain sparing opioid in newborn rats: Analgesia and hyperalgesia. NEUROBIOLOGY OF PAIN 2018; 3:1-7. [PMID: 31194154 PMCID: PMC6550121 DOI: 10.1016/j.ynpai.2018.01.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Revised: 01/08/2018] [Accepted: 01/09/2018] [Indexed: 01/16/2023]
Abstract
The peripherally acting opioid loperamide produces sustained antinociception in the newborn rat. Loperamide minimally crosses the blood brain barrier in the newborn rat. Daily systemic administration of loperamide produces opioid induced hyperalgesia in the newborn rat.
Effective pain management in neonates without the unwanted central nervous system (CNS) side effects remains an unmet need. To circumvent these central effects we tested the peripherally acting (brain sparing) opioid agonist loperamide in neonate rats. Our results show that: 1) loperamide (1 mg/kg, s.c.) does not affect the thermal withdrawal latency in the normal hind paw while producing antinociception in all pups with an inflamed hind paw. 2) A dose of loperamide 5 times higher resulted in only 6.9 ng/mL of loperamide in the cerebrospinal fluid (CSF), confirming that loperamide minimally crosses the blood–brain barrier (BBB). 3) Unexpectedly, sustained administration of loperamide for 5 days resulted in a hyperalgesic behavior, as well as increased excitability (sensitization) of dorsal root ganglia (DRGs) and spinal nociceptive neurons. This indicates that opioid induced hyperalgesia (OIH) can be induced through the peripheral nervous system. Unless prevented, OIH could in itself be a limiting factor in the use of brain sparing opioids in the neonate.
Collapse
Affiliation(s)
- Gong Kerui
- Department of Oral and Maxillofacial Surgery, University of California San Francisco, San Francisco, CA, United States
| | - Luc Jasmin
- Department of Oral and Maxillofacial Surgery, University of California San Francisco, San Francisco, CA, United States
| |
Collapse
|
23
|
Effects of menthol and its interaction with nicotine-conditioned cue on nicotine-seeking behavior in rats. Psychopharmacology (Berl) 2017; 234:3443-3453. [PMID: 28918457 PMCID: PMC5693741 DOI: 10.1007/s00213-017-4736-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 08/24/2017] [Indexed: 10/18/2022]
Abstract
RATIONALE Increasing clinical evidence suggests that menthol, a significant flavoring additive in tobacco products, may contribute to smoking and nicotine dependence. Relapse to smoking behavior presents a formidable challenge for the treatment of tobacco addiction. An unresolved issue is whether the mentholation of tobacco products precipitates relapse to tobacco use in abstinent smokers. OBJECTIVES The present study examined the effects of menthol on the perseverance and relapse of nicotine-seeking behavior in rats. METHODS Male Sprague-Dawley rats were trained to press a lever for intravenous nicotine self-administration (0.03 mg/kg/infusion) under a fixed-ratio five schedule of reinforcement. Each nicotine infusion was signaled by the presentation of a sensory stimulus that was established as a discrete nicotine-conditioned cue. Five minutes prior to the sessions, the rats received an intraperitoneal injection of menthol (0.1 mg/kg) or vehicle. In the subsequent extinction test sessions, nicotine was unavailable with or without menthol and/or the nicotine-conditioned cue. The reinstatement tests were performed the following day after the extinction criterion was met. Menthol was also tested on food-seeking responses. In a subset of nicotine-trained rats, a transient receptor potential melastatin 8 (TRPM8) antagonist RQ-00203078 was given prior to menthol administration. RESULTS Continued administration of menthol sustained responses on the previously active and nicotine-reinforced lever in the extinction tests. The readministration of menthol after extinction reinstated active lever responses. In both the extinction and the reinstatement tests, a combination of pre-session menthol administration and cue representation during the session produced a more robust behavioral effect than either menthol or the cue alone. No such effects of menthol was observed in food trained rats. RQ-00203078 did not change menthol effect on nicotine seeking. CONCLUSION These data demonstrated that menthol specifically sustained and reinstated nicotine-seeking behavior, and this effect was independent of TRPM8 activity. These findings suggest that menthol in most tobacco products, even not menthol labeled, may contribute to the perseverance of and relapse to tobacco-seeking behavior.
Collapse
|
24
|
Downregulations of TRPM8 expression and membrane trafficking in dorsal root ganglion mediate the attenuation of cold hyperalgesia in CCI rats induced by GFRα3 knockdown. Brain Res Bull 2017; 135:8-24. [PMID: 28867384 DOI: 10.1016/j.brainresbull.2017.08.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 07/07/2017] [Accepted: 08/04/2017] [Indexed: 12/22/2022]
Abstract
BACKGROUND Cold hyperalgesia is an intractable sensory abnormality commonly seen in peripheral neuropathies. Although glial cell line-derived neurotrophic factor family receptor alpha3 (GFRα3) is required for the formation of pathological cold pain has been revealed, potential transduction mechanism is poorly elucidated. We have previously demonstrated the contribution of enhanced activity of transient receptor potential melastatin 8 (TRPM8) to cold hyperalgesia in neuropathic pain using a rat model of chronic constriction injury (CCI) to the sciatic nerve. Recently, the enhancement of TRPM8 activity is attributed to the increased TRPM8 plasma membrane trafficking. In addition, TRPM8 can be sensitized by the activation of GFRα3, leading to increased cold responses in vivo. The aim of this study was to investigate whether GFRα3 could influence cold hyperalgesia of CCI rats via modulating TRPM8 expression and plasma membrane trafficking in dorsal root ganglion (DRG). METHODS Mechanical allodynia, cold and heat hyperalgesia were measured on 1day before CCI and the 1st, 4th, 7th, 10th and 14th day after CCI. TRPM8 total expression and membrane trafficking as well as GFRα3 expression in DRG were detected by immunofluorescence and western blot. Furthermore, GFRα3 small interfering RNA (siRNA) was intrathecally administrated to reduce GFRα3 expression in DRG, and the effects of GFRα3 knockdown on CCI-induced behavioral sensitization as well as TRPM8 total expression and membrane trafficking in both mRNA and protein levels were investigated, and the change in coexpression of TRPM8 with GFRα3 was also evaluated. Then, the effect of GFRα3 activation with artemin on pain behavior of CCI rats pretreated with the selective TRPM8 antagonist RQ-00203078 was observed. RESULTS Here we found that TRPM8 total expression and plasma membrane trafficking as well as GFRα3 expression in DRG were initially increased on the 4th day after CCI, and maintained at the peak level from the 10th to the 14th day, which entirely conformed with the induction and maintenance of behavioral-reflex facilitation following CCI. The coexpression of TRPM8 with GFRα3, which was mainly located in peptidergic C-fibers DRG neurons, was also increased after CCI. Downregulation of GFRα3 protein in DRG attenuated CCI-induced cold hyperalgesia without affecting mechanical allodynia and heat hyperalgesia, and reduced the upregulations of TRPM8 total expression and plasma membrane trafficking as well as coexpression of TRPM8 with GFRα3 induced by CCI. Additionally, the inhibition of TRPM8 abolished the influence of GFRα3 activation on cold hyperalgesia after CCI. CONCLUSION Our results demonstrate that GFRα3 knockdown specially inhibits cold hyperalgesia following CCI via decreasing the expression level and plasma membrane trafficking of TRPM8 in DRG. GFRα3 and its downstream mediator, TRPM8, represent a new analgesia axis which can be further exploited in sensitized cold reflex under the condition of chronic pain.
Collapse
|
25
|
Galor A, Small L, Feuer W, Levitt RC, Sarantopoulos KD, Yosipovitch G. The Relationship Between Ocular Itch, Ocular Pain, and Dry Eye Symptoms (An American Ophthalmological Society Thesis). TRANSACTIONS OF THE AMERICAN OPHTHALMOLOGICAL SOCIETY 2017; 115:T5. [PMID: 29391860 PMCID: PMC5774848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
PURPOSE To evaluate associations between sensations of ocular itch and dry eye (DE) symptoms, including ocular pain, and DE signs. METHODS A cross-sectional study of 324 patients seen in the Miami Veterans Affairs eye clinic was performed. The evaluation consisted of questionnaires regarding ocular itch, DE symptoms, descriptors of neuropathic-like ocular pain (NOP), and evoked pain sensitivity testing on the forehead and forearm, followed by a comprehensive ocular surface examination including corneal mechanical sensitivity testing. Analyses were performed to examine for differences between those with and without subjective complaints of ocular itch. RESULTS The mean age was 62 years with 92% being male. Symptoms of DE and NOP were more frequent in patients with moderate-severe ocular itch compared to those with no or mild ocular itch symptoms. With the exception of ocular surface inflammation (abnormal matrix metalloproteinase 9 testing) which was less common in those with moderate-severe ocular itch symptoms, DE signs were not related to ocular itch. Individuals with moderate-severe ocular itch also demonstrated greater sensitivity to evoked pain on the forearm and had higher non-ocular pain, depression, and post-traumatic stress disorders scores, compared to those with no or mild itch symptoms. CONCLUSIONS Subjects with moderate-severe ocular itch symptoms have more severe symptoms of DE, NOP, non-ocular pain and demonstrate abnormal somatosensory testing in the form of increased sensitivity to evoked pain at a site remote from the eye, consistent with generalized hypersensitivity.
Collapse
Affiliation(s)
- Anat Galor
- Corresponding Author: Anat Galor, MD, MSPH
| | | | | | | | | | | |
Collapse
|