1
|
Gupta A, Mishra SK, Lascelles BDX. Emerging evidence of artemin/GFRα3 signaling in musculoskeletal pain. Osteoarthritis Cartilage 2025; 33:196-206. [PMID: 39374825 PMCID: PMC11757073 DOI: 10.1016/j.joca.2024.09.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 09/17/2024] [Accepted: 09/30/2024] [Indexed: 10/09/2024]
Abstract
Chronic musculoskeletal pain is highly prevalent and poses a significant personal, societal, and economic burden. Management of chronic musculoskeletal pain remains a challenge. Long-term use of common analgesic medications such as nonsteroidal anti-inflammatory drugs and opioids is associated with adverse events, and in the case of opioids, drug addiction. Additionally, many individuals do not experience sufficient pain relief with these therapeutic approaches. Thus, there is an urgent need to develop clinically efficacious and safe therapeutics for musculoskeletal pain. Recent advances in our understanding of musculoskeletal pain neurobiology have helped identify the role of neurotrophic factors, specifically, the glial cell line-derived neurotrophic factor (GDNF) family of ligands (GFL) and their associated signaling pathways. This review outlines our current understanding of the GFL signaling systems, discusses their role in inflammatory and chronic musculoskeletal pain and sensitivity, and comments on the analgesic therapeutic potential of targeting the GFL signaling system.
Collapse
Affiliation(s)
- Ankita Gupta
- Translational Research in Pain Program, Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA
| | - Santosh K Mishra
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA.
| | - B Duncan X Lascelles
- Comparative Pain Research and Education Center, North Carolina State University, Raleigh, NC, USA; Thurston Arthritis Center, UNC School of Medicine, Chapel Hill, NC, USA; Center for Translational Pain Research, Department of Anesthesiology, Duke University, Durham, NC, USA.
| |
Collapse
|
2
|
Boullon L, Finn DP, Llorente-Berzal Á. Sex differences in the affective-cognitive dimension of neuropathic pain: Insights from the spared nerve injury rat model. THE JOURNAL OF PAIN 2025; 27:104752. [PMID: 39626836 DOI: 10.1016/j.jpain.2024.104752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 11/22/2024] [Accepted: 11/27/2024] [Indexed: 12/08/2024]
Abstract
Over 40% of neuropathic pain patients experience mood and cognitive disturbances, often showing reduced response to analgesics, with most affected individuals being female. This highlights the critical role of biological sex in pain-related affective and cognitive disorders, making it essential to understand the emotional and cognitive circuits linked to pain for improving treatment strategies. However, research on sex differences in preclinical pain models is lacking. This study aimed to investigate these differences using the spared nerve injury (SNI) rat model, conducting a comprehensive series of behavioural tests over 100 days post-injury to identify key time points for observing sex-specific behaviours indicative of pain-related conditions. The findings revealed that female rats exhibited greater mechanical and cold hypersensitivity compared to males following nerve injury and showed earlier onset of depression-related behaviours, while males were more prone to anxiety, social, and memory-related alterations. Interestingly, by the 14th week post-injury, females displayed no signs of these emotional and cognitive impairments. Additionally, fluctuations in the oestrous cycle or changes in testosterone and oestradiol levels did not correlate with sex differences in pain sensitivity or negative affect. Recognizing the influence of biological sex on pain-induced affective and cognitive alterations, especially in later stages post-injury, is crucial for enhancing our understanding of this complex pain disorder. PERSPECTIVE: This manuscript reports the relevance of long-term investigations of sex differences in chronic pain. It shows differential development of somatosensory sensitivity, negative affective states and cognitive impairments in males and females. It emphasizes the importance of including subjects of both sexes in the investigation of pain-related mechanisms and therapeutic management.
Collapse
Affiliation(s)
- Laura Boullon
- Pharmacology and Therapeutics, School of Medicine, Human Biology Building, University of Galway, Galway, Ireland; Galway Neuroscience Centre, University of Galway, Galway, Ireland; Centre for Pain Research, University of Galway, Galway, Ireland
| | - David P Finn
- Pharmacology and Therapeutics, School of Medicine, Human Biology Building, University of Galway, Galway, Ireland; Galway Neuroscience Centre, University of Galway, Galway, Ireland; Centre for Pain Research, University of Galway, Galway, Ireland
| | - Álvaro Llorente-Berzal
- Pharmacology and Therapeutics, School of Medicine, Human Biology Building, University of Galway, Galway, Ireland; Galway Neuroscience Centre, University of Galway, Galway, Ireland; Centre for Pain Research, University of Galway, Galway, Ireland; Department of Physiology, School of Medicine, Autonomous University of Madrid, Madrid, Spain.
| |
Collapse
|
3
|
Yang C, Wei C, Alam S, Chen X, McKemy DD. The neurotrophic factor artemin and its receptor GFRα3 mediate migraine-like pain via the ion channel TRPM8. Cephalalgia 2024; 44:3331024241297679. [PMID: 39552306 DOI: 10.1177/03331024241297679] [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: 11/19/2024]
Abstract
BACKGROUND Migraine has a strong genetic foundation, including both monogenic and polygenic types. The former are rare, with most migraine considered polygenic, supported by genome-wide association studies (GWAS) identifying numerous genetic variants linked with migraine risk. Surprisingly, some of the most common mutations are associated with transient receptor potential melastatin 8 (TRPM8), a non-selective cation channel that is the primary sensor of cold temperatures in cutaneous primary afferents of the somatosensory system. However, it is unlikely that the temperature sensitivity of TRPM8 is relevant in migraine-related tissues, such as the meninges, suggesting other activation mechanisms underly its role in migraine pathogenesis. Thus, to define the basis of the channel's involvement, we reasoned that cellular processes that increase cold sensitivity in the skin, such as the neurotrophic factor artemin, via its receptor glial cell-line derived neurotrophic factor family receptor alpha-3 (GFRα3), also mediate TRPM8-associated migraine-like pain in the meninges. METHODS To investigate the role of artemin and GFRα3 in preclinical rodent migraine models, we infused nitroglycerin acutely and chronically, and measured changes in periorbital and hind paw mechanical sensitivity in male and female mice lacking GFRα3, after neutralization of free artemin with specific monoclonal antibodies, or by systemic treatment with a TRPM8-specific antagonist. Further, in mice lacking GFRα3 we tested the effects of supradural infusions of a mix of inflammatory mediators, as well as tested if dura stimulation with artemin or a TRPM8-specific agonist induce migraine-related pain in mice. RESULTS We find that mechanical allodynia induced by systemic nitroglycerin, or supradural infusion of inflammatory mediators, involves GFRα3. In addition, neutralization of circulating artemin reduces the nitroglycerin phenotype, demonstrating the importance of this neurotrophic pathway in headaches. Further, we show TRPM8 expression in the meninges, and that direct supradural infusion of either a TRPM8-specific agonist or artemin itself produces mechanical allodynia, with the latter dependent on TRPM8 and ameliorated by concurrent treatment with sumatriptan. CONCLUSIONS These results indicate that neuroinflammatory events in the meninges can produce migraine-like pain in mice via artemin and GFRα3, likely acting upstream of TRPM8, providing a novel pathway that may contribute to headaches or migraine pathogenesis.
Collapse
Affiliation(s)
- Chenyu Yang
- Neurobiology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA
- Molecular and Computational Biology Graduate Program, University of Southern California, Los Angeles, CA, USA
| | - Chao Wei
- Neurobiology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA
- Department of Anesthesia and Critical Care, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Sanaa Alam
- Neurobiology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA
| | - Xunyang Chen
- Neurobiology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA
| | - David D McKemy
- Neurobiology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA
- Molecular and Computational Biology Graduate Program, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
4
|
Lewis CM, Griffith TN. Ion channels of cold transduction and transmission. J Gen Physiol 2024; 156:e202313529. [PMID: 39051992 PMCID: PMC11273221 DOI: 10.1085/jgp.202313529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 06/04/2024] [Accepted: 07/12/2024] [Indexed: 07/27/2024] Open
Abstract
Thermosensation requires the activation of a unique collection of ion channels and receptors that work in concert to transmit thermal information. It is widely accepted that transient receptor potential melastatin 8 (TRPM8) activation is required for normal cold sensing; however, recent studies have illuminated major roles for other ion channels in this important somatic sensation. In addition to TRPM8, other TRP channels have been reported to contribute to cold transduction mechanisms in diverse sensory neuron populations, with both leak- and voltage-gated channels being identified for their role in the transmission of cold signals. Whether the same channels that contribute to physiological cold sensing also mediate noxious cold signaling remains unclear; however, recent work has found a conserved role for the kainite receptor, GluK2, in noxious cold sensing across species. Additionally, cold-sensing neurons likely engage in functional crosstalk with nociceptors to give rise to cold pain. This Review will provide an update on our understanding of the relationship between various ion channels in the transduction and transmission of cold and highlight areas where further investigation is required.
Collapse
Affiliation(s)
- Cheyanne M Lewis
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA, USA
| | - Theanne N Griffith
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA, USA
| |
Collapse
|
5
|
Yang C, Wei C, Alam S, Chen X, McKemy DD. The neurotrophic factor artemin and its receptor GFRα3 mediate migraine-like pain via the ion channel TRPM8. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.09.611532. [PMID: 39314341 PMCID: PMC11419092 DOI: 10.1101/2024.09.09.611532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Background Migraine has a strong genetic foundation, including both monogenic and polygenic types. The former are rare, with most migraine considered polygenic, supported by genome-wide association studies (GWAS) identifying numerous genetic variants associated with migraine risk. Surprisingly, some of the most common mutations are associated with TRPM8, a non-selective cation channel that is the primary sensor of cold temperatures in primary afferent neurons of the somatosensory system. However, it is unlikely that the temperature sensitivity of TRPM8 underlies its role in migraine pathogenesis. To define the basis of the channel's involvement, we reasoned that cellular processes that increase cold sensitivity in the skin, such as the neurotrophic factor artemin, via its receptor GFRα3, also mediate TRPM8-associated migraine-like pain in the meninges. Methods To investigate the role of artemin and GFRα3 in preclinical rodent migraine models, we infused nitroglycerin acutely and chronically, and measured changes in periorbital and hind paw mechanical sensitivity in male and female mice lacking GFRα3, after neutralization of free artemin with specific monoclonal antibodies, or by systemic treatment with a TRPM8-specific antagonist. Further, in wildtypes and mice lacking either GFRα3 or TRPM8, we tested the effects of supradural infusions of a mix of inflammatory mediators, artemin, and a TRPM8-specific agonist on migraine-related pain in mice. Results We find that mechanical allodynia induced by systemic nitroglycerin, or supradural infusion of inflammatory mediators, involves GFRα3. In addition, neutralization of circulating artemin reduces the nitroglycerin phenotype, demonstrating the importance of this neurotrophic pathway. Further, we show TRPM8 expression in the meninges and that direct supradural infusion of either a TRPM8-specific agonist or artemin itself produces mechanical allodynia, the latter dependent on TRPM8 and ameliorated by concurrent treatment with sumatriptan. Conclusions These results indicate that neuroinflammatory events in the meninges can produce migraine-like pain in mice via artemin and GFRα3, likely acting upstream of TRPM8, providing a novel pathway that may contribute to migraine pathogenesis.
Collapse
Affiliation(s)
- Chenyu Yang
- Neurobiology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089
- Molecular and Computational Biology Graduate Program, University of Southern California, Los Angeles, CA 90089
| | - Chao Wei
- Neurobiology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089
- Department of Anesthesia and Critical Care, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215
| | - Sanaa Alam
- Neurobiology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089
| | - Xunyang Chen
- Neurobiology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089
| | - David D. McKemy
- Neurobiology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089
- Molecular and Computational Biology Graduate Program, University of Southern California, Los Angeles, CA 90089
| |
Collapse
|
6
|
Johnson SL, Missig G, Wang M, Ouk K, Gupta K, Nguyen HN, Toh MF, Ho TSY, Gray D, Zhang H, Choi-Sledeski YM, Barberis C, Stone DJ, Pin S, Lim J. Discovery of the first selective, small-molecule GFRα2/3 inhibitors through DNA-encoded library technology. Bioorg Med Chem Lett 2024; 110:129889. [PMID: 39004318 DOI: 10.1016/j.bmcl.2024.129889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 07/08/2024] [Accepted: 07/11/2024] [Indexed: 07/16/2024]
Abstract
Studies have shown that disrupting the formation of the ligand-RET-GFRα complex could be an effective way of treating pain and itch. Compared to traditional high-throughput screens, DNA encoded libraries (DELs) have distinguished themselves as a powerful technology for hit identification in recent years. The present work demonstrates the use of DEL technology identifying compound 16 as the first GFRa2/GFRa3 small molecule inhibitor (0.1/0.2 μM respectively) selective over RET. This molecule represents an opportunity to advance the development of small-molecule inhibitors targeting the GFRα-RET interface for the treatment of pain and itch.
Collapse
Affiliation(s)
- Shea L Johnson
- Cerevel Therapeutics, 222 Jacobs St., Cambridge, MA 02141, United States.
| | - Galen Missig
- Cerevel Therapeutics, 222 Jacobs St., Cambridge, MA 02141, United States
| | - Minghua Wang
- Cerevel Therapeutics, 222 Jacobs St., Cambridge, MA 02141, United States
| | - Kosalvisal Ouk
- Cerevel Therapeutics, 222 Jacobs St., Cambridge, MA 02141, United States
| | - Kushali Gupta
- Cerevel Therapeutics, 222 Jacobs St., Cambridge, MA 02141, United States
| | - Hanh Nho Nguyen
- Cerevel Therapeutics, 222 Jacobs St., Cambridge, MA 02141, United States
| | - May Fern Toh
- Cerevel Therapeutics, 222 Jacobs St., Cambridge, MA 02141, United States
| | - Tammy Szu-Yu Ho
- Cerevel Therapeutics, 222 Jacobs St., Cambridge, MA 02141, United States
| | - David Gray
- Cerevel Therapeutics, 222 Jacobs St., Cambridge, MA 02141, United States
| | - Hongjun Zhang
- Cerevel Therapeutics, 222 Jacobs St., Cambridge, MA 02141, United States
| | | | - Claude Barberis
- Cerevel Therapeutics, 222 Jacobs St., Cambridge, MA 02141, United States
| | - David J Stone
- Cerevel Therapeutics, 222 Jacobs St., Cambridge, MA 02141, United States
| | - Sokhom Pin
- Cerevel Therapeutics, 222 Jacobs St., Cambridge, MA 02141, United States
| | - Jongwon Lim
- Cerevel Therapeutics, 222 Jacobs St., Cambridge, MA 02141, United States
| |
Collapse
|
7
|
Sperduti M, Tagliamonte NL, Taffoni F, Guglielmelli E, Zollo L. Mechanical and thermal stimulation for studying the somatosensory system: a review on devices and methods. J Neural Eng 2024; 21:051001. [PMID: 39163886 DOI: 10.1088/1741-2552/ad716d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 08/20/2024] [Indexed: 08/22/2024]
Abstract
The somatosensory system is widely studied to understand its functioning mechanisms. Multiple tests, based on different devices and methods, have been performed not only on humans but also on animals andex-vivomodels. Depending on the nature of the sample under analysis and on the scientific aims of interest, several solutions for experimental stimulation and for investigations on sensation or pain have been adopted. In this review paper, an overview of the available devices and methods has been reported, also analyzing the representative values adopted during literature experiments. Among the various physical stimulations used to study the somatosensory system, we focused only on mechanical and thermal ones. Based on the analysis of their main features and on literature studies, we pointed out the most suitable solution for humans, rodents, andex-vivomodels and investigation aims (sensation and pain).
Collapse
Affiliation(s)
- M Sperduti
- Università Campus Bio-Medico di Roma, Research Unit of Advanced Robotics and Human-Centered Technologies, Via Alvaro del Portillo 21, 00128 Rome, Italy
| | - N L Tagliamonte
- Università Campus Bio-Medico di Roma, Research Unit of Advanced Robotics and Human-Centered Technologies, Via Alvaro del Portillo 21, 00128 Rome, Italy
| | - F Taffoni
- Università Campus Bio-Medico di Roma, Research Unit of Advanced Robotics and Human-Centered Technologies, Via Alvaro del Portillo 21, 00128 Rome, Italy
| | - E Guglielmelli
- Università Campus Bio-Medico di Roma, Research Unit of Advanced Robotics and Human-Centered Technologies, Via Alvaro del Portillo 21, 00128 Rome, Italy
| | - L Zollo
- Università Campus Bio-Medico di Roma, Research Unit of Advanced Robotics and Human-Centered Technologies, Via Alvaro del Portillo 21, 00128 Rome, Italy
| |
Collapse
|
8
|
Ferland S, Wang F, De Koninck Y, Ferrini F. An improved conflict avoidance assay reveals modality-specific differences in pain hypersensitivity across sexes. Pain 2024; 165:1304-1316. [PMID: 40323997 PMCID: PMC11090034 DOI: 10.1097/j.pain.0000000000003132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 10/17/2023] [Accepted: 11/06/2023] [Indexed: 01/27/2024]
Abstract
ABSTRACT Abnormal encoding of somatosensory modalities (ie, mechanical, cold, and heat) are a critical part of pathological pain states. Detailed phenotyping of patients' responses to these modalities have raised hopes that analgesic treatments could one day be tailored to a patient's phenotype. Such precise treatment would require a profound understanding of the underlying mechanisms of specific pain phenotypes at molecular, cellular, and circuitry levels. Although preclinical pain models have helped in that regard, the lack of a unified assay quantifying detailed mechanical, cold, and heat pain responses on the same scale precludes comparing how analgesic compounds act on different sensory phenotypes. The conflict avoidance assay is promising in that regard, but testing conditions require validation for its use with multiple modalities. In this study, we improve upon the conflict avoidance assay to provide a validated and detailed assessment of all 3 modalities within the same animal, in mice. We first optimized testing conditions to minimize the necessary amount of training and to reduce sex differences in performances. We then tested what range of stimuli produce dynamic stimulus-response relationships for different outcome measures in naive mice. We finally used this assay to show that nerve injury produces modality-specific sex differences in pain behavior. Our improved assay opens new avenues to study the basis of modality-specific abnormalities in pain behavior.
Collapse
Affiliation(s)
| | - Feng Wang
- CERVO Brain Research Centre, Québec, QC, Canada
- Faculty of Dentistry, Université Laval, Québec, QC, Canada
| | - Yves De Koninck
- CERVO Brain Research Centre, Québec, QC, Canada
- Department of Psychiatry and Neuroscience, Université Laval, Québec, QC, Canada
| | - Francesco Ferrini
- CERVO Brain Research Centre, Québec, QC, Canada
- Department of Psychiatry and Neuroscience, Université Laval, Québec, QC, Canada
- Department of Veterinary Sciences, University of Turin, Turin, Italy
| |
Collapse
|
9
|
Zhang Q, Weng W, Gu X, Xiang J, Yang Y, Zhu MX, Gu W, He Z, Li Y. hnRNPA1 SUMOylation promotes cold hypersensitivity in chronic inflammatory pain by stabilizing TRPA1 mRNA. Cell Rep 2023; 42:113401. [PMID: 37943660 DOI: 10.1016/j.celrep.2023.113401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 07/17/2023] [Accepted: 10/24/2023] [Indexed: 11/12/2023] Open
Abstract
TRPA1 is pivotal in cold hypersensitivity, but its regulatory mechanisms in inflammatory cold hyperalgesia remain poorly understood. We show here that the upregulation of SUMO1-conjugated protein levels in a complete Freund's adjuvant (CFA)-induced inflammatory pain model enhances TRPA1 mRNA stability, ultimately leading to increased expression levels. We further demonstrate that hnRNPA1 binds to TRPA1 mRNA, and its SUMOylation, upregulated in CFA-induced inflammatory pain, contributes to stabilizing TRPA1 mRNA by accumulating hnRNPA1 in the cytoplasm. Moreover, we find that wild-type hnRNPA1 viral infection in dorsal root ganglia neurons, and not infection with the SUMOylation-deficient hnRNPA1 mutant, can rescue the reduced ability of hnRNPA1-knockdown mice to develop inflammatory cold pain hypersensitivity. These results suggest that hnRNPA1 is a regulator of TRPA1 mRNA stability, the capability of which is enhanced upon SUMO1 conjugation at lysine 3 in response to peripheral inflammation, and the increased expression of TRPA1 in turn underlies the development of chronic inflammatory cold pain hypersensitivity.
Collapse
Affiliation(s)
- Qiao Zhang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Weiji Weng
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiaokun Gu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jinhua Xiang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yang Yang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Michael X Zhu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Weidong Gu
- Department of Anesthesiology, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China.
| | - Zhenzhou He
- Department of Anesthesiology, Minhang Hospital Affiliated to Fudan University, Shanghai 201199, China.
| | - Yong Li
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
10
|
Kocot-Kępska M, Pawlik K, Ciapała K, Makuch W, Zajączkowska R, Dobrogowski J, Przeklasa-Muszyńska A, Mika J. Phenytoin Decreases Pain-like Behaviors and Improves Opioid Analgesia in a Rat Model of Neuropathic Pain. Brain Sci 2023; 13:858. [PMID: 37371338 DOI: 10.3390/brainsci13060858] [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: 04/18/2023] [Revised: 05/18/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
Neuropathic pain remains a clinical challenge due to its complex and not yet fully understood pathomechanism, which result in limited analgesic effectiveness of the management offered, particularly for patients with acute, refractory neuropathic pain states. In addition to the introduction of several modern therapeutic approaches, such as neuromodulation or novel anti-neuropathic drugs, significant efforts have been made in the repurposing of well-known substances such as phenytoin. Although its main mechanism of action occurs at sodium channels in excitable and non-excitable cells and is well documented, how the drug affects the disturbed neuropathic interactions at the spinal cord level and how it influences morphine-induced analgesia have not been clarified, both being crucial from a clinical perspective. We demonstrated that single and repeated systemic administrations of phenytoin decreased tactile and thermal hypersensitivity in an animal model of neuropathic pain. Importantly, we observed an increase in the antinociceptive effect on thermal stimuli with repeated administrations of phenytoin. This is the first study to report that phenytoin improves morphine-induced antinociceptive effects and influences microglia/macrophage activity at the spinal cord and dorsal root ganglion levels in a neuropathic pain model. Our findings support the hypothesis that phenytoin may represent an effective strategy for neuropathic pain management in clinical practice, particularly when combination with opioids is needed.
Collapse
Affiliation(s)
- Magdalena Kocot-Kępska
- Department of Pain Research and Treatment, Jagiellonian University Medical College, 31-501 Krakow, Poland
| | - Katarzyna Pawlik
- Department of Pain Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, 31-343 Krakow, Poland
| | - Katarzyna Ciapała
- Department of Pain Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, 31-343 Krakow, Poland
| | - Wioletta Makuch
- Department of Pain Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, 31-343 Krakow, Poland
| | - Renata Zajączkowska
- Department of Interdisciplinary Intensive Care, Jagiellonian University Medical College, 30-688 Krakow, Poland
| | - Jan Dobrogowski
- Department of Pain Research and Treatment, Jagiellonian University Medical College, 31-501 Krakow, Poland
| | - Anna Przeklasa-Muszyńska
- Department of Pain Research and Treatment, Jagiellonian University Medical College, 31-501 Krakow, Poland
| | - Joanna Mika
- Department of Pain Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, 31-343 Krakow, Poland
| |
Collapse
|
11
|
Yang C, Yamaki S, Jung T, Kim B, Huyhn R, McKemy DD. Endogenous Inflammatory Mediators Produced by Injury Activate TRPV1 and TRPA1 Nociceptors to Induce Sexually Dimorphic Cold Pain That Is Dependent on TRPM8 and GFRα3. J Neurosci 2023; 43:2803-2814. [PMID: 36898840 PMCID: PMC10089246 DOI: 10.1523/jneurosci.2303-22.2023] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 02/06/2023] [Accepted: 03/04/2023] [Indexed: 03/12/2023] Open
Abstract
The detection of environmental temperatures is critical for survival, yet inappropriate responses to thermal stimuli can have a negative impact on overall health. The physiological effect of cold is distinct among somatosensory modalities in that it is soothing and analgesic, but also agonizing in the context of tissue damage. Inflammatory mediators produced during injury activate nociceptors to release neuropeptides, such as calcitonin gene-related peptide (CGRP) and substance P, inducing neurogenic inflammation, which further exasperates pain. Many inflammatory mediators induce sensitization to heat and mechanical stimuli but, conversely, inhibit cold responsiveness, and the identity of molecules inducing cold pain peripherally is enigmatic, as are the cellular and molecular mechanisms altering cold sensitivity. Here, we asked whether inflammatory mediators that induce neurogenic inflammation via the nociceptive ion channels TRPV1 (vanilloid subfamily of transient receptor potential channel) and TRPA1 (transient receptor potential ankyrin 1) lead to cold pain in mice. Specifically, we tested cold sensitivity in mice after intraplantar injection of lysophosphatidic acid or 4-hydroxy-2-nonenal, finding that each induces cold pain that is dependent on the cold-gated channel transient receptor potential melastatin 8 (TRPM8). Inhibition of CGRP, substance P, or toll-like receptor 4 (TLR4) signaling attenuates this phenotype, and each neuropeptide produces TRPM8-dependent cold pain directly. Further, the inhibition of CGRP or TLR4 signaling alleviates cold allodynia differentially by sex. Last, cold pain induced by both inflammatory mediators and neuropeptides requires TRPM8, as well as the neurotrophin artemin and its receptor GDNF receptor α3 (GFRα3). These results are consistent with artemin-induced cold allodynia requiring TRPM8, demonstrating that neurogenic inflammation alters cold sensitivity via localized artemin release that induces cold pain via GFRα3 and TRPM8.SIGNIFICANCE STATEMENT The cellular and molecular mechanisms that generate pain are complex with a diverse array of pain-producing molecules generated during injury that act to sensitize peripheral sensory neurons, thereby inducing pain. Here we identify a specific neuroinflammatory pathway involving the ion channel TRPM8 (transient receptor potential cation channel subfamily M member 8) and the neurotrophin receptor GFRα3 (GDNF receptor α3) that leads to cold pain, providing select targets for potential therapies for this pain modality.
Collapse
Affiliation(s)
- Chenyu Yang
- Neurobiology Section, Department of Biological Sciences, University of Southern California, Los Angeles, California 90089
- Molecular and Computational Biology Graduate Program, University of Southern California, Los Angeles, California 90089
| | - Shanni Yamaki
- Neurobiology Section, Department of Biological Sciences, University of Southern California, Los Angeles, California 90089
- Molecular and Computational Biology Graduate Program, University of Southern California, Los Angeles, California 90089
| | - Tyler Jung
- Neurobiology Section, Department of Biological Sciences, University of Southern California, Los Angeles, California 90089
| | - Brian Kim
- Neurobiology Section, Department of Biological Sciences, University of Southern California, Los Angeles, California 90089
| | - Ryan Huyhn
- Neurobiology Section, Department of Biological Sciences, University of Southern California, Los Angeles, California 90089
| | - David D McKemy
- Neurobiology Section, Department of Biological Sciences, University of Southern California, Los Angeles, California 90089
- Molecular and Computational Biology Graduate Program, University of Southern California, Los Angeles, California 90089
| |
Collapse
|
12
|
Yang C, Yamaki S, Jung T, Kim B, Huyhn R, McKemy DD. Endogenous inflammatory mediators produced by injury activate TRPV1 and TRPA1 nociceptors to induce sexually dimorphic cold pain that is dependent on TRPM8 and GFRα3. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.23.525238. [PMID: 36747719 PMCID: PMC9900806 DOI: 10.1101/2023.01.23.525238] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The detection of environmental temperatures is critical for survival, yet inappropriate responses to thermal stimuli can have a negative impact on overall health. The physiological effect of cold is distinct among somatosensory modalities in that it is soothing and analgesic, but also agonizing in the context of tissue damage. Inflammatory mediators produced during injury activate nociceptors to release neuropeptides, such as CGRP and substance P, inducing neurogenic inflammation which further exasperates pain. Many inflammatory mediators induce sensitization to heat and mechanical stimuli but, conversely, inhibit cold responsiveness, and the identity of molecules inducing cold pain peripherally is enigmatic, as are the cellular and molecular mechanisms altering cold sensitivity. Here, we asked if inflammatory mediators that induce neurogenic inflammation via the nociceptive ion channels TRPV1 and TRPA1 lead to cold pain in mice. Specifically, we tested cold sensitivity in mice after intraplantar injection of lysophosphatidic acid (LPA) or 4-hydroxy-2-nonenal (4HNE), finding each induces cold pain that is dependent on the cold-gated channel TRPM8. Inhibition of either CGRP, substance P, or toll-like receptor 4 (TLR4) signaling attenuates this phenotype, and each neuropeptide produces TRPM8-dependent cold pain directly. Further, the inhibition of CGRP or TLR4 signaling alleviates cold allodynia differentially by sex. Lastly, we find that cold pain induced by inflammatory mediators and neuropeptides requires the neurotrophin artemin and its receptor GFRα3. These results demonstrate that tissue damage alters cold sensitivity via neurogenic inflammation, likely leading to localized artemin release that induces cold pain via GFRα3 and TRPM8. Significance Statement The cellular and molecular mechanisms that generate pain are complex with a diverse array of pain-producing molecules generated during injury that act to sensitize peripheral sensory neurons, thereby inducing pain. Here we identify a specific neuroinflammatory pathway involving the ion channel TRPM8 and the neurotrophin receptor GFRα3 that leads to cold pain, providing select targets for potential therapies for this pain modality.
Collapse
Affiliation(s)
- Chenyu Yang
- Neurobiology Section, Department of Biological Sciences; University of Southern California, Los Angeles, CA 90089.,Molecular and Computational Biology Graduate Program; University of Southern California, Los Angeles, CA 90089
| | - Shanni Yamaki
- Neurobiology Section, Department of Biological Sciences; University of Southern California, Los Angeles, CA 90089.,Molecular and Computational Biology Graduate Program; University of Southern California, Los Angeles, CA 90089
| | - Tyler Jung
- Neurobiology Section, Department of Biological Sciences; University of Southern California, Los Angeles, CA 90089
| | - Brian Kim
- Neurobiology Section, Department of Biological Sciences; University of Southern California, Los Angeles, CA 90089
| | - Ryan Huyhn
- Neurobiology Section, Department of Biological Sciences; University of Southern California, Los Angeles, CA 90089
| | - David D McKemy
- Neurobiology Section, Department of Biological Sciences; University of Southern California, Los Angeles, CA 90089.,Molecular and Computational Biology Graduate Program; University of Southern California, Los Angeles, CA 90089
| |
Collapse
|
13
|
Niu M, Zhao F, Chen R, Li P, Bi L. The transient receptor potential channels in rheumatoid arthritis: Need to pay more attention. Front Immunol 2023; 14:1127277. [PMID: 36926330 PMCID: PMC10013686 DOI: 10.3389/fimmu.2023.1127277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 02/06/2023] [Indexed: 03/06/2023] Open
Abstract
Rheumatoid arthritis (RA) is characterized by the augment of vascular permeability, increased inflammatory cells infiltration, dysregulated immune cells activation, pannus formation and unbearable pain hyperalgesia. Ca2+ affect almost every aspect of cellular functions, involving cell migration, signal transduction, proliferation, and apoptosis. Transient receptor potential channels (TRPs) as a type of non-selective permeable cation channels, can regulate Ca2+ entry and intracellular Ca2+ signal in cells including immune cells and neurons. Researches have demonstrated that TRPs in the mechanisms of inflammatory diseases have achieved rapid progress, while the roles of TRPs in RA pathogenesis and pain hyperalgesia are still not well understood. To solve this problem, this review presents the evidence of TRPs on vascular endothelial cells in joint swelling, neutrophils activation and their trans-endothelial migration, as well as their bridging role in the reactive oxygen species/TRPs/Ca2+/peptidyl arginine deiminases networks in accelerating citrullinated proteins formation. It also points out the distinct functions of TRPs subfamilies expressed in the nervous systems of joints in cold hyperalgesia and neuro-inflammation mutually influenced inflammatory pain in RA. Thus, more attention could be paid on the impact of TRPs in RA and TRPs are useful in researches on the molecular mechanisms of anti-inflammation and analgesic therapeutic strategies.
Collapse
Affiliation(s)
- Mengwen Niu
- Department of Rheumatology and Immunology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Feng Zhao
- Department of Rheumatology and Immunology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Rui Chen
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Ping Li
- Department of Rheumatology and Immunology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Liqi Bi
- Department of Rheumatology and Immunology, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
14
|
Jeong SL, Zhang H, Yamaki S, Yang C, McKemy D, Lieber M, Pham P, Goodman M. Immunoglobulin somatic hypermutation in a defined biochemical system recapitulates affinity maturation and permits antibody optimization. Nucleic Acids Res 2022; 50:11738-11754. [PMID: 36321646 PMCID: PMC9723645 DOI: 10.1093/nar/gkac995] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 10/10/2022] [Accepted: 10/18/2022] [Indexed: 11/05/2022] Open
Abstract
We describe a purified biochemical system to produce monoclonal antibodies (Abs) in vitro using activation-induced deoxycytidine deaminase (AID) and DNA polymerase η (Polη) to diversify immunoglobulin variable gene (IgV) libraries within a phage display format. AID and Polη function during B-cell affinity maturation by catalyzing somatic hypermutation (SHM) of immunoglobulin variable genes (IgV) to generate high-affinity Abs. The IgV mutational motif specificities observed in vivo are conserved in vitro. IgV mutations occurred in antibody complementary determining regions (CDRs) and less frequently in framework (FW) regions. A unique feature of our system is the use of AID and Polη to perform repetitive affinity maturation on libraries reconstructed from a preceding selection step. We have obtained scFv Abs against human glucagon-like peptide-1 receptor (GLP-1R), a target in the treatment of type 2 diabetes, and VHH nanobodies targeting Fatty Acid Amide Hydrolase (FAAH), involved in chronic pain, and artemin, a neurotropic factor that regulates cold pain. A round of in vitro affinity maturation typically resulted in a 2- to 4-fold enhancement in Ab-Ag binding, demonstrating the utility of the system. We tested one of the affinity matured nanobodies and found that it reduced injury-induced cold pain in a mouse model.
Collapse
Affiliation(s)
- Soo Lim Jeong
- Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Hongyu Zhang
- Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Shanni Yamaki
- Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Chenyu Yang
- Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - David D McKemy
- Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Michael R Lieber
- Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA,Departments of Pathology, Biochemistry & Molecular Biology, and Molecular Microbiology & Immunology, Norris Comprehensive Cancer Center, University of Southern California Keck School of Medicine, Los Angeles, CA 90033, USA
| | - Phuong Pham
- Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Myron F Goodman
- To whom correspondence should be addressed. Tel: +1 213 740 5190; Fax: +1 213 821 1138;
| |
Collapse
|
15
|
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: 1.7] [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
|
16
|
Formagio ASN, Vilegas W, Volobuff CRF, Kassuya CAL, Cardoso CAL, Pereira ZV, Silva RMMF, Dos Santos Yamazaki DA, de Freitas Gauze G, Manfron J, Marangoni JA. Exploration of essential oil from Psychotria poeppigiana as an anti-hyperalgesic and anti-acetylcholinesterase agent: Chemical composition, biological activity and molecular docking. JOURNAL OF ETHNOPHARMACOLOGY 2022; 296:115220. [PMID: 35358624 DOI: 10.1016/j.jep.2022.115220] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 02/17/2022] [Accepted: 03/20/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Leaves from Psychotria poeppigiana Müll. Arg. (accepted as Palicourea tomentosa (Aubl.) Borhidi), Rubiaceae, has traditionally been used in medicine for treatments of inflammation and pain; Synonymously, Cephaelis elata for the treatment of dementia; However, few scientific studies have been evidence demonstrating this activity. AIM OF THE STUDY The aim of this study was to investigate the chemical composition of P. poeppigiana essential oil obtained from leaves (EOPP) and its antioxidant, anti-inflammatory and acetylcholinesterase (AChE) activities. Molecular docking simulations were carried out with the main constituents. MATERIALS AND METHODS EOPP (hydrodistillation) was analysed by gas chromatography-mass spectrometry (GC-MS). The fractionation of EOPP afforded germacrene D and bicyclogermacrene. The antioxidant activity of EOPP was determined by MDA assay. The inflammatory parameters were evaluated using CFA model (with paw edema, mechanical, thermal hyperalgesia, MPO and NAG) in EOPP (30, 100 and 300 mg/kg), germacrene D and bicyclogermacrene (30 mg/kg). The AChE inhibition was evaluated in rat brain structures and molecular docking simulations were carried out using Autodock v.4.3.2. RESULTS GC-MS analysis identified 19 compounds, and the major compounds were germacrene D (29.38%) and bicyclogermacrene (25.21%). EOPP exhibited high antioxidant capacity (IC50 = 12.78 ± 1.36 μg/mL). All the tested doses of EOPP and both major constituents significantly inhibited cold and mechanical hyperalgesia and significantly blocked the increase in MPO activity 24 h after the CFA injection. There was significant AChE inhibition by EOPP and germacrene D in the cerebral cortex and hippocampus (>50%). Enzyme-ligand molecular modelling showed that the major constituents of EOPP interacted differently with AChE. CONCLUSIONS The chemical compounds of the essential oil from the leaves of P. poeppigiana is based mainly on terpenes, the sesquiterpenes germacrene D (29.38%) and bicyclogermacrene (25.21%) being the major compounds. EOPP presented antioxidant, anti-inflammatory and anti-acetylcholinesterase (AChE) activities. Besides, enzyme-ligand molecular modelling showed the EOPP may act as an anti-hyperalgesic and AChE inhibitory agent. Taken together, these results might be in accordance with if folk use for pain- and inflammation-related symptoms.
Collapse
Affiliation(s)
| | - Wagner Vilegas
- Institute of Biosciences, UNESP - São Paulo State University, São Vicente, SP, Brazil.
| | | | | | | | - Zefa Valdevina Pereira
- Faculty of Biological and Environmental Sciences, Federal University of Grande Dourados, Dourados, MS, Brazil.
| | | | | | | | - Jane Manfron
- Postgraduate Program in Pharmaceutical Sciences, State University of Ponta Grossa, Ponta Grossa, PR, Brazil.
| | - Janaine Alberto Marangoni
- Faculty of Biological and Environmental Sciences, Federal University of Grande Dourados, Dourados, MS, Brazil.
| |
Collapse
|
17
|
Haghani M, Jafari M, Meftahi GH, Behzadnia MJ, Bahari Z, Salimi-Sabour E, Jangravi Z. Analgesic effects of Terminalia chebula extract are mediated by the suppression of the protein expression of nerve growth factor and nuclear factor-κB in the brain and oxidative markers following neuropathic pain in rats. Mol Biol Rep 2022; 49:10457-10467. [DOI: 10.1007/s11033-022-07870-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 08/15/2022] [Indexed: 11/24/2022]
|
18
|
Lewis CM, Griffith TN. The mechanisms of cold encoding. Curr Opin Neurobiol 2022; 75:102571. [DOI: 10.1016/j.conb.2022.102571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 03/31/2022] [Accepted: 05/06/2022] [Indexed: 11/15/2022]
|
19
|
Nazari Formagio AS, Vilegas W, Ferreira Volobuff CR, Leite Kassuya CA, Paes de Almeida V, Manfron J, Pereira ZV, Pereira Cabral MR, Sarragiotto MH. Palicourea tomentosa (Aubl.) Borhidi: Microscopy, chemical composition and the analgesic, anti-inflammatory and anti-acetylcholinesterase potential. JOURNAL OF ETHNOPHARMACOLOGY 2022; 291:115050. [PMID: 35150816 DOI: 10.1016/j.jep.2022.115050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 01/12/2022] [Accepted: 01/25/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Palicourea tomentosa (Aubl.) Borhidi (synonym Psychotria poeppigiana Müll. Arg.) leaves are used in the popular treatments of inflammation and pain; however, there are no scientific studies demonstrating their activity as the methanolic extract of P. tomentosa. AIM OF STUDY This study was undertaken to investigate the potential antioxidant, anti-acetylcholinesterase, anti-hyperalgesic, anti-nociceptive and anti-inflammatory properties, as well as the chemical composition and concentrations of constituents of the methanolic extract of P. tomentosa leaves (MEPT). The study also analyzes the micromorphology and histochemistry of leaves of P. tomentosa. MATERIALS AND METHODS The MEPT was analysed by ultra-high-pressure liquid chromatography-high resolution mass spectrometry (UHPLC-HRMS/MS). The concentrations of total phenols, flavonoids, flavonols and condensed tannin were determined. The micromorphology and histochemistry of leaves were performed using standard reagents, light and field emission scanning electron microscopy, beyond energy-dispersive X-ray spectroscopy. The antioxidant activity was evaluated for DPPH, β-carotene and MDA. The anti-inflammatory activity of MEPT (30, 100, and 300 mg/kg) was assayed in carrageenan-induced models of paw oedema, mechanical hyperalgesia (Von Frey), cold allodynia (acetone) and pleurisy in mice. The anti-nociceptive potential of MEPT (30, 100, and 300 mg/kg) was evaluated by the formalin method in mice. The anti-acetylcholinesterase properties were evaluated in vivo in four rat brain structures. RESULTS The total ion chromatogram of MEPT demonstrated two alkaloids, one coumarin, one iridoid and two terpene derivatives. The highest phenol, flavonoid, flavonol and condensed tannin concentrations were found in the extract. A comprehensive explanation of the leaf micromorphology and histochemistry was presented. MEPT was significantly inhibited by the DPPH, β-carotene and MDA models. MEPT (30, 100 and 300 mg/kg) reduced the inflammation and hyperalgesic parameters in a carrageenan model and reduced formalin-induced nociception in both phases, which were cold sensitivity and oedema formation. The oral administration of 30 and 100 mg/kg MEPT significantly inhibited AChE activity in the frontal cortex. CONCLUSION This is the first chemical and biological study performed with a P. tomentosa methanolic extract and anatomical and histochemical analysis. The present study showed that MEPT inhibited pain and inflammatory parameters contributing, at least in part, to explain the popular use of this plant as analgesic natural agent. Also, anatomical and histochemistry of leaves described in the present study provide microscopical information, which aids species identification.
Collapse
Affiliation(s)
| | - Wagner Vilegas
- Institute of Biosciences, UNESP - São Paulo State University, São Vicente, SP, Brazil
| | | | | | - Valter Paes de Almeida
- Postgraduate Program in Pharmaceutical Sciences, State University of Ponta Grossa, Ponta Grossa, PR, Brazil
| | - Jane Manfron
- Postgraduate Program in Pharmaceutical Sciences, State University of Ponta Grossa, Ponta Grossa, PR, Brazil
| | - Zefa Valdevina Pereira
- Faculty of Biological and Environmental Sciences, Federal University of Grande Dourados - UFGD, Dourados, MS, Brazil
| | | | | |
Collapse
|
20
|
Mert I, Cetinkaya A, Gurler M, Turel CA, Celik H, Torun IE, Turel I. Anti-inflammatory potential of liraglutide, a glucagon-like peptide-1 receptor agonist, in rats with peripheral acute inflammation. Inflammopharmacology 2022; 30:1093-1105. [PMID: 35412166 DOI: 10.1007/s10787-022-00978-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 03/18/2022] [Indexed: 11/30/2022]
Abstract
The present study aimed to explore the possible anti-inflammatory actions of liraglutide (LRG), a glucagon-like peptide-1 (GLP-1) receptor agonist, and to compare with tramadol (TR) or LRG, and TR combination treatment by investigating the inflammatory signs such as pain hypersensitivity, edema, and fever in carrageenan (CG)-induced acute peripheral inflammation model in rats. The levels of several biomarkers for inflammatory status, angiogenesis, and oxidative stress were also measured in inflamed tissues. CG induced inflammation in the paws of rats identified by hypersensitivities, redness, edema and fever. LRG, significantly improved the hypersensitivity to mechanical (from 4 to 35.5 g) or cold (from 5 to 44.2 s) stimuli, reduced the edema (paw mass, from 2.54 to 1.85 g), and fever (paw temperature, from 33.6 to 27.3 °C). LRG dramatically suppressed the inflammatory signs when compared to those of TR. In addition, co-administration of TR and LRG resulted in further reduction of sensitivity to mechanical and cold stimuli. Anti-inflammatory potential of LRG altered depending on their inhibitory effects in the biomarkers of inflamed paws. Consequently, the suppressive actions of LRG in the inflammation induced hypersensitivities, edema, and fever, indicating that these drugs have significant anti-inflammatory potential with anti-hypersensitivities, anti-edema, and anti-pyretic effects. LRG with anti-inflammatory actions may be a highly promising therapeutic option for the management of inflammatory conditions or inflammatory-related various diseases.
Collapse
Affiliation(s)
- Irem Mert
- Department of Pharmacology, Faculty of Medicine, Bolu Abant Izzet Baysal University, 14030, Bolu, Turkey.
| | - Ayhan Cetinkaya
- Department of Physiology, Faculty of Medicine, Bolu Abant Izzet Baysal University, Bolu, Turkey
| | - Mujgan Gurler
- Department of Internal Medicine, Faculty of Medicine, Bolu Abant Izzet Baysal University, Bolu, Turkey
| | - Canan Akünal Turel
- Department of Neurology, Faculty of Medicine, Bolu Abant Izzet Baysal University, Bolu, Turkey
| | - Humeyra Celik
- Department of Physiology, Faculty of Medicine, Bolu Abant Izzet Baysal University, Bolu, Turkey
| | - Ibrahim Ethem Torun
- Department of Physiology, Faculty of Medicine, Bolu Abant Izzet Baysal University, Bolu, Turkey
| | - Idris Turel
- Department of Pharmacology, Faculty of Medicine, Bolu Abant Izzet Baysal University, 14030, Bolu, Turkey
| |
Collapse
|
21
|
Minnema L, Gupta A, Mishra SK, Lascelles BDX. Investigating the Role of Artemin and Its Cognate Receptor, GFRα3, in Osteoarthritis Pain. Front Neurosci 2022; 16:738976. [PMID: 35153665 PMCID: PMC8829392 DOI: 10.3389/fnins.2022.738976] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 01/05/2022] [Indexed: 11/13/2022] Open
Abstract
Osteoarthritis (OA) associated pain (OA-pain) is a significant global problem. OA-pain limits limb use and mobility and is associated with widespread sensitivity. Therapeutic options are limited, and the available options are often associated with adverse effects. The lack of therapeutic options is partly due to a lack of understanding of clinically relevant underlying neural mechanisms of OA-pain. In previous work in naturally occurring OA-pain in dogs, we identified potential signaling molecules (artemin/GFRα3) that were upregulated. Here, we use multiple approaches, including cellular, mouse genetic, immunological suppression in a mouse model of OA, and clinically relevant measures of sensitivity and limb use to explore the functional role of artemin/GFRα3 signaling in OA-pain. We found the monoiodoacetate (MIA)-induced OA-pain in mice is associated with decreased limb use and hypersensitivity. Exogenous artemin induces mechanical, heat, and cold hypersensitivity, and systemic intraperitoneal anti-artemin monoclonal antibody administration reverses this hypersensitivity and restores limb use in mice with MIA-induced OA-pain. An artemin receptor GFRα3 expression is increased in sensory neurons in the MIA model. Our results provide a molecular basis of arthritis pain linked with artemin/GFRα3 signaling and indicate that further work is warranted to investigate the neuronal plasticity and the pathways that drive pain in OA.
Collapse
Affiliation(s)
- Laura Minnema
- Translational Research in Pain Program, Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| | - Ankita Gupta
- Translational Research in Pain Program, Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| | - Santosh K. Mishra
- Department of Molecular and Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
- Comparative Pain Research and Education Center, North Carolina State University, Raleigh, NC, United States
- *Correspondence: Santosh K. Mishra,
| | - B. Duncan X. Lascelles
- Translational Research in Pain Program, Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
- Comparative Pain Research and Education Center, North Carolina State University, Raleigh, NC, United States
- Thurston Arthritis Center, UNC School of Medicine, Chapel Hill, NC, United States
- B. Duncan X. Lascelles,
| |
Collapse
|
22
|
Boakye PA, Tang SJ, Smith PA. Mediators of Neuropathic Pain; Focus on Spinal Microglia, CSF-1, BDNF, CCL21, TNF-α, Wnt Ligands, and Interleukin 1β. FRONTIERS IN PAIN RESEARCH 2021; 2:698157. [PMID: 35295524 PMCID: PMC8915739 DOI: 10.3389/fpain.2021.698157] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 07/14/2021] [Indexed: 01/04/2023] Open
Abstract
Intractable neuropathic pain is a frequent consequence of nerve injury or disease. When peripheral nerves are injured, damaged axons undergo Wallerian degeneration. Schwann cells, mast cells, fibroblasts, keratinocytes and epithelial cells are activated leading to the generation of an "inflammatory soup" containing cytokines, chemokines and growth factors. These primary mediators sensitize sensory nerve endings, attract macrophages, neutrophils and lymphocytes, alter gene expression, promote post-translational modification of proteins, and alter ion channel function in primary afferent neurons. This leads to increased excitability and spontaneous activity and the generation of secondary mediators including colony stimulating factor 1 (CSF-1), chemokine C-C motif ligand 21 (CCL-21), Wnt3a, and Wnt5a. Release of these mediators from primary afferent neurons alters the properties of spinal microglial cells causing them to release tertiary mediators, in many situations via ATP-dependent mechanisms. Tertiary mediators such as BDNF, tumor necrosis factor α (TNF-α), interleukin 1β (IL-1β), and other Wnt ligands facilitate the generation and transmission of nociceptive information by increasing excitatory glutamatergic transmission and attenuating inhibitory GABA and glycinergic transmission in the spinal dorsal horn. This review focusses on activation of microglia by secondary mediators, release of tertiary mediators from microglia and a description of their actions in the spinal dorsal horn. Attention is drawn to the substantial differences in the precise roles of various mediators in males compared to females. At least 25 different mediators have been identified but the similarity of their actions at sensory nerve endings, in the dorsal root ganglia and in the spinal cord means there is considerable redundancy in the available mechanisms. Despite this, behavioral studies show that interruption of the actions of any single mediator can relieve signs of pain in experimental animals. We draw attention this paradox. It is difficult to explain how inactivation of one mediator can relieve pain when so many parallel pathways are available.
Collapse
Affiliation(s)
- Paul A. Boakye
- Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, United States
| | - Shao-Jun Tang
- Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, United States
| | - Peter A. Smith
- Neuroscience and Mental Health Institute and Department of Pharmacology, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
23
|
Mihai DP, Ungurianu A, Ciotu CI, Fischer MJM, Olaru OT, Nitulescu GM, Andrei C, Zbarcea CE, Zanfirescu A, Seremet OC, Chirita C, Negres S. Effects of Venlafaxine, Risperidone and Febuxostat on Cuprizone-Induced Demyelination, Behavioral Deficits and Oxidative Stress. Int J Mol Sci 2021; 22:7183. [PMID: 34281235 PMCID: PMC8268376 DOI: 10.3390/ijms22137183] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 06/25/2021] [Accepted: 06/28/2021] [Indexed: 12/27/2022] Open
Abstract
Multiple sclerosis (MS) is a demyelinating, autoimmune disease that affects a large number of young adults. Novel therapies for MS are needed considering the efficiency and safety limitations of current treatments. In our study, we investigated the effects of venlafaxine (antidepressant, serotonin-norepinephrine reuptake inhibitor), risperidone (atypical antipsychotic) and febuxostat (gout medication, xanthine oxidase inhibitor) in the cuprizone mouse model of acute demyelination, hypothesizing an antagonistic effect on TRPA1 calcium channels. Cuprizone and drugs were administered to C57BL6/J mice for five weeks and locomotor activity, motor performance and cold sensitivity were assessed. Mice brains were harvested for histological staining and assessment of oxidative stress markers. Febuxostat and metabolites of venlafaxine (desvenlafaxine) and risperidone (paliperidone) were tested for TRPA1 antagonistic activity. Following treatment, venlafaxine and risperidone significantly improved motor performance and sensitivity to a cold stimulus. All administered drugs ameliorated the cuprizone-induced deficit of superoxide dismutase activity. Desvenlafaxine and paliperidone showed no activity on TRPA1, while febuxostat exhibited agonistic activity at high concentrations. Our findings indicated that all three drugs offered some protection against the effects of cuprizone-induced demyelination. The agonistic activity of febuxostat can be of potential use for discovering novel TRPA1 ligands.
Collapse
Affiliation(s)
- Dragos Paul Mihai
- Faculty of Pharmacy, “Carol Davila”, University of Medicine and Pharmacy, 020956 Bucharest, Romania; (D.P.M.); (O.T.O.); (G.M.N.); (C.A.); (C.E.Z.); (A.Z.); (O.C.S.); (C.C.); (S.N.)
| | - Anca Ungurianu
- Faculty of Pharmacy, “Carol Davila”, University of Medicine and Pharmacy, 020956 Bucharest, Romania; (D.P.M.); (O.T.O.); (G.M.N.); (C.A.); (C.E.Z.); (A.Z.); (O.C.S.); (C.C.); (S.N.)
| | - Cosmin I. Ciotu
- Center for Physiology and Pharmacology, Institute of Physiology, Medical University of Vienna, 1090 Vienna, Austria; (C.I.C.); (M.J.M.F.)
| | - Michael J. M. Fischer
- Center for Physiology and Pharmacology, Institute of Physiology, Medical University of Vienna, 1090 Vienna, Austria; (C.I.C.); (M.J.M.F.)
| | - Octavian Tudorel Olaru
- Faculty of Pharmacy, “Carol Davila”, University of Medicine and Pharmacy, 020956 Bucharest, Romania; (D.P.M.); (O.T.O.); (G.M.N.); (C.A.); (C.E.Z.); (A.Z.); (O.C.S.); (C.C.); (S.N.)
| | - George Mihai Nitulescu
- Faculty of Pharmacy, “Carol Davila”, University of Medicine and Pharmacy, 020956 Bucharest, Romania; (D.P.M.); (O.T.O.); (G.M.N.); (C.A.); (C.E.Z.); (A.Z.); (O.C.S.); (C.C.); (S.N.)
| | - Corina Andrei
- Faculty of Pharmacy, “Carol Davila”, University of Medicine and Pharmacy, 020956 Bucharest, Romania; (D.P.M.); (O.T.O.); (G.M.N.); (C.A.); (C.E.Z.); (A.Z.); (O.C.S.); (C.C.); (S.N.)
| | - Cristina Elena Zbarcea
- Faculty of Pharmacy, “Carol Davila”, University of Medicine and Pharmacy, 020956 Bucharest, Romania; (D.P.M.); (O.T.O.); (G.M.N.); (C.A.); (C.E.Z.); (A.Z.); (O.C.S.); (C.C.); (S.N.)
| | - Anca Zanfirescu
- Faculty of Pharmacy, “Carol Davila”, University of Medicine and Pharmacy, 020956 Bucharest, Romania; (D.P.M.); (O.T.O.); (G.M.N.); (C.A.); (C.E.Z.); (A.Z.); (O.C.S.); (C.C.); (S.N.)
| | - Oana Cristina Seremet
- Faculty of Pharmacy, “Carol Davila”, University of Medicine and Pharmacy, 020956 Bucharest, Romania; (D.P.M.); (O.T.O.); (G.M.N.); (C.A.); (C.E.Z.); (A.Z.); (O.C.S.); (C.C.); (S.N.)
| | - Cornel Chirita
- Faculty of Pharmacy, “Carol Davila”, University of Medicine and Pharmacy, 020956 Bucharest, Romania; (D.P.M.); (O.T.O.); (G.M.N.); (C.A.); (C.E.Z.); (A.Z.); (O.C.S.); (C.C.); (S.N.)
| | - Simona Negres
- Faculty of Pharmacy, “Carol Davila”, University of Medicine and Pharmacy, 020956 Bucharest, Romania; (D.P.M.); (O.T.O.); (G.M.N.); (C.A.); (C.E.Z.); (A.Z.); (O.C.S.); (C.C.); (S.N.)
| |
Collapse
|
24
|
Kotliarova A, Sidorova YA. Glial Cell Line-Derived Neurotrophic Factor Family Ligands, Players at the Interface of Neuroinflammation and Neuroprotection: Focus Onto the Glia. Front Cell Neurosci 2021; 15:679034. [PMID: 34220453 PMCID: PMC8250866 DOI: 10.3389/fncel.2021.679034] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 05/21/2021] [Indexed: 12/25/2022] Open
Abstract
Well-known effects of neurotrophic factors are related to supporting the survival and functioning of various neuronal populations in the body. However, these proteins seem to also play less well-documented roles in glial cells, thus, influencing neuroinflammation. This article summarizes available data on the effects of glial cell line derived neurotrophic factor (GDNF) family ligands (GFLs), proteins providing trophic support to dopaminergic, sensory, motor and many other neuronal populations, in non-neuronal cells contributing to the development and maintenance of neuropathic pain. The paper also contains our own limited data describing the effects of small molecules targeting GFL receptors on the expression of the satellite glial marker IBA1 in dorsal root ganglia of rats with surgery- and diabetes-induced neuropathy. In our experiments activation of GFLs receptors with either GFLs or small molecule agonists downregulated the expression of IBA1 in this tissue of experimental animals. While it can be a secondary effect due to a supportive role of GFLs in neuronal cells, growing body of evidence indicates that GFL receptors are expressed in glial and peripheral immune system cells. Thus, targeting GFL receptors with either proteins or small molecules may directly suppress the activation of glial and immune system cells and, therefore, reduce neuroinflammation. As neuroinflammation is considered to be an important contributor to the process of neurodegeneration these data further support research efforts to modulate the activity of GFL receptors in order to develop disease-modifying treatments for neurodegenerative disorders and neuropathic pain that target both neuronal and glial cells.
Collapse
Affiliation(s)
- Anastasiia Kotliarova
- Laboratory of Molecular Neuroscience, Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Yulia A Sidorova
- Laboratory of Molecular Neuroscience, Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| |
Collapse
|
25
|
Serum artemin is not correlated with sensitivity within dogs with naturally occurring osteoarthritis pain. Sci Rep 2021; 11:6682. [PMID: 33758254 PMCID: PMC7988108 DOI: 10.1038/s41598-021-85976-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 03/03/2021] [Indexed: 11/14/2022] Open
Abstract
Osteoarthritis (OA) pain is associated with peripheral and central sensitization in humans and results in widespread increased sensitivity across the body. Sensitization contributes to the OA-associated pain (OAP) state. We recently identified increased levels of an endogenous neurotrophic factor, artemin (ARTN), in dogs with OAP compared to healthy pain-free controls. Circulating ARTN released from damaged tissues in OA, may play a central role in widespread sensitivity and pain. However, the relationship between ARTN and somatosensory sensitivity remains unknown. The study aimed to assess the relationship between serum ARTN concentrations and measures of sensitivity in dogs with OAP using quantitative sensory testing. We hypothesized that there would be a positive association between circulating ARTN and increased sensitivity to mechanical and thermal stimuli in dogs with OAP. We used linear and logistic regression models to assess the relationship between ARTN, sensitization, and pain within a cohort of 43 dogs with spontaneous OAP. Serum ARTN was not associated with the degree of sensitization within dogs with OAP. Further, across dogs with varying OAP severity, we did not find any association between ARTN, and clinical measures of joint pain and disability. Although a relationship between ARTN and joint pain was not ruled out.
Collapse
|
26
|
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: 1.5] [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
|
27
|
Esancy K, Dhaka A. Unveiling new mechanisms for cold sensitization. Pain 2021; 162:323-324. [PMID: 32826762 PMCID: PMC7855638 DOI: 10.1097/j.pain.0000000000002044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 08/04/2020] [Indexed: 11/26/2022]
Affiliation(s)
- Kali Esancy
- Department of Biological Structure, University of Washington, Seattle, United States
| | - Ajay Dhaka
- Department of Biological Structure, University of Washington, Seattle, United States
- Graduate Program in Neuroscience, University of Washington, Seattle, United States
| |
Collapse
|
28
|
Efficacy assessment of salicylidene salicylhydrazide in chemotherapy associated peripheral neuropathy. Eur J Pharmacol 2020; 888:173481. [DOI: 10.1016/j.ejphar.2020.173481] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 08/07/2020] [Accepted: 08/08/2020] [Indexed: 12/13/2022]
|
29
|
Mahato AK, Sidorova YA. Glial cell line-derived neurotrophic factors (GFLs) and small molecules targeting RET receptor for the treatment of pain and Parkinson's disease. Cell Tissue Res 2020; 382:147-160. [PMID: 32556722 PMCID: PMC7529621 DOI: 10.1007/s00441-020-03227-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 04/27/2020] [Indexed: 02/07/2023]
Abstract
Rearranged during transfection (RET), in complex with glial cell line-derived (GDNF) family receptor alpha (GFRα), is the canonical signaling receptor for GDNF family ligands (GFLs) expressed in both central and peripheral parts of the nervous system and also in non-neuronal tissues. RET-dependent signaling elicited by GFLs has an important role in the development, maintenance and survival of dopamine and sensory neurons. Both Parkinson's disease and neuropathic pain are devastating disorders without an available cure, and at the moment are only treated symptomatically. GFLs have been studied extensively in animal models of Parkinson's disease and neuropathic pain with remarkable outcomes. However, clinical trials with recombinant or viral vector-encoded GFL proteins have produced inconclusive results. GFL proteins are not drug-like; they have poor pharmacokinetic properties and activate multiple receptors. Targeting RET and/or GFRα with small molecules may resolve the problems associated with using GFLs as drugs and can result in the development of therapeutics for disease-modifying treatments against Parkinson's disease and neuropathic pain.
Collapse
Affiliation(s)
- Arun Kumar Mahato
- Institute of Biotechnology, HiLIFE, University of Helsinki, Viikinkaari 5D, 00014, Helsinki, Finland
| | - Yulia A Sidorova
- Institute of Biotechnology, HiLIFE, University of Helsinki, Viikinkaari 5D, 00014, Helsinki, Finland.
| |
Collapse
|
30
|
Mahato AK, Sidorova YA. RET Receptor Tyrosine Kinase: Role in Neurodegeneration, Obesity, and Cancer. Int J Mol Sci 2020; 21:ijms21197108. [PMID: 32993133 PMCID: PMC7583994 DOI: 10.3390/ijms21197108] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 09/23/2020] [Accepted: 09/24/2020] [Indexed: 02/06/2023] Open
Abstract
Rearranged during transfection (RET) is the tyrosine kinase receptor that under normal circumstances interacts with ligand at the cell surface and mediates various essential roles in a variety of cellular processes such as proliferation, differentiation, survival, migration, and metabolism. RET plays a pivotal role in the development of both peripheral and central nervous systems. RET is expressed from early stages of embryogenesis and remains expressed throughout all life stages. Mutations either activating or inhibiting RET result in several aggressive diseases, namely cancer and Hirschsprung disease. However, the physiological ligand-dependent activation of RET receptor is important for the survival and maintenance of several neuronal populations, appetite, and weight gain control, thus providing an opportunity for the development of disease-modifying therapeutics against neurodegeneration and obesity. In this review, we describe the structure of RET, its signaling, and its role in both normal conditions as well as in several disorders. We highlight the differences in the signaling and outcomes of constitutive and ligand-induced RET activation. Finally, we review the data on recently developed small molecular weight RET agonists and their potential for the treatment of various diseases.
Collapse
|
31
|
Small Molecules and Peptides Targeting Glial Cell Line-Derived Neurotrophic Factor Receptors for the Treatment of Neurodegeneration. Int J Mol Sci 2020; 21:ijms21186575. [PMID: 32911810 PMCID: PMC7554781 DOI: 10.3390/ijms21186575] [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: 07/31/2020] [Revised: 08/31/2020] [Accepted: 09/06/2020] [Indexed: 12/14/2022] Open
Abstract
Glial cell line-derived neurotrophic factor (GDNF) family ligands (GFLs) are able to promote the survival of multiple neuronal populations in the body and, therefore, hold considerable promise for disease-modifying treatments of diseases and conditions caused by neurodegeneration. Available data reveal the potential of GFLs for the therapy of Parkinson's disease, neuropathic pain and diseases caused by retinal degeneration but, also, amyotrophic lateral sclerosis and, possibly, Alzheimer's disease. Despite promising data collected in preclinical models, clinical translation of GFLs is yet to be conducted. The main reasons for the limited success of GFLs clinical development are the poor pharmacological characteristics of GFL proteins, such as the inability of GFLs to cross tissue barriers, poor diffusion in tissues, biphasic dose-response and activation of several receptors in the organism in different cell types, along with ethical limitations on patients' selection in clinical trials. The development of small molecules selectively targeting particular GFL receptors with improved pharmacokinetic properties can overcome many of the difficulties and limitations associated with the clinical use of GFL proteins. The current review lists several strategies to target the GFL receptor complex with drug-like molecules, discusses their advantages, provides an overview of available chemical scaffolds and peptides able to activate GFL receptors and describes the effects of these molecules in cultured cells and animal models.
Collapse
|
32
|
Zhu S, Li Y, Bennett S, Chen J, Weng IZ, Huang L, Xu H, Xu J. The role of glial cell line-derived neurotrophic factor family member artemin in neurological disorders and cancers. Cell Prolif 2020; 53:e12860. [PMID: 32573073 PMCID: PMC7377943 DOI: 10.1111/cpr.12860] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 05/17/2020] [Accepted: 06/02/2020] [Indexed: 12/12/2022] Open
Abstract
Artemin (ARTN) is a member of the glial cell line‐derived neurotrophic factor (GDNF) family ligands (GFLs), which encompasses family members, GDNF, neurturin (NRTN) and persephin (PSPN). ARTN is also referred to as Enovin or Neublastin, and bears structural characteristics of the TGF‐β superfamily. ARTN contains a dibasic cleavage site (RXXR) that is predicted to be cleaved by furin to yield a carboxy‐terminal 113 amino acid mature form. ARTN binds preferentially to receptor GFRα3, coupled to a receptor tyrosine kinase RET, forming a signalling complex for the regulation of intracellular pathways that affect diverse outcomes of nervous system development and homoeostasis. Standard signalling cascades activated by GFLs via RET include the phosphorylation of mitogen‐activated protein kinase or MAPK (p‐ERK, p‐p38 and p‐JNK), PI3K‐AKT and Src. Neural cell adhesion molecule (NCAM) is an alternative signalling receptor for ARTN in the presence of GFRα1, leading to activation of Fyn and FAK. Further, ARTN also interacts with heparan sulphate proteoglycan syndecan‐3 and mediates non‐RET signalling via activation of Src kinases. This review discusses the role of ARTN in spinal cord injury, neuropathic pain and other neurological disorders. Additionally, ARTN plays a role in non‐neuron tissues, such as the formation of Peyer's patch‐like structures in the lymphoid tissue of the gut. The emerging role of ARTN in cancers and therapeutic resistance to cancers is also explored. Further research is necessary to determine the function of ARTN in a tissue‐specific manner, including its signalling mechanisms, in order to improve the therapeutic potential of ARTN in human diseases.
Collapse
Affiliation(s)
- Sipin Zhu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| | - Yihe Li
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| | - Samuel Bennett
- School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| | - Junhao Chen
- School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| | - Isabel Ziwai Weng
- School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| | - Lin Huang
- School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia.,Department of Spine Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Department of Orthopaedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Huazi Xu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jiake Xu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| |
Collapse
|
33
|
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: 21] [Impact Index Per Article: 4.2] [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
|
34
|
Minnema L, Wheeler J, Enomoto M, Pitake S, Mishra SK, Lascelles BDX. Correlation of Artemin and GFRα3 With Osteoarthritis Pain: Early Evidence From Naturally Occurring Osteoarthritis-Associated Chronic Pain in Dogs. Front Neurosci 2020; 14:77. [PMID: 32116521 PMCID: PMC7031206 DOI: 10.3389/fnins.2020.00077] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 01/20/2020] [Indexed: 12/11/2022] Open
Abstract
Arthritis, including osteoarthritis (OA) and other musculoskeletal-associated pain, is a worldwide problem, however, effective drug options are limited. Several receptors, neurotransmitters, and endogenous mediators have been identified in rodent models, but the relevance of these molecules in disease-associated pain is not always clear. Artemin, a neurotrophic factor, and its receptor, glial-derived neurotrophic factor (GDNF) family receptor alpha-3 (GFRα3), have been identified as involved in pain in rodents. Their role in OA-associated pain is unknown. To explore a possible association, we analyzed tissue from naturally occurring OA in dogs to characterize the correlation with chronic pain. We used behavioral assessment, objective measures of limb use, and molecular tools to identify whether artemin and GFRα3 might be associated with OA pain. Our results using banked tissue from well-phenotyped dogs indicates that artemin/GFRα3 may play an important, and hitherto unrecognized, role in chronic OA-associated pain. Elevated serum levels of artemin from osteoarthritic humans compared to healthy individuals suggest translational relevance. Our data provide compelling evidence that the artemin/GFRα3 signaling pathway may be important in OA pain in both non-humans and humans and may ultimately lead to novel therapeutics.
Collapse
Affiliation(s)
- Laura Minnema
- Translational Research in Pain Program, Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States.,Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| | - Joshua Wheeler
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States.,Comparative Pain Research and Education Center, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| | - Masataka Enomoto
- Translational Research in Pain Program, Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States.,Comparative Pain Research and Education Center, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| | - Saumitra Pitake
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| | - Santosh K Mishra
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States.,Comparative Pain Research and Education Center, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| | - B Duncan X Lascelles
- Translational Research in Pain Program, Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States.,Comparative Pain Research and Education Center, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States.,Thurston Arthritis Research Center, UNC School of Medicine, Chapel Hill, NC, United States.,Center for Translational Pain Research, Department of Anesthesiology, Duke University, Durham, NC, United States
| |
Collapse
|
35
|
Dominant Role of the Gut Microbiota in Chemotherapy Induced Neuropathic Pain. Sci Rep 2019; 9:20324. [PMID: 31889131 PMCID: PMC6937259 DOI: 10.1038/s41598-019-56832-x] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 12/10/2019] [Indexed: 01/07/2023] Open
Abstract
Chemotherapy induced peripheral neuropathy (CIPN), a toxic side effect of some cancer treatments, negatively impacts patient outcomes and drastically reduces survivor’s quality of life (QOL). Uncovering the mechanisms driving chemotherapy-induced CIPN is urgently needed to facilitate the development of effective treatments, as currently there are none. Observing that C57BL/6 (B6) and 129SvEv (129) mice are respectively sensitive and resistant to Paclitaxel-induced pain, we investigated the involvement of the gut microbiota in this extreme phenotypic response. Reciprocal gut microbiota transfers between B6 and 129 mice as well as antibiotic depletion causally linked gut microbes to Paclitaxel-induced pain sensitivity and resistance. Microglia proliferated in the spinal cords of Paclitaxel treated mice harboring the pain-sensitive B6 microbiota but not the pain-resistant 129 microbiota, which exhibited a notable absence of infiltrating immune cells. Paclitaxel decreased the abundance of Akkermansia muciniphila, which could compromise barrier integrity resulting in systemic exposure to bacterial metabolites and products – that acting via the gut-immune-brain axis – could result in altered brain function. Other bacterial taxa that consistently associated with both bacteria and pain as well as microglia and pain were identified, lending support to our hypothesis that microglia are causally involved in CIPN, and that gut bacteria are drivers of this phenotype.
Collapse
|
36
|
Cobos EJ, Nickerson CA, Gao F, Chandran V, Bravo-Caparrós I, González-Cano R, Riva P, Andrews NA, Latremoliere A, Seehus CR, Perazzoli G, Nieto FR, Joller N, Painter MW, Ma CHE, Omura T, Chesler EJ, Geschwind DH, Coppola G, Rangachari M, Woolf CJ, Costigan M. Mechanistic Differences in Neuropathic Pain Modalities Revealed by Correlating Behavior with Global Expression Profiling. Cell Rep 2019; 22:1301-1312. [PMID: 29386116 PMCID: PMC5908229 DOI: 10.1016/j.celrep.2018.01.006] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 08/23/2017] [Accepted: 01/02/2018] [Indexed: 01/23/2023] Open
Abstract
Chronic neuropathic pain is a major morbidity of neural injury, yet its mechanisms are incompletely understood. Hypersensitivity to previously non-noxious stimuli (allodynia) is a common symptom. Here, we demonstrate that the onset of cold hypersensitivity precedes tactile allodynia in a model of partial nerve injury, and this temporal divergence was associated with major differences in global gene expression in innervating dorsal root ganglia. Transcripts whose expression change correlates with the onset of cold allodynia were nociceptor related, whereas those correlating with tactile hypersensitivity were immune cell centric. Ablation of TrpV1 lineage nociceptors resulted in mice that did not acquire cold allodynia but developed normal tactile hypersensitivity, whereas depletion of macrophages or T cells reduced neuropathic tactile allodynia but not cold hypersensitivity. We conclude that neuropathic pain incorporates reactive processes of sensory neurons and immune cells, each leading to distinct forms of hypersensitivity, potentially allowing drug development targeted to each pain type.
Collapse
Affiliation(s)
- Enrique J Cobos
- Kirby Neurobiology Center, Boston Children's Hospital and Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA; Department of Pharmacology and Institute of Neuroscience, Faculty of Medicine and Biomedical Research Center, University of Granada, 18071 Granada, Spain; Biosanitary Research Institute, University Hospital Complex of Granada, 18012 Granada, Spain
| | - Chelsea A Nickerson
- Kirby Neurobiology Center, Boston Children's Hospital and Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Fuying Gao
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Vijayendran Chandran
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Pediatrics, School of Medicine, University of Florida, Gainesville, FL 32610-0296, USA
| | - Inmaculada Bravo-Caparrós
- Department of Pharmacology and Institute of Neuroscience, Faculty of Medicine and Biomedical Research Center, University of Granada, 18071 Granada, Spain
| | - Rafael González-Cano
- Kirby Neurobiology Center, Boston Children's Hospital and Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Priscilla Riva
- Kirby Neurobiology Center, Boston Children's Hospital and Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Nick A Andrews
- Kirby Neurobiology Center, Boston Children's Hospital and Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Alban Latremoliere
- Kirby Neurobiology Center, Boston Children's Hospital and Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Corey R Seehus
- Kirby Neurobiology Center, Boston Children's Hospital and Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Gloria Perazzoli
- Department of Pharmacology and Institute of Neuroscience, Faculty of Medicine and Biomedical Research Center, University of Granada, 18071 Granada, Spain; Department of Anatomy and Embryology, School of Medicine, University of Granada, 18071 Granada, Spain
| | - Francisco R Nieto
- Department of Pharmacology and Institute of Neuroscience, Faculty of Medicine and Biomedical Research Center, University of Granada, 18071 Granada, Spain; Biosanitary Research Institute, University Hospital Complex of Granada, 18012 Granada, Spain
| | - Nicole Joller
- Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Michio W Painter
- Kirby Neurobiology Center, Boston Children's Hospital and Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Chi Him Eddie Ma
- Kirby Neurobiology Center, Boston Children's Hospital and Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Takao Omura
- Kirby Neurobiology Center, Boston Children's Hospital and Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Elissa J Chesler
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | - Daniel H Geschwind
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Giovanni Coppola
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Manu Rangachari
- Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Neurosciences, Centre de recherche du CHU de Québec, Université Laval, Québec, QC, Canada; Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec, QC G1V 0A6, Canada
| | - Clifford J Woolf
- Kirby Neurobiology Center, Boston Children's Hospital and Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Michael Costigan
- Kirby Neurobiology Center, Boston Children's Hospital and Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA; Department of Anesthesia, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
37
|
Selective cold pain inhibition by targeted block of TRPM8-expressing neurons with quaternary lidocaine derivative QX-314. Commun Biol 2018; 1:53. [PMID: 30271936 PMCID: PMC6123689 DOI: 10.1038/s42003-018-0062-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 05/02/2018] [Indexed: 01/04/2023] Open
Abstract
Treatment of pain with local anesthetics leads to an unfavorable decrease in general sensory acuity due to their indiscriminate block of both pain sensing (nociceptors) and non-pain sensing nerves. However, the cell impermeant lidocaine derivative QX-314 can be selectively targeted to only nociceptors by permeation through ligand-gated cation channels. Here we show that localized injection of QX-314 with agonists for the menthol receptor TRPM8 specifically blocks cold-evoked behaviors in mice, including cold allodynia and hyperalgesia. Remarkably, cooling stimuli also promotes QX-314-mediated inhibition of cold behaviors, and can be used to block cold allodynia, while retaining relatively normal cold sensation. The effects of both agonist and thermally evoked uptake of QX-314 are TRPM8-dependent, results demonstrating an effective approach to treat localized cold pain without altering general somatosensation. Serra Ongun, Angela Sarkisian and David McKemy show that localized co-injection of lidocaine derivative QX-314 and receptor agonists is able to block cold sensitivity in mice in a targeted way, with implications for treating cold pain associated with injury and disease.
Collapse
|
38
|
Birder LA, Kullmann FA. Role of neurogenic inflammation in local communication in the visceral mucosa. Semin Immunopathol 2018; 40:261-279. [PMID: 29582112 PMCID: PMC5960632 DOI: 10.1007/s00281-018-0674-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 03/06/2018] [Indexed: 12/27/2022]
Abstract
Intense research has focused on the involvement of the nervous system in regard to cellular mechanisms underlying neurogenic inflammation in the pelvic viscera. Evidence supports the neural release of inflammatory factors, trophic factors, and neuropeptides in the initiation of inflammation. However, more recently, non-neuronal cells including epithelia, endothelial, mast cells, and paraneurons are likely important participants in nervous system functions. For example, the urinary bladder urothelial cells are emerging as key elements in the detection and transmission of both physiological and nociceptive stimuli in the lower urinary tract. There is mounting evidence that these cells are involved in sensory mechanisms and can release mediators. Further, localization of afferent nerves next to the urothelium suggests these cells may be targets for transmitters released from bladder nerves and that chemicals released by urothelial cells may alter afferent excitability. Modifications of this type of communication in a number of pathological conditions can result in altered release of epithelial-derived mediators, which can activate local sensory nerves. Taken together, these and other findings highlighted in this review suggest that neurogenic inflammation involves complex anatomical and physiological interactions among a number of cell types in the bladder wall. The specific factors and pathways that mediate inflammatory responses in both acute and chronic conditions are not well understood and need to be further examined. Elucidation of mechanisms impacting on these pathways may provide insights into the pathology of various types of disorders involving the pelvic viscera.
Collapse
Affiliation(s)
- Lori A Birder
- Department of Medicine, University of Pittsburgh School of Medicine, A 1217 Scaife Hall, 3550 Terrace Street, Pittsburgh, PA, 15261, USA.
- Department of Chemical Biology and Pharmacology, University of Pittsburgh School of Medicine, A 1217 Scaife Hall, 3550 Terrace Street, Pittsburgh, PA, 15261, USA.
| | - F Aura Kullmann
- Department of Medicine, University of Pittsburgh School of Medicine, A 1217 Scaife Hall, 3550 Terrace Street, Pittsburgh, PA, 15261, USA
| |
Collapse
|
39
|
Moore C, Gupta R, Jordt SE, Chen Y, Liedtke WB. Regulation of Pain and Itch by TRP Channels. Neurosci Bull 2018; 34:120-142. [PMID: 29282613 PMCID: PMC5799130 DOI: 10.1007/s12264-017-0200-8] [Citation(s) in RCA: 215] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 10/27/2017] [Indexed: 02/07/2023] Open
Abstract
Nociception is an important physiological process that detects harmful signals and results in pain perception. In this review, we discuss important experimental evidence involving some TRP ion channels as molecular sensors of chemical, thermal, and mechanical noxious stimuli to evoke the pain and itch sensations. Among them are the TRPA1 channel, members of the vanilloid subfamily (TRPV1, TRPV3, and TRPV4), and finally members of the melastatin group (TRPM2, TRPM3, and TRPM8). Given that pain and itch are pro-survival, evolutionarily-honed protective mechanisms, care has to be exercised when developing inhibitory/modulatory compounds targeting specific pain/itch-TRPs so that physiological protective mechanisms are not disabled to a degree that stimulus-mediated injury can occur. Such events have impeded the development of safe and effective TRPV1-modulating compounds and have diverted substantial resources. A beneficial outcome can be readily accomplished via simple dosing strategies, and also by incorporating medicinal chemistry design features during compound design and synthesis. Beyond clinical use, where compounds that target more than one channel might have a place and possibly have advantageous features, highly specific and high-potency compounds will be helpful in mechanistic discovery at the structure-function level.
Collapse
Affiliation(s)
- Carlene Moore
- Department of Neurology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Rupali Gupta
- Department of Neurology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Sven-Eric Jordt
- Department of Anesthesiology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Yong Chen
- Department of Neurology, Duke University Medical Center, Durham, NC, 27710, USA.
| | - Wolfgang B Liedtke
- Department of Neurology, Duke University Medical Center, Durham, NC, 27710, USA.
- Department of Anesthesiology, Duke University Medical Center, Durham, NC, 27710, USA.
| |
Collapse
|
40
|
Abstract
Of somatosensory modalities cold is one of the more ambiguous percepts, evoking the pleasant sensation of cooling, the stinging bite of cold pain, and welcome relief from chronic pain. Moreover, unlike the precipitous thermal thresholds for heat activation of thermosensitive afferent neurons, thresholds for cold fibers are across a range of cool to cold temperatures that spans over 30°C. Until recently, how cold produces this myriad of biologic effects was unknown. However, recent advances in our understanding of cold mechanisms at the behavioral, physiologic, and cellular level have begun to provide insights into this sensory modality. The identification of a number of ion channels that either serve as the principal detectors of a cold stimulus in the peripheral nervous system, or are part of a differential expression pattern of channels that maintain cell excitability in the cold, endows select neurons with properties that are amenable to electric signaling in the cold. This chapter highlights the current understanding of the molecules involved in cold transduction in the mammalian peripheral nervous system, as well as presenting a hypothetic model to account for the broad range of cold thermal thresholds and distinct functions of cold fibers in perception, pain, and analgesia.
Collapse
Affiliation(s)
- David D McKemy
- Section of Neurobiology, Department of Biological Sciences, University of Southern California, Los Angeles, CA, United States.
| |
Collapse
|
41
|
Hsieh YL, Kan HW, Chiang H, Lee YC, Hsieh ST. Distinct TrkA and Ret modulated negative and positive neuropathic behaviors in a mouse model of resiniferatoxin-induced small fiber neuropathy. Exp Neurol 2017; 300:87-99. [PMID: 29106982 DOI: 10.1016/j.expneurol.2017.10.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 10/21/2017] [Accepted: 10/25/2017] [Indexed: 12/18/2022]
Abstract
Neurotrophic factors and their corresponding receptors play key roles in the maintenance of different phenotypic dorsal root ganglion (DRG) neurons, the axons of which degenerate in small fiber neuropathy, leading to various neuropathic manifestations. Mechanisms underlying positive and negative symptoms of small fiber neuropathy have not been systematically explored. This study investigated the molecular basis of these seemingly paradoxical neuropathic behaviors according to the profiles of TrkA and Ret with immunohistochemical and pharmacological interventions in a mouse model of resiniferatoxin (RTX)-induced small fiber neuropathy. Mice with RTX neuropathy exhibited thermal hypoalgesia and mechanical allodynia, reduced skin innervation, and altered DRG expression profiles with decreased TrkA(+) neurons and increased Ret(+) neurons. RTX neuropathy induced the expression of activating transcription factor 3 (ATF3), and ATF3(+) neurons were colocalized with Ret but not with TrkA (P<0.001). As a neuroprotectant, 4-Methylcatechol (4MC) promoted skin reinnervation partially with correlated reversal of the neuropathic behaviors and altered neurochemical expression. Gambogic amide, a selective TrkA agonist, normalized thermal hypoalgesia, and GW441756, a TrkA kinase inhibitor, induced thermal hypoalgesia, which was already reversed by 4MC. Mechanical allodynia was reversed by a Ret kinase inhibitor, AST487, which induced thermal hyperalgesia in naïve mice. The activation of Ret signaling by XIB4035 induced mechanical allodynia and thermal hypoalgesia in RTX neuropathy mice in which the neuropathic behaviors were previously normalized by 4MC. Distinct neurotrophic factor receptors, TrkA and Ret, accounted for negative and positive neuropathic behaviors in RTX-induced small fiber neuropathy, respectively: TrkA for thermal hypoalgesia and Ret for mechanical allodynia and thermal hypoalgesia.
Collapse
Affiliation(s)
- Yu-Lin Hsieh
- Department of Anatomy, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan.
| | - Hung-Wei Kan
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
| | - Hao Chiang
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan; Department of Otology and Laryngology, Harvard Medical School, Boston, MA 02115, USA; Eaton-Peabody Laboratory, Massachusetts Eye and Ear Infirmary, Boston, MA 02114, USA
| | - Yi-Chen Lee
- Department of Anatomy, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
| | - Sung-Tsang Hsieh
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan; Department of Neurology, National Taiwan University Hospital, Taipei 10002, Taiwan; Graduate Institute of Brain and Mind Science, College of Medicine, National Taiwan University, Taipei 10051, Taiwan.
| |
Collapse
|
42
|
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: 1.0] [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
|
43
|
Role of the Excitability Brake Potassium Current I KD in Cold Allodynia Induced by Chronic Peripheral Nerve Injury. J Neurosci 2017; 37:3109-3126. [PMID: 28179555 DOI: 10.1523/jneurosci.3553-16.2017] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 01/24/2017] [Accepted: 01/26/2017] [Indexed: 11/21/2022] Open
Abstract
Cold allodynia is a common symptom of neuropathic and inflammatory pain following peripheral nerve injury. The mechanisms underlying this disabling sensory alteration are not entirely understood. In primary somatosensory neurons, cold sensitivity is mainly determined by a functional counterbalance between cold-activated TRPM8 channels and Shaker-like Kv1.1-1.2 channels underlying the excitability brake current IKD Here we studied the role of IKD in damage-triggered painful hypersensitivity to innocuous cold. We found that cold allodynia induced by chronic constriction injury (CCI) of the sciatic nerve in mice, was related to both an increase in the proportion of cold-sensitive neurons (CSNs) in DRGs contributing to the sciatic nerve, and a decrease in their cold temperature threshold. IKD density was reduced in high-threshold CSNs from CCI mice compared with sham animals, with no differences in cold-induced TRPM8-dependent current density. The electrophysiological properties and neurochemical profile of CSNs revealed an increase of nociceptive-like phenotype among neurons from CCI animals compared with sham mice. These results were validated using a mathematical model of CSNs, including IKD and TRPM8, showing that a reduction in IKD current density shifts the thermal threshold to higher temperatures and that the reduction of this current induces cold sensitivity in former cold-insensitive neurons expressing low levels of TRPM8-like current. Together, our results suggest that cold allodynia is largely due to a functional downregulation of IKD in both high-threshold CSNs and in a subpopulation of polymodal nociceptors expressing TRPM8, providing a general molecular and neural mechanism for this sensory alteration.SIGNIFICANCE STATEMENT This paper unveils the critical role of the brake potassium current IKD in damage-triggered cold allodynia. Using a well-known form of nerve injury and combining behavioral analysis, calcium imaging, patch clamping, and pharmacological tools, validated by mathematical modeling, we determined that the functional expression of IKD is reduced in sensory neurons in response to peripheral nerve damage. This downregulation not only enhances cold sensitivity of high-threshold cold thermoreceptors signaling cold discomfort, but it also transforms a subpopulation of polymodal nociceptors signaling pain into neurons activated by mild temperature drops. Our results suggest that cold allodynia is linked to a reduction of IKD in both high-threshold cold thermoreceptors and nociceptors expressing TRPM8, providing a general model for this form of cold-induced pain.
Collapse
|