1
|
Bavencoffe A, Lopez ER, Johnson KN, Tian J, Gorgun FM, Shen BQ, Domagala DM, Zhu MX, Dessauer CW, Walters ET. Widespread hyperexcitability of nociceptor somata outlasts enhanced avoidance behavior after incision injury. Pain 2025; 166:1088-1104. [PMID: 39432803 PMCID: PMC12003080 DOI: 10.1097/j.pain.0000000000003443] [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: 02/05/2024] [Accepted: 09/05/2024] [Indexed: 10/23/2024]
Abstract
ABSTRACT Nociceptors with somata in dorsal root ganglia (DRGs) readily switch from an electrically silent state to a hyperactive state of tonic, nonaccommodating, low-frequency, irregular discharge of action potentials (APs). Spontaneous activity (SA) during this state is present in vivo in rats months after spinal cord injury (SCI) and has been causally linked to SCI pain. Intrinsically generated SA and, more generally, ongoing activity (OA) are induced by various neuropathic conditions in rats, mice, and humans and are retained in nociceptor somata after dissociation and culturing, providing a powerful tool for investigating its mechanisms and functions. The present study shows that long-lasting hyperexcitability that can generate OA during modest depolarization in probable nociceptors dissociated from DRGs of male and female rats is induced by plantar incision injury. OA occurred when the soma was artificially depolarized to a level within the normal range of membrane potentials where large, transient depolarizing spontaneous fluctuations (DSFs) can approach AP threshold. This hyperexcitability persisted for at least 3 weeks, whereas behavioral indicators of affective pain-hind paw guarding and increased avoidance of a noxious substrate in an operant conflict test-persisted for 1 week or less. The most consistent electrophysiological alteration associated with OA was enhancement of DSFs. An unexpected discovery after plantar incisions was hyperexcitability in neurons from thoracic DRGs that innervate dermatomes distant from the injured tissue. Potential in vivo functions of widespread, low-frequency nociceptor OA consistent with these and other findings are to contribute to hyperalgesic priming and to drive anxiety-related hypervigilance.
Collapse
Affiliation(s)
- Alexis Bavencoffe
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston
| | - Elia R. Lopez
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston
| | - Kayla N. Johnson
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston
| | - Jinbin Tian
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston
| | - Falih M. Gorgun
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston
| | - Breanna Q. Shen
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston
| | - Drue M. Domagala
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston
| | - Michael X. Zhu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston
| | - Carmen W. Dessauer
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston
| | - Edgar T. Walters
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston
| |
Collapse
|
2
|
Hu J, Fan W, Xu Y, Li X, Zhang H, Li S, Xue L. Maladaptive changes in the homeostasis of AEA-TRPV1/CB1R induces pain-related hyperactivity of nociceptors after spinal cord injury. Cell Biosci 2025; 15:2. [PMID: 39789637 PMCID: PMC11720958 DOI: 10.1186/s13578-025-01345-6] [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: 10/30/2024] [Accepted: 01/02/2025] [Indexed: 01/12/2025] Open
Abstract
BACKGROUND Neuropathic pain resulting from spinal cord injury (SCI) is associated with persistent hyperactivity of primary nociceptors. Anandamide (AEA) has been reported to modulate neuronal excitability and synaptic transmission through activation of cannabinoid type-1 receptors (CB1Rs) and transient receptor potential vanilloid 1 (TRPV1). However, the role of AEA and these receptors in the hyperactivity of nociceptors after SCI remains unclear. RESULTS In this study, we investigated the effects of AEA and its receptors on the hyperexcitability of mouse dorsal root ganglion (DRG) neurons after SCI. Using a whole-cell patch-clamp technique, we found that the timing of SCI-induced hyperexcitability in nociceptors paralleled an increase in the endocannabinoid AEA content. The expression of TRPV1 and CB1R was also upregulated at different time points after SCI. High-dose extracellular administration of AEA increased the excitability of naive DRG neurons, leading to the transition from a rapidly accommodating (RA) hypoexcitable state to a highly excitable non-accommodating (NA) state. These AEA-induced transitions were facilitated by increased TRPV1 transcription. Pharmacological and Ca2+ imaging experiments revealed that AEA induced hyperexcitability in nociceptors after SCI via the AEA-TRPV1-Ca2+ pathway, whereas activation of CB1Rs reduced SCI-induced hyperexcitability and maintained cytosolic Ca2+ concentration ([Ca2+]cyto) at low levels in the early stages of SCI. As the AEA and TRPV1 levels increased after SCI, adaptive neuroprotection transitioned to a maladaptive hyperactive state, leading to sustained pain. CONCLUSIONS Taken together, this study provides new insights into how endocannabinoids regulate nociceptor activity after SCI, offering potential targets for the treatment of neuropathic pain.
Collapse
Affiliation(s)
- JiaQi Hu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200438, People's Republic of China
- Department of Physiology and Neurobiology, School of Life Sciences, Fudan University, Shanghai, 200438, People's Republic of China
- Center for Rehabilitation Medicine, Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, People's Republic of China
| | - WenYong Fan
- Department of Physiology and Neurobiology, School of Life Sciences, Fudan University, Shanghai, 200438, People's Republic of China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration, Ministry of Education, Department of Orthopedics, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200092, People's Republic of China
| | - Yue Xu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200438, People's Republic of China
- Department of Physiology and Neurobiology, School of Life Sciences, Fudan University, Shanghai, 200438, People's Republic of China
| | - XiaoFei Li
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200438, People's Republic of China
- Department of Physiology and Neurobiology, School of Life Sciences, Fudan University, Shanghai, 200438, People's Republic of China
| | - HaoYang Zhang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200438, People's Republic of China
- Department of Physiology and Neurobiology, School of Life Sciences, Fudan University, Shanghai, 200438, People's Republic of China
| | - Shun Li
- Center for Rehabilitation Medicine, Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, People's Republic of China
| | - Lei Xue
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200438, People's Republic of China.
- Department of Physiology and Neurobiology, School of Life Sciences, Fudan University, Shanghai, 200438, People's Republic of China.
- Research Institute of Intelligent Complex Systems, Fudan University, Shanghai, 200433, People's Republic of China.
| |
Collapse
|
3
|
Dangi A, Sharma SS. Pharmacological agents targeting transient receptor potential (TRP) channels in neuropathic pain: Preclinical and clinical status. Eur J Pharmacol 2024; 980:176845. [PMID: 39067564 DOI: 10.1016/j.ejphar.2024.176845] [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: 04/19/2024] [Revised: 06/19/2024] [Accepted: 07/23/2024] [Indexed: 07/30/2024]
Abstract
Neuropathic pain generally affects 7-10% population worldwide and an estimated ∼1 in every 20 individuals in western countries suffer and burden to society. The most limiting factor with existing therapies includes dose escalation issues, off-target side effects and poor translation of randomized trials into clinical practice. Neuropathic pain is a broad term that comprises direct injury/damage to the central and/or peripheral nervous system, leads to maladaptive changes in neuronal as well as in non-neuronal cells, which further contributes to the spontaneous pain, sensory and motor deficit along with altered sensitivity towards the noxious as well as non-noxious stimulus. Transient receptor potential (TRP) channels are polymodal, non-specific cation channels that operate as biosensors to various mechanical and chemical stimuli, including hyperosmolarity, shear stress, heat, mechanical stretch, extracellular ATP, and other products of inflammation. Modulation of these channels leads to various physiological and pathophysiological manifestations at molecular and cellular levels, leading to diseases including neuropathic pain. There are several molecules targeting TRP channels for neuropathic pain in pre-clinical studies, clinical trials and in the market. This review highlights the critical involvement of various pharmacological modulators for TRP channels targeting neuropathic pain and their possible outcomes to harness the therapeutic potential of TRP channels.
Collapse
Affiliation(s)
- Ashish Dangi
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Mohali, Punjab, India
| | - Shyam Sunder Sharma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Mohali, Punjab, India.
| |
Collapse
|
4
|
Gao N, Li M, Wang W, Liu Z, Guo Y. The dual role of TRPV1 in peripheral neuropathic pain: pain switches caused by its sensitization or desensitization. Front Mol Neurosci 2024; 17:1400118. [PMID: 39315294 PMCID: PMC11417043 DOI: 10.3389/fnmol.2024.1400118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 08/12/2024] [Indexed: 09/25/2024] Open
Abstract
The transient receptor potential vanilloid 1 (TRPV1) channel plays a dual role in peripheral neuropathic pain (NeuP) by acting as a "pain switch" through its sensitization and desensitization. Hyperalgesia, commonly resulting from tissue injury or inflammation, involves the sensitization of TRPV1 channels, which modulates sensory transmission from primary afferent nociceptors to spinal dorsal horn neurons. In chemotherapy-induced peripheral neuropathy (CIPN), TRPV1 is implicated in neuropathic pain mechanisms due to its interaction with ion channels, neurotransmitter signaling, and oxidative stress. Sensitization of TRPV1 in dorsal root ganglion neurons contributes to CIPN development, and inhibition of TRPV1 channels can reduce chemotherapy-induced mechanical hypersensitivity. In diabetic peripheral neuropathy (DPN), TRPV1 is involved in pain modulation through pathways including reactive oxygen species and cytokine production. TRPV1's interaction with TRPA1 channels further influences chronic pain onset and progression. Therapeutically, capsaicin, a TRPV1 agonist, can induce analgesia through receptor desensitization, while TRPV1 antagonists and siRNA targeting TRPV1 show promise in preclinical studies. Cannabinoid modulation of TRPV1 provides another potential pathway for alleviating neuropathic pain. This review summarizes recent preclinical research on TRPV1 in association with peripheral NeuP.
Collapse
Affiliation(s)
- Ning Gao
- Department of Acupuncture and Moxibustion, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Meng Li
- Department of Gastroenterology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Weiming Wang
- Department of Acupuncture and Moxibustion, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhen Liu
- Department of Gastroenterology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yufeng Guo
- Department of Acupuncture and Moxibustion, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
5
|
Socała K, Jakubiec M, Abram M, Mlost J, Starowicz K, Kamiński RM, Ciepiela K, Andres-Mach M, Zagaja M, Metcalf CS, Zawadzki P, Wlaź P, Kamiński K. TRPV1 channel in the pathophysiology of epilepsy and its potential as a molecular target for the development of new antiseizure drug candidates. Prog Neurobiol 2024; 240:102634. [PMID: 38834133 DOI: 10.1016/j.pneurobio.2024.102634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 04/26/2024] [Accepted: 05/28/2024] [Indexed: 06/06/2024]
Abstract
Identification of transient receptor potential cation channel, subfamily V member 1 (TRPV1), also known as capsaicin receptor, in 1997 was a milestone achievement in the research on temperature sensation and pain signalling. Very soon after it became evident that TRPV1 is implicated in a wide array of physiological processes in different peripheral tissues, as well as in the central nervous system, and thereby could be involved in the pathophysiology of numerous diseases. Increasing evidence suggests that modulation of TRPV1 may also affect seizure susceptibility and epilepsy. This channel is localized in brain regions associated with seizures and epilepsy, and its overexpression was found both in animal models of seizures and in brain samples from epileptic patients. Moreover, modulation of TRPV1 on non-neuronal cells (microglia, astrocytes, and/or peripheral immune cells) may have an impact on the neuroinflammatory processes that play a role in epilepsy and epileptogenesis. In this paper, we provide a comprehensive and critical overview of currently available data on TRPV1 as a possible molecular target for epilepsy management, trying to identify research gaps and future directions. Overall, several converging lines of evidence implicate TRPV1 channel as a potentially attractive target in epilepsy research but more studies are needed to exploit the possible role of TRPV1 in seizures/epilepsy and to evaluate the value of TRPV1 ligands as candidates for new antiseizure drugs.
Collapse
Affiliation(s)
- Katarzyna Socała
- Department of Animal Physiology and Pharmacology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Akademicka 19, Lublin PL 20-033, Poland.
| | - Marcin Jakubiec
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Cracow PL 30-688, Poland
| | - Michał Abram
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Cracow PL 30-688, Poland
| | - Jakub Mlost
- Department of Neurochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, Cracow PL 31-343, Poland
| | - Katarzyna Starowicz
- Department of Neurochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, Cracow PL 31-343, Poland
| | - Rafał M Kamiński
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Cracow PL 30-688, Poland
| | - Katarzyna Ciepiela
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Cracow PL 30-688, Poland; Selvita S.A., Bobrzyńskiego 14, Cracow PL 30-348, Poland
| | - Marta Andres-Mach
- Department of Experimental Pharmacology, Institute of Rural Health, Jaczewskiego 2, Lublin PL 20-090, Poland
| | - Mirosław Zagaja
- Department of Experimental Pharmacology, Institute of Rural Health, Jaczewskiego 2, Lublin PL 20-090, Poland
| | - Cameron S Metcalf
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT 84112, USA
| | - Przemysław Zawadzki
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Cracow PL 30-688, Poland
| | - Piotr Wlaź
- Department of Animal Physiology and Pharmacology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Akademicka 19, Lublin PL 20-033, Poland
| | - Krzysztof Kamiński
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Cracow PL 30-688, Poland
| |
Collapse
|
6
|
Bavencoffe A, Zhu MY, Neerukonda SV, Johnson KN, Dessauer CW, Walters ET. Induction of long-term hyperexcitability by memory-related cAMP signaling in isolated nociceptor cell bodies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.13.603393. [PMID: 39071414 PMCID: PMC11275899 DOI: 10.1101/2024.07.13.603393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Persistent hyperactivity of nociceptors is known to contribute significantly to long-lasting sensitization and ongoing pain in many clinical conditions. It is often assumed that nociceptor hyperactivity is mainly driven by continuing stimulation from inflammatory mediators. We have tested an additional possibility: that persistent increases in excitability promoting hyperactivity can be induced by a prototypical cellular signaling pathway long known to induce late-phase long-term potentiation (LTP) of synapses in brain regions involved in memory formation. This cAMP-PKA-CREB-gene transcription-protein synthesis pathway was tested using whole-cell current clamp methods on small dissociated sensory neurons (primarily nociceptors) from dorsal root ganglia (DRGs) excised from previously uninjured ("naïve") rats. Six-hour treatment with the specific Gαs-coupled 5-HT4 receptor agonist, prucalopride, or with the adenylyl cyclase activator, forskolin, induced long-term hyperexcitability (LTH) in DRG neurons that manifested 12-24 hours later as action potential (AP) discharge (ongoing activity, OA) during artificial depolarization to -45 mV, a membrane potential that is normally subthreshold for AP generation. Prucalopride treatment also induced significant long-lasting depolarization of resting membrane potential (from -69 to -66 mV), enhanced depolarizing spontaneous fluctuations (DSFs) of membrane potential, and indications of reduced AP threshold and rheobase. LTH was prevented by co-treatment of prucalopride with inhibitors of PKA, CREB, gene transcription, and protein synthesis. As in the induction of synaptic memory, many other cellular signals are likely to be involved. However, the discovery that this prototypical memory induction pathway can induce nociceptor LTH, along with reports that cAMP signaling and CREB activity in DRGs can induce hyperalgesic priming, suggest that early, temporary, cAMP-induced transcriptional and translational mechanisms can induce nociceptor LTH that might last for long periods. An interesting possibility is that these mechanisms can also be reactivated by re-exposure to inflammatory mediators such as serotonin during subsequent challenges to bodily integrity, "reconsolidating" the cellular memory and thereby extending the duration of persistent nociceptor hyperexcitability.
Collapse
Affiliation(s)
- Alexis Bavencoffe
- Department of Integrative Biology and Pharmacology, McGovern Medical School at UTHealth, Houston, Texas, USA 77030
| | - Michael Y. Zhu
- Department of Integrative Biology and Pharmacology, McGovern Medical School at UTHealth, Houston, Texas, USA 77030
| | - Sanjay V. Neerukonda
- Medical Scientist Training Program, McGovern Medical School at UTHealth, Houston, Texas, USA 77030
| | - Kayla N. Johnson
- Department of Integrative Biology and Pharmacology, McGovern Medical School at UTHealth, Houston, Texas, USA 77030
| | - Carmen W. Dessauer
- Department of Integrative Biology and Pharmacology, McGovern Medical School at UTHealth, Houston, Texas, USA 77030
| | - Edgar T. Walters
- Department of Integrative Biology and Pharmacology, McGovern Medical School at UTHealth, Houston, Texas, USA 77030
| |
Collapse
|
7
|
Bavencoffe A, Zhu MY, Neerukonda SV, Johnson KN, Dessauer CW, Walters ET. Induction of long-term hyperexcitability by memory-related cAMP signaling in isolated nociceptor cell bodies. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2024; 16:100166. [PMID: 39399224 PMCID: PMC11470187 DOI: 10.1016/j.ynpai.2024.100166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/10/2024] [Accepted: 09/11/2024] [Indexed: 10/15/2024]
Abstract
Persistent hyperactivity of nociceptors is known to contribute significantly to long-lasting sensitization and ongoing pain in many clinical conditions. It is often assumed that nociceptor hyperactivity is mainly driven by continuing stimulation from inflammatory mediators. We have tested an additional possibility: that persistent increases in excitability promoting hyperactivity can be induced by a prototypical cellular signaling pathway long known to induce late-phase long-term potentiation (LTP) of synapses in brain regions involved in memory formation. This cAMP-PKA-CREB-gene transcription-protein synthesis pathway was tested using whole-cell current clamp methods on small dissociated sensory neurons (primarily nociceptors) from dorsal root ganglia (DRGs) excised from previously uninjured ("naïve") male rats. Six-hour treatment with the specific Gαs-coupled 5-HT4 receptor agonist, prucalopride, or with the adenylyl cyclase activator forskolin induced long-term hyperexcitability (LTH) in DRG neurons that manifested 12-24 h later as action potential (AP) discharge (ongoing activity, OA) during artificial depolarization to -45 mV, a membrane potential that is normally subthreshold for AP generation. Prucalopride treatment also induced significant long-lasting depolarization of resting membrane potential (from -69 to -66 mV), enhanced depolarizing spontaneous fluctuations (DSFs) of membrane potential, and produced trends for reduced AP threshold and rheobase. LTH was prevented by co-treatment of prucalopride with inhibitors of PKA, CREB, gene transcription, or protein synthesis. As in the induction of synaptic memory, many other cellular signals are likely to be involved. However, the discovery that this prototypical memory induction pathway can induce nociceptor LTH, along with reports that cAMP signaling and CREB activity in DRGs can induce hyperalgesic priming, suggest that early, temporary, cAMP-induced transcriptional and translational mechanisms can induce nociceptor LTH that might last for long periods. The present results also raise the question of whether reactivation of primed signaling mechanisms by re-exposure to inflammatory mediators linked to cAMP synthesis during subsequent challenges to bodily integrity can "reconsolidate" nociceptor memory, extending the duration of persistent hyperexcitability.
Collapse
Affiliation(s)
- Alexis Bavencoffe
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - Michael Y. Zhu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - Sanjay V. Neerukonda
- Medical Scientist Training Program, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Kayla N. Johnson
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - Carmen W. Dessauer
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - Edgar T. Walters
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| |
Collapse
|
8
|
Maximiano TKE, Carneiro JA, Fattori V, Verri WA. TRPV1: Receptor structure, activation, modulation and role in neuro-immune interactions and pain. Cell Calcium 2024; 119:102870. [PMID: 38531262 DOI: 10.1016/j.ceca.2024.102870] [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: 11/30/2023] [Revised: 03/06/2024] [Accepted: 03/06/2024] [Indexed: 03/28/2024]
Abstract
In the 1990s, the identification of a non-selective ion channel, especially responsive to capsaicin, revolutionized the studies of somatosensation and pain that were to follow. The TRPV1 channel is expressed mainly in neuronal cells, more specifically, in sensory neurons responsible for the perception of noxious stimuli. However, its presence has also been detected in other non-neuronal cells, such as immune cells, β- pancreatic cells, muscle cells and adipocytes. Activation of the channel occurs in response to a wide range of stimuli, such as noxious heat, low pH, gasses, toxins, endocannabinoids, lipid-derived endovanilloid, and chemical agents, such as capsaicin and resiniferatoxin. This activation results in an influx of cations through the channel pore, especially calcium. Intracellular calcium triggers different responses in sensory neurons. Dephosphorylation of the TRPV1 channel leads to its desensitization, which disrupts its function, while its phosphorylation increases the channel's sensitization and contributes to the channel's rehabilitation after desensitization. Kinases, phosphoinositides, and calmodulin are the main signaling pathways responsible for the channel's regulation. Thus, in this review we provide an overview of TRPV1 discovery, its tissue expression as well as on the mechanisms by which TRPV1 activation (directly or indirectly) induces pain in different disease models.
Collapse
Affiliation(s)
- Thaila Kawane Euflazio Maximiano
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Center of Biological Sciences, Londrina State University, Londrina, Paraná, Brazil
| | - Jessica Aparecida Carneiro
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Center of Biological Sciences, Londrina State University, Londrina, Paraná, Brazil
| | - Victor Fattori
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital-Harvard Medical School, Karp Research Building, 300 Longwood Ave, 02115, Boston, Massachusetts, United States.
| | - Waldiceu A Verri
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Center of Biological Sciences, Londrina State University, Londrina, Paraná, Brazil.
| |
Collapse
|
9
|
Tian J, Bavencoffe AG, Zhu MX, Walters ET. Readiness of nociceptor cell bodies to generate spontaneous activity results from background activity of diverse ion channels and high input resistance. Pain 2024; 165:893-907. [PMID: 37862056 PMCID: PMC10950548 DOI: 10.1097/j.pain.0000000000003091] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 08/09/2023] [Indexed: 10/21/2023]
Abstract
ABSTRACT Nociceptor cell bodies generate "spontaneous" discharge that can promote ongoing pain in persistent pain conditions. Little is known about the underlying mechanisms. Recordings from nociceptor cell bodies (somata) dissociated from rodent and human dorsal root ganglia have shown that previous pain in vivo is associated with low-frequency discharge controlled by irregular depolarizing spontaneous fluctuations of membrane potential (DSFs), likely produced by transient inward currents across the somal input resistance. Using mouse nociceptors, we show that DSFs are associated with high somal input resistance over a wide range of membrane potentials, including depolarized levels where DSFs approach action potential (AP) threshold. Input resistance and both the amplitude and frequency of DSFs were increased in neurons exhibiting spontaneous activity. Ion substitution experiments indicated that the depolarizing phase of DSFs is generated by spontaneous opening of channels permeable to Na + or Ca 2+ and that Ca 2+ -permeable channels are especially important for larger DSFs. Partial reduction of the amplitude or frequency of DSFs by perfusion of pharmacological inhibitors indicated small but significant contributions from Nav1.7, Nav1.8, TRPV1, TRPA1, TRPM4, and N-type Ca 2+ channels. Less specific blockers suggested a contribution from NALCN channels, and global knockout suggested a role for Nav1.9. The combination of high somal input resistance plus background activity of diverse ion channels permeable to Na + or Ca 2+ produces DSFs that are poised to reach AP threshold if resting membrane potential depolarizes, AP threshold decreases, or DSFs become enhanced-all of which can occur under painful neuropathic and inflammatory conditions.
Collapse
Affiliation(s)
- Jinbin Tian
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston
| | - Alexis G. Bavencoffe
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston
| | - Michael X. Zhu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston
| | - Edgar T. Walters
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston
| |
Collapse
|
10
|
Qiao Z, Liu S, Zhai W, Jiang L, Ma Y, Zhang Z, Wang B, Shao J, Qian H, Zhao F, Yan L. Novel dual-target FAAH and TRPV1 ligands as potential pharmacotherapeutics for pain management. Eur J Med Chem 2024; 267:116208. [PMID: 38325006 DOI: 10.1016/j.ejmech.2024.116208] [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: 12/12/2023] [Revised: 01/17/2024] [Accepted: 01/31/2024] [Indexed: 02/09/2024]
Abstract
Dual-acting drugs that simultaneously inhibit fatty acid amide hydrolase (FAAH) and antagonize the transient receptor potential vanilloid 1 (TRPV1) is a promising stronger therapeutic approach for pain management without side effects associated with single-target agents. Here, several series of dual FAAH/TRPV1 blockers were designed and synthesized through rational molecular hybridization between the pharmacophore of classical TRPV1 antagonists and FAAH inhibitors. The studies resulted in compound 2r, which exhibited strong dual FAAH/TRPV1 inhibition/antagonism in vitro, exerted powerful analgesic effects in formalin-induced pain test (phase II, in mice), desirable anti-inflammatory activity in carrageenan-induced paw edema in rats, no TRPV1-related hyperthermia side effect, and favorable pharmacokinetic properties. Meanwhile, the contributions of TRPV1 and FAAH to its antinociceptive effects were verified by target engagement and molecular docking studies. Overall, compound 2r can serve as a new scaffold for developing FAAH/TRPV1 dual-activie ligands to counteract pain.
Collapse
Affiliation(s)
- Zhenrui Qiao
- State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan, 475004, China
| | - Shuyu Liu
- State Key Laboratory of Natural Medicines, Center of Drug Discovery, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, Jiangsu, 210009, China
| | - Weibin Zhai
- State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan, 475004, China
| | - Lei Jiang
- State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan, 475004, China
| | - Yunmeng Ma
- State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan, 475004, China
| | - Zhikang Zhang
- State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan, 475004, China
| | - Bingxin Wang
- State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan, 475004, China
| | - Jingwen Shao
- State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan, 475004, China
| | - Hai Qian
- State Key Laboratory of Natural Medicines, Center of Drug Discovery, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, Jiangsu, 210009, China
| | - Fenqin Zhao
- State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan, 475004, China.
| | - Lin Yan
- State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan, 475004, China.
| |
Collapse
|
11
|
Bavencoffe AG, Lopez ER, Johnson KN, Tian J, Gorgun FM, Shen BQ, Zhu MX, Dessauer CW, Walters ET. Widespread latent hyperactivity of nociceptors outlasts enhanced avoidance behavior following incision injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.30.578108. [PMID: 38352319 PMCID: PMC10862851 DOI: 10.1101/2024.01.30.578108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/24/2024]
Abstract
Nociceptors with somata in dorsal root ganglia (DRGs) exhibit an unusual readiness to switch from an electrically silent state to a hyperactive state of tonic, nonaccommodating, low-frequency, irregular discharge of action potentials (APs). Ongoing activity (OA) during this state is present in vivo in rats months after spinal cord injury (SCI), and has been causally linked to SCI pain. OA induced by various neuropathic conditions in rats, mice, and humans is retained in nociceptor somata after dissociation and culturing, providing a powerful tool for investigating its mechanisms and functions. An important question is whether similar nociceptor OA is induced by painful conditions other than neuropathy. The present study shows that probable nociceptors dissociated from DRGs of rats subjected to postsurgical pain (induced by plantar incision) exhibit OA. The OA was most apparent when the soma was artificially depolarized to a level within the normal range of membrane potentials where large, transient depolarizing spontaneous fluctuations (DSFs) can approach AP threshold. This latent hyperactivity persisted for at least 3 weeks, whereas behavioral indicators of affective pain - hindpaw guarding and increased avoidance of a noxious substrate in an operant conflict test - persisted for 1 week or less. An unexpected discovery was latent OA in neurons from thoracic DRGs that innervate dermatomes distant from the injured tissue. The most consistent electrophysiological alteration associated with OA was enhancement of DSFs. Potential in vivo functions of widespread, low-frequency nociceptor OA consistent with these and other findings are to amplify hyperalgesic priming and to drive anxiety-related hypervigilance.
Collapse
Affiliation(s)
- Alexis G. Bavencoffe
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston
| | - Elia R. Lopez
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston
| | - Kayla N. Johnson
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston
| | - Jinbin Tian
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston
| | - Falih M. Gorgun
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston
| | - Breanna Q. Shen
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston
| | - Michael X. Zhu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston
| | - Carmen W. Dessauer
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston
| | - Edgar T. Walters
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston
| |
Collapse
|
12
|
Zeng J, Lu Y, Chu H, Lu L, Chen Y, Ji K, Lin Y, Li J, Wang S. Research trends and frontier hotspots of TRPV1 based on bibliometric and visualization analyses. Heliyon 2024; 10:e24153. [PMID: 38293347 PMCID: PMC10827456 DOI: 10.1016/j.heliyon.2024.e24153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 11/30/2023] [Accepted: 01/04/2024] [Indexed: 02/01/2024] Open
Abstract
Background Transient receptor potential vanilloid type1 (TRPV1) is a non-selective cation channel with multiple activation mechanisms, which has received increasing attention since it was first cloned in 1997. Methods We used bibliometric and visualization analyses to evaluate the theme trends and knowledge structure of TRPV1 research-papers on TRPV1 from 2002 to 2022 obtained from the Web of Science Core Collection. VOSviewer and CiteSpace were used to analyze authors, institutions, countries, co-cited references, and keywords. Results A total of 7413 papers were included. The main research area of TRPV1 was neuroscience; the most published country was the United States, and the University of California, San Francisco, had the highest centrality. Two major collaborative sub-networks were formed between the authors. The distribution of keywords shows that TRPV1 was initially studied extensively, and the recent studies focused on TRPV1 structure and diseases. "Oxidative stress," "TRPV1 structure," "cancer," and "model" have been the research hotspots in recent years. Conclusions This research provides valuable information for the study of TRPV1. Disease research was focused on pain, cancer, and neurodegenerative diseases. Both agonists and antagonists of TRPV1 are gradually being used in clinical practice, and acupuncture was effective in treating TRPV1-mediated inflammatory pain. TRPV1 is involved in classical endogenous cannabis system signaling, and new signaling pathways continue to be revealed.
Collapse
Affiliation(s)
- Jingchun Zeng
- Rehabilitation Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yiqian Lu
- The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Hui Chu
- The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Liming Lu
- Clinical Research and Data Center, South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yuexuan Chen
- The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Kaisong Ji
- The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yeze Lin
- Rehabilitation Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jingjing Li
- Bao'an Traditional Chinese Medicine Hospital//Seventh Clinical Medical College of Guangzhou University of Traditional Chinese Medicine, Shenzhen, Guangdong, China
| | - Shuxin Wang
- Rehabilitation Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
13
|
Saunders MN, Griffin KV, Kalashnikova I, Kolpek D, Smith DR, Saito E, Cummings BJ, Anderson AJ, Shea LD, Park J. Biodegradable nanoparticles targeting circulating immune cells reduce central and peripheral sensitization to alleviate neuropathic pain following spinal cord injury. Pain 2024; 165:92-101. [PMID: 37463227 PMCID: PMC10787809 DOI: 10.1097/j.pain.0000000000002989] [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: 12/27/2022] [Accepted: 05/26/2023] [Indexed: 07/20/2023]
Abstract
ABSTRACT Neuropathic pain is a critical source of comorbidity following spinal cord injury (SCI) that can be exacerbated by immune-mediated pathologies in the central and peripheral nervous systems. In this article, we investigate whether drug-free, biodegradable, poly(lactide- co -glycolide) (PLG) nanoparticle treatment mitigates the development of post-SCI neuropathic pain in female mice. Our results show that acute treatment with PLG nanoparticles following thoracic SCI significantly reduces tactile and cold hypersensitivity scores in a durable fashion. Nanoparticles primarily reduce peripheral immune-mediated mechanisms of neuropathic pain, including neuropathic pain-associated gene transcript frequency, transient receptor potential ankyrin 1 nociceptor expression, and MCP-1 (CCL2) chemokine production in the subacute period after injury. Altered central neuropathic pain mechanisms during this period are limited to reduced innate immune cell cytokine expression. However, in the chronic phase of SCI, nanoparticle treatment induces changes in both central and peripheral neuropathic pain signaling, driving reductions in cytokine production and other immune-relevant markers. This research suggests that drug-free PLG nanoparticles reprogram peripheral proalgesic pathways subacutely after SCI to reduce neuropathic pain outcomes and improve chronic central pain signaling.
Collapse
Affiliation(s)
- Michael N Saunders
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI USA
| | - Kate V Griffin
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI USA
| | - Irina Kalashnikova
- Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY USA
| | - Daniel Kolpek
- Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY USA
| | - Dominique R Smith
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI USA
| | - Eiji Saito
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI USA
| | - Brian J Cummings
- Department of Anatomy and Neurobiology, University of California, Irvine, CA USA
- Department of Physical Medicine and Rehabilitation, University of California, Irvine, CA USA
| | - Aileen J Anderson
- Department of Anatomy and Neurobiology, University of California, Irvine, CA USA
- Department of Physical Medicine and Rehabilitation, University of California, Irvine, CA USA
| | - Lonnie D Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI USA
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI USA
| | - Jonghyuck Park
- Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY USA
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY USA
| |
Collapse
|
14
|
Wang W, Sun T. Impact of TRPV1 on Pathogenesis and Therapy of Neurodegenerative Diseases. Molecules 2023; 29:181. [PMID: 38202764 PMCID: PMC10779880 DOI: 10.3390/molecules29010181] [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: 10/25/2023] [Revised: 12/11/2023] [Accepted: 12/11/2023] [Indexed: 01/12/2024] Open
Abstract
Transient receptor potential vanilloid 1 (TRPV1) is a transmembrane and non-selective cation channel protein, which can be activated by various physical and chemical stimuli. Recent studies have shown the strong pathogenetic associations of TRPV1 with neurodegenerative diseases (NDs), in particular Alzheimer's disease (AD), Parkinson's disease (PD) and multiple sclerosis (MS) via regulating neuroinflammation. Therapeutic effects of TRPV1 agonists and antagonists on the treatment of AD and PD in animal models also are emerging. We here summarize the current understanding of TRPV1's effects and its agonists and antagonists as a therapeutic means in neurodegenerative diseases, and highlight future treatment strategies using natural TRPV1 agonists. Developing new targets and applying natural products are becoming a promising direction in the treatment of chronic disorders, especially neurodegenerative diseases.
Collapse
Affiliation(s)
| | - Tao Sun
- Center for Precision Medicine, School of Medicine and School of Biomedical Sciences, Huaqiao University, Xiamen 361021, China;
| |
Collapse
|
15
|
Liu H, Lauzadis J, Gunaratna K, Sipple E, Kaczocha M, Puopolo M. Inhibition of T-Type Calcium Channels With TTA-P2 Reduces Chronic Neuropathic Pain Following Spinal Cord Injury in Rats. THE JOURNAL OF PAIN 2023; 24:1681-1695. [PMID: 37169156 DOI: 10.1016/j.jpain.2023.05.002] [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: 12/20/2022] [Revised: 04/03/2023] [Accepted: 05/02/2023] [Indexed: 05/13/2023]
Abstract
Spinal cord injury (SCI)-induced neuropathic pain (SCI-NP) develops in up to 60 to 70% of people affected by traumatic SCI, leading to a major decline in quality of life and increased risk for depression, anxiety, and addiction. Gabapentin and pregabalin, together with antidepressant drugs, are commonly prescribed to treat SCI-NP, but their efficacy is unsatisfactory. The limited efficacy of current pharmacological treatments for SCI-NP likely reflects our limited knowledge of the underlying mechanism(s) responsible for driving the maintenance of SCI-NP. The leading hypothesis in the field supports a major role for spontaneously active injured nociceptors in driving the maintenance of SCI-NP. Recent data from our laboratory provided additional support for this hypothesis and identified the T-type calcium channels as key players in driving the spontaneous activity of SCI-nociceptors, thus providing a rational pharmacological target to treat SCI-NP. To test whether T-type calcium channels contribute to the maintenance of SCI-NP, male and female SCI and sham rats were treated with TTA-P2 (a blocker of T-type calcium channels) to determine its effects on mechanical hypersensitivity (as measured with the von Frey filaments) and spontaneous ongoing pain (as measured with the conditioned place preference paradigm), and compared them to the effects of gabapentin, a blocker of high voltage-activated calcium channels. We found that both TTA-P2 and gabapentin reduced mechanical hypersensitivity in male and females SCI rats, but surprisingly only TTA-P2 reduced spontaneous ongoing pain in male SCI rats. PERSPECTIVES: SCI-induced neuropathic pain, and in particular the spontaneous ongoing pain component, is notoriously very difficult to treat. Our data provide evidence that inhibition of T-type calcium channels reduces spontaneous ongoing pain in SCI rats, supporting a clinically relevant role for T-type channels in the maintenance of SCI-induced neuropathic pain.
Collapse
Affiliation(s)
- Huilin Liu
- Department of Anesthesiology, Stony Brook Pain and Analgesia Research Center (SPARC), Health Sciences Center L4-072, Stony Brook Renaissance School of Medicine, Stony Brook, New York
| | - Justas Lauzadis
- Department of Anesthesiology, Stony Brook Pain and Analgesia Research Center (SPARC), Health Sciences Center L4-072, Stony Brook Renaissance School of Medicine, Stony Brook, New York
| | - Kavindu Gunaratna
- Department of Anesthesiology, Stony Brook Pain and Analgesia Research Center (SPARC), Health Sciences Center L4-072, Stony Brook Renaissance School of Medicine, Stony Brook, New York
| | - Erin Sipple
- Department of Anesthesiology, Stony Brook Pain and Analgesia Research Center (SPARC), Health Sciences Center L4-072, Stony Brook Renaissance School of Medicine, Stony Brook, New York
| | - Martin Kaczocha
- Department of Anesthesiology, Stony Brook Pain and Analgesia Research Center (SPARC), Health Sciences Center L4-072, Stony Brook Renaissance School of Medicine, Stony Brook, New York
| | - Michelino Puopolo
- Department of Anesthesiology, Stony Brook Pain and Analgesia Research Center (SPARC), Health Sciences Center L4-072, Stony Brook Renaissance School of Medicine, Stony Brook, New York.
| |
Collapse
|
16
|
Mustafa S, Bajic JE, Barry B, Evans S, Siemens KR, Hutchinson MR, Grace PM. One immune system plays many parts: The dynamic role of the immune system in chronic pain and opioid pharmacology. Neuropharmacology 2023; 228:109459. [PMID: 36775098 PMCID: PMC10015343 DOI: 10.1016/j.neuropharm.2023.109459] [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: 12/06/2022] [Revised: 02/06/2023] [Accepted: 02/08/2023] [Indexed: 02/12/2023]
Abstract
The transition from acute to chronic pain is an ongoing major problem for individuals, society and healthcare systems around the world. It is clear chronic pain is a complex multidimensional biological challenge plagued with difficulties in pain management, specifically opioid use. In recent years the role of the immune system in chronic pain and opioid pharmacology has come to the forefront. As a highly dynamic and versatile network of cells, tissues and organs, the immune system is perfectly positioned at the microscale level to alter nociception and drive structural adaptations that underpin chronic pain and opioid use. In this review, we highlight the need to understand the dynamic and adaptable characteristics of the immune system and their role in the transition, maintenance and resolution of chronic pain. The complex multidimensional interplay of the immune system with multiple physiological systems may provide new transformative insight for novel targets for clinical management and treatment of chronic pain. This article is part of the Special Issue on "Opioid-induced changes in addiction and pain circuits".
Collapse
Affiliation(s)
- Sanam Mustafa
- School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia; Australian Research Council Centre of Excellence for Nanoscale BioPhotonics, The University of Adelaide, Adelaide, SA, Australia; Davies Livestock Research Centre, The University of Adelaide, Roseworthy, SA, Australia.
| | - Juliana E Bajic
- School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia; Davies Livestock Research Centre, The University of Adelaide, Roseworthy, SA, Australia
| | - Benjamin Barry
- School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
| | - Samuel Evans
- School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia; Davies Livestock Research Centre, The University of Adelaide, Roseworthy, SA, Australia
| | - Kariel R Siemens
- School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
| | - Mark R Hutchinson
- School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia; Australian Research Council Centre of Excellence for Nanoscale BioPhotonics, The University of Adelaide, Adelaide, SA, Australia; Davies Livestock Research Centre, The University of Adelaide, Roseworthy, SA, Australia
| | - Peter M Grace
- Laboratories of Neuroimmunology, Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX, USA; MD Anderson Pain Research Consortium, Houston, TX, USA
| |
Collapse
|
17
|
Jang W, Oh M, Cho EH, Baek M, Kim C. Drosophila pain sensitization and modulation unveiled by a novel pain model and analgesic drugs. PLoS One 2023; 18:e0281874. [PMID: 36795675 PMCID: PMC9934396 DOI: 10.1371/journal.pone.0281874] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 02/01/2023] [Indexed: 02/17/2023] Open
Abstract
In mammals, pain is regulated by the combination of an ascending stimulating and descending inhibitory pain pathway. It remains an intriguing question whether such pain pathways are of ancient origin and conserved in invertebrates. Here we report a new Drosophila pain model and use it to elucidate the pain pathways present in flies. The model employs transgenic flies expressing the human capsaicin receptor TRPV1 in sensory nociceptor neurons, which innervate the whole fly body, including the mouth. Upon capsaicin sipping, the flies abruptly displayed pain-related behaviors such as running away, scurrying around, rubbing vigorously, and pulling at their mouth parts, suggesting that capsaicin stimulated nociceptors in the mouth via activating TRPV1. When reared on capsaicin-containing food, the animals died of starvation, demonstrating the degree of pain experienced. This death rate was reduced by treatment both with NSAIDs and gabapentin, analgesics that inhibit the sensitized ascending pain pathway, and with antidepressants, GABAergic agonists, and morphine, analgesics that strengthen the descending inhibitory pathway. Our results suggest Drosophila to possess intricate pain sensitization and modulation mechanisms similar to mammals, and we propose that this simple, non-invasive feeding assay has utility for high-throughput evaluation and screening of analgesic compounds.
Collapse
Affiliation(s)
- Wijeong Jang
- School of Biological Sciences and Technology, Chonnam National University, Gwangju, Korea
| | - Myungsok Oh
- School of Biological Sciences and Technology, Chonnam National University, Gwangju, Korea
| | - Eun-Hee Cho
- School of Biological Sciences and Technology, Chonnam National University, Gwangju, Korea
| | - Minwoo Baek
- School of Biological Sciences and Technology, Chonnam National University, Gwangju, Korea
| | - Changsoo Kim
- School of Biological Sciences and Technology, Chonnam National University, Gwangju, Korea
- * E-mail:
| |
Collapse
|
18
|
Talifu Z, Pan Y, Gong H, Xu X, Zhang C, Yang D, Gao F, Yu Y, Du L, Li J. The role of KCC2 and NKCC1 in spinal cord injury: From physiology to pathology. Front Physiol 2022; 13:1045520. [PMID: 36589461 PMCID: PMC9799334 DOI: 10.3389/fphys.2022.1045520] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
The balance of ion concentrations inside and outside the cell is an essential homeostatic mechanism in neurons and serves as the basis for a variety of physiological activities. In the central nervous system, NKCC1 and KCC2, members of the SLC12 cation-chloride co-transporter (CCC) family, participate in physiological and pathophysiological processes by regulating intracellular and extracellular chloride ion concentrations, which can further regulate the GABAergic system. Over recent years, studies have shown that NKCC1 and KCC2 are essential for the maintenance of Cl- homeostasis in neural cells. NKCC1 transports Cl- into cells while KCC2 transports Cl- out of cells, thereby regulating chloride balance and neuronal excitability. An imbalance of NKCC1 and KCC2 after spinal cord injury will disrupt CI- homeostasis, resulting in the transformation of GABA neurons from an inhibitory state into an excitatory state, which subsequently alters the spinal cord neural network and leads to conditions such as spasticity and neuropathic pain, among others. Meanwhile, studies have shown that KCC2 is also an essential target for motor function reconstruction after spinal cord injury. This review mainly introduces the physiological structure and function of NKCC1 and KCC2 and discusses their pathophysiological roles after spinal cord injury.
Collapse
Affiliation(s)
- Zuliyaer Talifu
- School of Rehabilitation, Capital Medical University, Beijing, China,Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China,Chinese Institute of Rehabilitation Science, Beijing, China,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China,School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Yunzhu Pan
- School of Rehabilitation, Capital Medical University, Beijing, China,Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China,Chinese Institute of Rehabilitation Science, Beijing, China,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China,School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Han Gong
- School of Rehabilitation, Capital Medical University, Beijing, China,Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China,Chinese Institute of Rehabilitation Science, Beijing, China,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Xin Xu
- School of Rehabilitation, Capital Medical University, Beijing, China,Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China,Chinese Institute of Rehabilitation Science, Beijing, China,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Chunjia Zhang
- School of Rehabilitation, Capital Medical University, Beijing, China,Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China,Chinese Institute of Rehabilitation Science, Beijing, China,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Degang Yang
- School of Rehabilitation, Capital Medical University, Beijing, China,Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Feng Gao
- School of Rehabilitation, Capital Medical University, Beijing, China,Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Yan Yu
- School of Rehabilitation, Capital Medical University, Beijing, China,Chinese Institute of Rehabilitation Science, Beijing, China,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Liangjie Du
- School of Rehabilitation, Capital Medical University, Beijing, China,Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China,*Correspondence: Liangjie Du, ; Jianjun Li,
| | - Jianjun Li
- School of Rehabilitation, Capital Medical University, Beijing, China,Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China,Chinese Institute of Rehabilitation Science, Beijing, China,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China,School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, China,*Correspondence: Liangjie Du, ; Jianjun Li,
| |
Collapse
|
19
|
Fernández-Carvajal A, Fernández-Ballester G, Ferrer-Montiel A. TRPV1 in chronic pruritus and pain: Soft modulation as a therapeutic strategy. Front Mol Neurosci 2022; 15:930964. [PMID: 36117910 PMCID: PMC9478410 DOI: 10.3389/fnmol.2022.930964] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 08/09/2022] [Indexed: 11/17/2022] Open
Abstract
Chronic pain and pruritus are highly disabling pathologies that still lack appropriate therapeutic intervention. At cellular level the transduction and transmission of pain and pruritogenic signals are closely intertwined, negatively modulating each other. The molecular and cellular pathways involved are multifactorial and complex, including peripheral and central components. Peripherally, pain and itch are produced by subpopulations of specialized nociceptors that recognize and transduce algesic and pruritogenic signals. Although still under intense investigation, cumulative evidence is pointing to the thermosensory channel TRPV1 as a hub for a large number of pro-algesic and itchy agents. TRPV1 appears metabolically coupled to most neural receptors that recognize algesic and pruritic molecules. Thus, targeting TRPV1 function appears as a valuable and reasonable therapeutic strategy. In support of this tenet, capsaicin, a desensitizing TRPV1 agonist, has been shown to exhibit clinically relevant analgesic, anti-inflammatory, and anti-pruritic activities. However, potent TRPV1 antagonists have been questioned due to an hyperthermic secondary effect that prevented their clinical development. Thus, softer strategies directed to modulate peripheral TRPV1 function appear warranted to alleviate chronic pain and itch. In this regard, soft, deactivatable TRPV1 antagonists for topical or local application appear as an innovative approach for improving the distressing painful and itchy symptoms of patients suffering chronic pain or pruritus. Here, we review the data on these compounds and propose that this strategy could be used to target other peripheral therapeutic targets.
Collapse
|
20
|
The Effect of Ginger and Its Sub-Components on Pain. PLANTS 2022; 11:plants11172296. [PMID: 36079679 PMCID: PMC9460519 DOI: 10.3390/plants11172296] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 08/27/2022] [Accepted: 08/31/2022] [Indexed: 11/16/2022]
Abstract
Zingiber officinale Roscoe (ginger) has long been used as an herbal medicine to treat various diseases, and its main sub-components, [6]-gingerol and [6]-shogaol, were also reported to have anti-inflammatory, anti-oxidant, and anti-tumor effects. However, their effects on various types of pain and their underlying mechanisms of action have not been clearly analyzed and understood yet. Thus, in this review, by analyzing 16 studies that used Z. officinale, [6]-gingerol, and [6]-shogaol on mechanical, spontaneous and thermal pain, their effects and mechanisms of action have been analyzed. Pain was induced by either nerve injury or chemical injections in rodents. Nine studies analyzed the analgesic effect of Z. officinale, and four and three studies focused on [6]-gingerol and [6]-shogaol, respectively. Seven papers have demonstrated the underlying mechanism of action of their analgesic effects. Studies have focused on the spinal cord and one on the dorsal root ganglion (DRG) neurons. Involvement and change in the function of serotonergic receptors (5-HT1A, B, D, and 5A), transient receptor potential vanilloid 1 (TRPV1), N-methyl-D-aspartate (NMDA) receptors, phosphorylated extracellular signal-regulated kinase 1/2 (pERK1/2), histone deacetylase 1 (HDAC1), voltage-gated sodium channel 1.8 (Nav1.8), substance P (SP), and sciatic nerve’s morphology have been observed.
Collapse
|
21
|
North RY, Odem MA, Li Y, Tatsui CE, Cassidy RM, Dougherty PM, Walters ET. Electrophysiological Alterations Driving Pain-Associated Spontaneous Activity in Human Sensory Neuron Somata Parallel Alterations Described in Spontaneously Active Rodent Nociceptors. THE JOURNAL OF PAIN 2022; 23:1343-1357. [PMID: 35292377 PMCID: PMC9357108 DOI: 10.1016/j.jpain.2022.02.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 02/21/2022] [Accepted: 02/24/2022] [Indexed: 06/10/2023]
Abstract
Neuropathic pain in rodents can be driven by ectopic spontaneous activity (SA) generated by sensory neurons in dorsal root ganglia (DRG). The recent demonstration that SA in dissociated human DRG neurons is associated with reported neuropathic pain in patients enables a detailed comparison of pain-linked electrophysiological alterations driving SA in human DRG neurons to alterations that distinguish SA in nociceptors from SA in low-threshold mechanoreceptors (LTMRs) in rodent neuropathy models. Analysis of recordings from dissociated somata of patient-derived DRG neurons showed that SA and corresponding pain in both sexes were significantly associated with the three functional electrophysiological alterations sufficient to generate SA in the absence of extrinsic depolarizing inputs. These include enhancement of depolarizing spontaneous fluctuations of membrane potential (DSFs), which were analyzed quantitatively for the first time in human DRG neurons. The functional alterations were indistinguishable from SA-driving alterations reported for nociceptors in rodent chronic pain models. Irregular, low-frequency DSFs in human DRG neurons closely resemble DSFs described in rodent nociceptors while differing substantially from the high-frequency sinusoidal oscillations described in rodent LTMRs. These findings suggest that conserved physiological mechanisms of SA in human nociceptor somata can drive neuropathic pain despite documented cellular differences between human and rodent DRG neurons. PERSPECTIVE: Electrophysiological alterations in human sensory neurons associated with patient-reported neuropathic pain include all three of the functional alterations that logically can promote spontaneous activity. The similarity of distinctively altered spontaneous depolarizations in human DRG neurons and rodent nociceptors suggests that spontaneously active human nociceptors can persistently promote neuropathic pain in patients.
Collapse
Affiliation(s)
- Robert Y North
- Department of Neurosurgery, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Max A Odem
- Department of Microbiology and Molecular Genetics, McGovern Medical School at UTHealth, Houston, Texas
| | - Yan Li
- Department of Anesthesia and Pain Medicine, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Claudio Esteves Tatsui
- Department of Neurosurgery, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Ryan M Cassidy
- M.D. Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas
| | - Patrick M Dougherty
- Department of Anesthesia and Pain Medicine, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Edgar T Walters
- Department of Integrative Biology and Pharmacology, McGovern Medical School at UTHealth, Houston, Texas..
| |
Collapse
|
22
|
Central Neuropathic Pain Syndromes: Current and Emerging Pharmacological Strategies. CNS Drugs 2022; 36:483-516. [PMID: 35513603 DOI: 10.1007/s40263-022-00914-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/10/2022] [Indexed: 12/31/2022]
Abstract
Central neuropathic pain is caused by a disease or lesion of the brain or spinal cord. It is difficult to predict which patients will develop central pain syndromes after a central nervous system injury, but depending on the etiology, lifetime prevalence may be greater than 50%. The resulting pain is often highly distressing and difficult to treat, with no specific treatment guidelines currently available. This narrative review discusses mechanisms contributing to central neuropathic pain, and focuses on pharmacological approaches for managing common central neuropathic pain conditions such as central post-stroke pain, spinal cord injury-related pain, and multiple sclerosis-related neuropathic pain. Tricyclic antidepressants, serotonin-norepinephrine reuptake inhibitors, and gabapentinoids have some evidence for efficacy in central neuropathic pain. Medications from other pharmacologic classes may also provide pain relief, but current evidence is limited. Certain non-pharmacologic approaches, neuromodulation in particular, may be helpful in refractory cases. Emerging data suggest that modulating the primary afferent input may open new horizons for the treatment of central neuropathic pain. For most patients, effective treatment will likely require a multimodal therapy approach.
Collapse
|
23
|
Akhilesh, Uniyal A, Gadepalli A, Tiwari V, Allani M, Chouhan D, Ummadisetty O, Verma N, Tiwari V. Unlocking the potential of TRPV1 based siRNA therapeutics for the treatment of chemotherapy-induced neuropathic pain. Life Sci 2022; 288:120187. [PMID: 34856209 DOI: 10.1016/j.lfs.2021.120187] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 11/24/2021] [Accepted: 11/24/2021] [Indexed: 01/23/2023]
Abstract
Chemotherapy-induced neuropathic pain (CINP) is among the most common clinical complications associated with the use of anti-cancer drugs. CINP occurs in nearly 68.1% of the cancer patients receiving chemotherapeutic drugs. Most of the clinically available analgesics are ineffective in the case of CINP patients as the pathological mechanisms involved with different chemotherapeutic drugs are distinct from each other. CINP triggers the somatosensory nervous system, increases the neuronal firing and activation of nociceptive mediators including transient receptor protein vanilloid 1 (TRPV1). TRPV1 is widely present in the peripheral nociceptive nerve cells and it has been reported that the higher expression of TRPV1 in DRGs serves a critical role in the potentiation of CINP. The therapeutic glory of TRPV1 is well recognized in clinics which gives a promising insight into the treatment of pain. But the adverse effects associated with some of the antagonists directed the scientists towards RNA interference (RNAi), a tool to silence gene expression. Thus, ongoing research is focused on developing small interfering RNA (siRNA)-based therapeutics targeting TRPV1. In this review, we have discussed the involvement of TRPV1 in the nociceptive signaling associated with CINP and targeting this nociceptor, using siRNA will potentially arm us with effective therapeutic interventions for the clinical management of CINP.
Collapse
Affiliation(s)
- Akhilesh
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Ankit Uniyal
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Anagha Gadepalli
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Vineeta Tiwari
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Meghana Allani
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Deepak Chouhan
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Obulapathi Ummadisetty
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Nimisha Verma
- Department of Anaesthesiology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Vinod Tiwari
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi, Uttar Pradesh, India.
| |
Collapse
|
24
|
Zhao F, Wang S, Li Y, Wang J, Wang Y, Zhang C, Li Y, Huang L, Yu Y, Zheng J, Yu B, Pessah IN, Cao Z. Surfactant cocamide monoethanolamide causes eye irritation by activating nociceptor TRPV1 channels. Br J Pharmacol 2021; 178:3448-3462. [PMID: 33837959 PMCID: PMC11164132 DOI: 10.1111/bph.15491] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 12/19/2020] [Accepted: 04/02/2021] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND PURPOSE Cocamide monoethanolamide (CMEA) is commonly used as a surfactant-foam booster in cosmetic formulations. Upon contact with the eye or other sensitive skin areas, CMEA elicits stinging and lasting irritation. We hypothesized a specific molecular interaction with TRPV1 channels by which CMEA caused eye irritation. EXPERIMENTAL APPROACH Eye irritancy was evaluated using eye-wiping tests in rabbits and mice. Intracellular Ca2+ concentrations and action potentials were measured using Ca2+ imaging and current clamp respectively. Voltage clamp, site-direct mutagenesis and molecular modelling were used to identify binding pockets for CMEA on TRPV1 channels. KEY RESULTS CMEA-induced eye irritation is ameliorated by selective ablation of TRPV1 channels.Rodents exhibit much stronger responses to CMEA than rabbits. In trigeminal ganglion neurons, CMEA induced Ca2+ influx and neuronal excitability, effects mitigated by a TRPV1 channel inhibition and absent in TRPV1 knockout neurons. In HEK-293 cells expressing TRPV1 channels, CMEA increased whole-cell currents by increasing channel open probability (EC50 = 10.2 μM), without affecting TRPV2, TRPV3, TRPV4, and TRPA1 channel activities. Lauric acid monoethanolamide (LAMEA), the most abundant constituent of CMEA, was the most efficacious and potent TRPV1 channel activator, binding to the capsaicin-binding pocket of the channel. The T550I mutants of rabbit and human TRPV1 channels exhibit much lower sensitivity to LAMEA. CONCLUSIONS AND IMPLICATION CMEA directly activates TRPV1 channels to produce eye irritation. Rabbits, the standard animal used for eye irritancy tests are poor models for evaluating human eye irritants structurally related to CMEA. Our study identifies potential alternatives to CMEA as non-irritating surfactants.
Collapse
Affiliation(s)
- Fang Zhao
- State Key Laboratory of Natural Medicines and Department of TCM pharmacology, School of Traditional Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China, 211198
| | - Shuangyan Wang
- State Key Laboratory of Natural Medicines and Department of TCM pharmacology, School of Traditional Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China, 211198
| | - Yan Li
- State Key Laboratory of Natural Medicines and Department of TCM pharmacology, School of Traditional Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China, 211198
| | - Jin Wang
- Department of Basic Medicine, School of Basic Medicine and Clinic Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China, 211198
| | - Yujing Wang
- State Key Laboratory of Natural Medicines and Department of TCM pharmacology, School of Traditional Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China, 211198
| | - Chunlei Zhang
- State Key Laboratory of Natural Medicines and Department of TCM pharmacology, School of Traditional Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China, 211198
| | - Yong Li
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China, 200025
| | - Longjiang Huang
- College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, China, 266042
| | - Ye Yu
- Department of Basic Medicine, School of Basic Medicine and Clinic Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China, 211198
| | - Jie Zheng
- Department of Physiology and Membrane Biology, University of California, Davis, CA, USA, 95616
| | - Boyang Yu
- State Key Laboratory of Natural Medicines and Department of TCM pharmacology, School of Traditional Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China, 211198
| | - Isaac N Pessah
- Department of Molecular Bioscience, School of Veterinary Medicine, University of California, Davis, California, USA, 95616
| | - Zhengyu Cao
- State Key Laboratory of Natural Medicines and Department of TCM pharmacology, School of Traditional Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China, 211198
| |
Collapse
|
25
|
Fang J, Wang S, Zhou J, Shao X, Sun H, Liang Y, He X, Jiang Y, Liu B, Jin X, Fang J, Du J. Electroacupuncture Regulates Pain Transition Through Inhibiting PKCε and TRPV1 Expression in Dorsal Root Ganglion. Front Neurosci 2021; 15:685715. [PMID: 34354561 PMCID: PMC8329384 DOI: 10.3389/fnins.2021.685715] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 05/28/2021] [Indexed: 11/16/2022] Open
Abstract
Many cases of acute pain can be resolved with few side effects. However, some cases of acute pain may persist beyond the time required for tissue injury recovery and transit to chronic pain, which is hard to treat. The mechanisms underlying pain transition are not entirely understood, and treatment strategies are lacking. In this study, the hyperalgesic priming model was established on rats to study pain transition by injection of carrageenan (Car) and prostaglandin E2 (PGE2). The expression levels of protein kinase C epsilon (PKCε) and transient receptor potential vanilloid 1 (TRPV1) in the L4–L6 dorsal root ganglion (DRG) were investigated. Electroacupuncture (EA) is a form of acupuncture in which a small electric current is passed between a pair of acupuncture needles. EA was administrated, and its effect on hyperalgesia and PKCε and TRPV1 expression was investigated. The PKCε–TRPV1 signaling pathway in DRG was implicated in the pain transition. EA increased the pain threshold of model animals and regulated the high expression of PKCε and TRPV1. Moreover, EA also regulated hyperalgesia and high TRPV1 expression induced by selective PKCε activation. We also found that EA partly increased chronic pain threshold, even though it was only administered between the Car and PGE2 injections. These findings suggested that EA could prevent the transition from acute to chronic pain by inhibiting the PKCε and TRPV1 expression in the peripheral nervous system.
Collapse
Affiliation(s)
- Junfan Fang
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou, China
| | - Sisi Wang
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou, China
| | - Jie Zhou
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou, China
| | - Xiaomei Shao
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou, China
| | - Haiju Sun
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou, China
| | - Yi Liang
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou, China
| | - Xiaofen He
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou, China
| | - Yongliang Jiang
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou, China
| | - Boyi Liu
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou, China
| | - Xiaoming Jin
- Department of Anatomy, Cell Biology and Physiology, Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Jianqiao Fang
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou, China
| | - Junying Du
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou, China
| |
Collapse
|
26
|
Lee K, Choi YI, Im ST, Hwang SM, Lee HK, Im JZ, Kim YH, Jung SJ, Park CK. Riboflavin Inhibits Histamine-Dependent Itch by Modulating Transient Receptor Potential Vanilloid 1 (TRPV1). Front Mol Neurosci 2021; 14:643483. [PMID: 34220447 PMCID: PMC8249943 DOI: 10.3389/fnmol.2021.643483] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 05/18/2021] [Indexed: 11/13/2022] Open
Abstract
Riboflavin, also known as vitamin B2, isfound in foods and is used as a dietary supplement. Its deficiency (also called ariboflavinosis) results in some skin lesions and inflammations, such as stomatitis, cheilosis, oily scaly skin rashes, and itchy, watery eyes. Various therapeutic effects of riboflavin, such as anticancer, antioxidant, anti-inflammatory, and anti-nociceptive effects, are well known. Although some studies have identified the clinical effect of riboflavin on skin problems, including itch and inflammation, its underlying mechanism of action remains unknown. In this study, we investigated the molecular mechanism of the effects of riboflavin on histamine-dependent itch based on behavioral tests and electrophysiological experiments. Riboflavin significantly reduced histamine-induced scratching behaviors in mice and histamine-induced discharges in single-nerve fiber recordings, while it did not alter motor function in the rotarod test. In cultured dorsal root ganglion (DRG) neurons, riboflavin showed a dose-dependent inhibitory effect on the histamine- and capsaicin-induced inward current. Further tests wereconducted to determine whether two endogenous metabolites of riboflavin, flavin mononucleotide (FMN) and flavin adenine dinucleotide (FAD), have similar effects to those of riboflavin. Here, FMN, but not FAD, significantly inhibited capsaicin-induced currents and itching responses caused by histamine. In addition, in transient receptor potential vanilloid 1 (TRPV1)-transfected HEK293 cells, both riboflavin and FMN blocked capsaicin-induced currents, whereas FAD did not. These results revealed that riboflavin inhibits histamine-dependent itch by modulating TRPV1 activity. This study will be helpful in understanding how riboflavin exerts antipruritic effects and suggests that it might be a useful drug for the treatment of histamine-dependent itch.
Collapse
Affiliation(s)
- Kihwan Lee
- Tooth-Periodontium Complex Medical Research Center (MRC), Department of Physiology, School of Dentistry, Seoul National University, Seoul, South Korea
| | - Young In Choi
- Department of Physiology, College of Medicine, Hanyang University, Seoul, South Korea
| | - Sang-Taek Im
- Gachon Pain Center and Department of Physiology, College of Medicine, Gachon University, Incheon, South Korea
| | - Sung-Min Hwang
- Gachon Pain Center and Department of Physiology, College of Medicine, Gachon University, Incheon, South Korea
| | - Han-Kyu Lee
- Department of Physiology, College of Medicine, Hanyang University, Seoul, South Korea
| | - Jay-Zoon Im
- Department of Physiology, College of Medicine, Hanyang University, Seoul, South Korea
| | - Yong Ho Kim
- Gachon Pain Center and Department of Physiology, College of Medicine, Gachon University, Incheon, South Korea
| | - Sung Jun Jung
- Department of Physiology, College of Medicine, Hanyang University, Seoul, South Korea
| | - Chul-Kyu Park
- Gachon Pain Center and Department of Physiology, College of Medicine, Gachon University, Incheon, South Korea
| |
Collapse
|
27
|
Deng SY, Tang XC, Chang YC, Xu ZZ, Chen QY, Cao N, Kong LJY, Wang Y, Ma KT, Li L, Si JQ. Improving NKCC1 Function Increases the Excitability of DRG Neurons Exacerbating Pain Induced After TRPV1 Activation of Primary Sensory Neurons. Front Cell Neurosci 2021; 15:665596. [PMID: 34113239 PMCID: PMC8185156 DOI: 10.3389/fncel.2021.665596] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/13/2021] [Indexed: 11/13/2022] Open
Abstract
Background Our aim was to investigate the effects of the protein expression and the function of sodium, potassium, and chloride co-transporter (NKCC1) in the dorsal root ganglion (DRG) after activation of transient receptor potential vanilloid 1 receptor (TRPV1) in capsaicin-induced acute inflammatory pain and the possible mechanism of action. Methods Male Sprague–Dawley rats were randomly divided into control, capsaicin, and inhibitor groups. The expression and distribution of TRPV1 and NKCC1 in rat DRG were observed by immunofluorescence. Thermal radiation and acetone test were used to detect the pain threshold of heat and cold noxious stimulation in each group. The expressions of NKCC1 mRNA, NKCC1 protein, and p-NKCC1 in the DRG were detected by PCR and western blotting (WB). Patch clamp and chloride fluorescent probe were used to observe the changes of GABA activation current and intracellular chloride concentration. After intrathecal injection of protein kinase C (PKC) inhibitor (GF109203X) or MEK/extracellular signal-regulated kinase (ERK) inhibitor (U0126), the behavioral changes and the expression of NKCC1 and p-ERK protein in L4–6 DRG were observed. Result: TRPV1 and NKCC1 were co-expressed in the DRG. Compared with the control group, the immunofluorescence intensity of NKCC1 and p-NKCC1 in the capsaicin group was significantly higher, and the expression of NKCC1 in the nuclear membrane was significantly higher than that in the control group. The expression of NKCC1 mRNA and protein of NKCC1 and p-NKCC1 in the capsaicin group were higher than those in the control group. After capsaicin injection, GF109203X inhibited the protein expression of NKCC1 and p-ERK, while U0126 inhibited the protein expression of NKCC1. In the capsaicin group, paw withdrawal thermal latency (WTL) was decreased, while cold withdrawal latency (CWL) was prolonged. Bumetanide, GF109203X, or U0126 could reverse the effect. GABA activation current significantly increased in the DRG cells of the capsaicin group, which could be reversed by bumetanide. The concentration of chloride in the DRG cells of the capsaicin group increased, but decreased after bumetanide, GF109203X, and U0126 were administered. Conclusion Activation of TRPV1 by exogenous agonists can increase the expression and function of NKCC1 protein in DRG, which is mediated by activation of PKC/p-ERK signaling pathway. These results suggest that DRG NKCC1 may participate in the inflammatory pain induced by TRPV1.
Collapse
Affiliation(s)
- Shi-Yu Deng
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Department of Physiology, Shihezi University Medical College, Shihezi, China.,Department of Anesthesia, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Xue-Chun Tang
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Department of Physiology, Shihezi University Medical College, Shihezi, China.,NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China.,Department of Cardiology, First Affiliated Hospital of Shihezi University, Shihezi, China
| | - Yue-Chen Chang
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Department of Physiology, Shihezi University Medical College, Shihezi, China.,NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China.,Medical Teaching Experimental Center, Shihezi University Medical College, Shihezi, China
| | - Zhen-Zhen Xu
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Department of Physiology, Shihezi University Medical College, Shihezi, China.,NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China.,Department of Anesthesiology, Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qin-Yi Chen
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Department of Physiology, Shihezi University Medical College, Shihezi, China.,Department of Anesthesiology, Xiangyang Central Hospital, Xiangyang Central Hospital, China
| | - Nan Cao
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Department of Physiology, Shihezi University Medical College, Shihezi, China.,NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Liang-Jing-Yuan Kong
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Department of Physiology, Shihezi University Medical College, Shihezi, China.,NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Yang Wang
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Department of Physiology, Shihezi University Medical College, Shihezi, China.,NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Ke-Tao Ma
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Department of Physiology, Shihezi University Medical College, Shihezi, China.,NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Li Li
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Department of Physiology, Shihezi University Medical College, Shihezi, China.,NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China.,Department of Physiology, Medical College of Jiaxing University, Jiaxing, China
| | - Jun-Qiang Si
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Department of Physiology, Shihezi University Medical College, Shihezi, China.,NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| |
Collapse
|
28
|
Kong F, Sun K, Zhu J, Li F, Lin F, Sun X, Luo X, Ren C, Lu L, Zhao S, Sun J, Wang Y, Shi J. PD-L1 Improves Motor Function and Alleviates Neuropathic Pain in Male Mice After Spinal Cord Injury by Inhibiting MAPK Pathway. Front Immunol 2021; 12:670646. [PMID: 33936116 PMCID: PMC8081847 DOI: 10.3389/fimmu.2021.670646] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 03/29/2021] [Indexed: 12/17/2022] Open
Abstract
Background Traumatic spinal cord injury (SCI) causes severe motor dysfunction and persistent central neuropathic pain (Nep), which has not yet been effectively cured. Programmed cell death ligand-1 (PD-L1) is typically produced by cancer cells and contributes to the immune-suppressive in tumor microenvironment. However, the role of PD-L1 in regulating inflammatory response and Nep after SCI remains unclear. A growing amount of researches have begun to investigate the effect of PD-L1 on macrophages and microglia in recent years. Considering the pivotal role of macrophages/microglia in the inflammatory response after SCI, we proposed the hypothesis that PD-L1 improved the recovery of locomotor and sensory functions after SCI through regulating macrophages and microglia. Methods The mice SCI model was established to determine the changes in expression patterns of PD-L1. Meanwhile, we constructed PD-L1 knockout mice to observe differences in functional recovery and phenotypes of macrophages/microglia post-SCI. Results In present study, PD-L1 was significantly upregulated after SCI and highly expressed on macrophages/microglia at the injury epicenter. PD-L1 knockout (KO) mice showed worse locomotor recovery and more serious pathological pain compared with wild-type (WT) mice. Furthermore, deletion of PD-L1 significantly increased the polarization of M1-like macrophages/microglia. Mechanistic analysis revealed that PD-L1 may improve functional outcomes following SCI by inhibiting phosphorylation of p38 and ERK1/2. Conclusions Our observations implicate the involvement of PD-L1 in recovery of SCI and provide a new treatment strategy for the prevention and treatment of this traumatic condition.
Collapse
Affiliation(s)
- Fanqi Kong
- Department of Orthopedic Surgery, Spine Center, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Kaiqiang Sun
- Department of Orthopedic Surgery, Spine Center, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Jian Zhu
- Department of Orthopedic Surgery, Spine Center, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Fudong Li
- Department of Orthopedic Surgery, Spine Center, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Feng Lin
- Department of Orthopedic Surgery, Spine Center, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Xiaofei Sun
- Department of Orthopedic Surgery, Spine Center, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Xi Luo
- Department of Orthopedic Surgery, Spine Center, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Changzhen Ren
- Department of Cardiology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Lantao Lu
- Department of Orthopedic Surgery, Spine Center, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - ShuJie Zhao
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jingchuan Sun
- Department of Orthopedic Surgery, Spine Center, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Yuan Wang
- Department of Orthopedic Surgery, Spine Center, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Jiangang Shi
- Department of Orthopedic Surgery, Spine Center, Changzheng Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
29
|
Morrison D, Arcese AA, Parrish J, Gibbs K, Beaufort A, Herman P, Stein AB, Bloom O. Systemic gene expression profiles according to pain types in individuals with chronic spinal cord injury. Mol Pain 2021; 17:17448069211007289. [PMID: 33853401 PMCID: PMC8053765 DOI: 10.1177/17448069211007289] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Pain affects most individuals with traumatic spinal cord injury (SCI). Major pain types after SCI are neuropathic or nociceptive, often experienced concurrently. Pain after SCI may be refractory to treatments and negatively affects quality of life. Previously, we analyzed whole blood gene expression in individuals with chronic SCI compared to able-bodied (AB) individuals. Most participants with SCI reported pain (N = 19/28). Here, we examined gene expression of participants with SCI by pain status. Compared to AB, participants with SCI with pain had 468 differentially expressed (DE) genes; participants without pain had 564 DE genes (FDR < 0.05). Among DE genes distinct to participants with SCI with pain, Gene Ontology Biological Process (GOBP) analysis showed upregulated genes were enriched in categories related to T cell activation or inflammation; downregulated genes were enriched in categories related to protein proteolysis and catabolism. Although most participants with pain reported multiple pain types concurrently, we performed a preliminary comparison of gene expression by worst pain problem type. Compared to AB, participants with SCI who ranked neuropathic (N = 9) as worst had one distinct DE gene (TMEM156); participants who ranked nociceptive (N = 10) as worst had 61 distinct DE genes (FDR < 0.05). In the nociceptive group, the GOBP category with the lowest P-value identified among upregulated genes was “positive regulation of T cell activation”; among downregulated genes it was “receptor tyrosine kinase binding”. An exploratory comparison of pain groups by principal components analysis also showed that the nociceptive group was enriched in T-cell related genes. A correlation analysis identified genes significantly correlated with pain intensity in the neuropathic or nociceptive groups (N = 145, 65, respectively, Pearson’s correlation r > 0.8). While this pilot study highlights challenges of identifying gene expression profiles that correlate with specific types of pain in individuals with SCI, it suggests that T-cell signaling should be further investigated in this context.
Collapse
Affiliation(s)
- Debra Morrison
- Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Anthony A Arcese
- Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Janay Parrish
- Donald and Barbara Zucker School of Medicine at Hofstra-Northwell, Hempstead, NY, USA
| | - Katie Gibbs
- Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY, USA.,Department of Physical Medicine and Rehabilitation, Zucker School of Medicine at Hofstra-Northwell, Hempstead, NY, USA
| | - Andrew Beaufort
- Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Paige Herman
- Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Adam B Stein
- Department of Physical Medicine and Rehabilitation, Zucker School of Medicine at Hofstra-Northwell, Hempstead, NY, USA
| | - Ona Bloom
- Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY, USA.,Department of Physical Medicine and Rehabilitation, Zucker School of Medicine at Hofstra-Northwell, Hempstead, NY, USA.,Department of Molecular Medicine, Zucker School of Medicine at Hofstra-Northwell, Hempstead, NY, USA
| |
Collapse
|
30
|
Miao B, Yin Y, Mao G, Zhao B, Wu J, Shi H, Fei S. The implication of transient receptor potential canonical 6 in BDNF-induced mechanical allodynia in rat model of diabetic neuropathic pain. Life Sci 2021; 273:119308. [PMID: 33667520 DOI: 10.1016/j.lfs.2021.119308] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 02/18/2021] [Accepted: 02/21/2021] [Indexed: 02/01/2023]
Abstract
AIMS Brain-derived neurotrophic factor (BDNF) is vital in the pathogenesis of mechanical allodynia with a paucity of reports available regarding diabetic neuropathy pain (DNP). Herein we identified the involvement of BDNF in driving mechanical allodynia in DNP rats via the activation of transient receptor potential canonical 6 (TRPC6) channel. MATERIALS AND METHODS The DNP rat model was established via streptozotocin (STZ) injection, and allodynia was assessed by paw withdrawal mechanical threshold (PWMT) and paw withdrawal thermal latency (PWTL). The expression profiles of BDNF and TRPC6 in dorsal root ganglia (DRG) and spinal cord were illustrated by immunofluorescence and Western blotting. Intrathecal administration of K252a or TrkB-Fc was performed to inhibit BNDF/TrkB expression, and respective injection of GsMTX-4, BTP2 and TRPC6 antisense oligodeoxynucleotides (TRPC6-AS) was likewise conducted to inhibit TRPC6 expression in DNP rats. Calcium influx in DRG was monitored by calcium imaging. KEY FINDINGS The time-dependent increase of BDNF and TRPC6 expression in DRG and spinal cord was observed since the 7th post-STZ day, correlated with the development of mechanical allodynia in DNP rats. Intrathecal administration of K252a, TrkB-Fc, GsMTX-4 and BTP2 prevented mechanical allodynia in DNP rats. Pre-treatment of TRPC6-AS reversed the BDNF-induced pain-like responses in DNP rats rather than the naïve rats. In addition, the TRPC6-AS reversed BDNF-induced increase of calcium influx in DRG neurons in DNP rats. SIGNIFICANCE The intrathecal inhibition of TRPC6 alleviated the BDNF-induced mechanical allodynia in DNP rat model. This finding may validate the application of TRPC6 antagonists as interesting strategy for DNP management.
Collapse
Affiliation(s)
- Bei Miao
- Department of Gastroenterology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou 221002, Jiangsu Province, China; Institute of Digestive Diseases, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou 221002, Jiangsu Province, China
| | - Yue Yin
- Department of Anesthesiology, Xuzhou Central Hospital, 199 Jiefang South Road, Xuzhou 221009, Jiangsu Province, China
| | - Guangtong Mao
- Department of Pathology, Xinyi People's Hospital, 16 Renmin Road, Xinyi 221400, Jiangsu Province, China
| | - Benhuo Zhao
- Department of Gastroenterology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou 221002, Jiangsu Province, China
| | - Jiaojiao Wu
- Department of Gastroenterology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou 221002, Jiangsu Province, China
| | - Hengliang Shi
- Central Laboratory, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou 221002, Jiangsu Province, China.
| | - Sujuan Fei
- Department of Gastroenterology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou 221002, Jiangsu Province, China; Institute of Digestive Diseases, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou 221002, Jiangsu Province, China.
| |
Collapse
|
31
|
BmK NSPK, a Potent Potassium Channel Inhibitor from Scorpion Buthus martensii Karsch, Promotes Neurite Outgrowth via NGF/TrkA Signaling Pathway. Toxins (Basel) 2021; 13:toxins13010033. [PMID: 33466524 PMCID: PMC7824859 DOI: 10.3390/toxins13010033] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 12/30/2020] [Accepted: 12/31/2020] [Indexed: 12/31/2022] Open
Abstract
Scorpion toxins represent a variety of tools to explore molecular mechanisms and cellular signaling pathways of many biological functions. These toxins are also promising lead compounds for developing treatments for many neurological diseases. In the current study, we purified a new scorpion toxin designated as BmK NSPK (Buthus martensii Karsch neurite-stimulating peptide targeting Kv channels) from the BmK venom. The primary structure was determined using Edman degradation. BmK NSPK directly inhibited outward K+ current without affecting sodium channel activities, depolarized membrane, and increased spontaneous calcium oscillation in spinal cord neurons (SCNs) at low nanomolar concentrations. BmK NSPK produced a nonmonotonic increase on the neurite extension that peaked at ~10 nM. Mechanistic studies demonstrated that BmK NSPK increased the release of nerve growth factor (NGF). The tyrosine kinases A (TrkA) receptor inhibitor, GW 441756, eliminated the BmK NSPK-induced neurite outgrowth. BmK NSPK also increased phosphorylation levels of protein kinase B (Akt) that is the downstream regulator of TrkA receptors. These data demonstrate that BmK NSPK is a new voltage-gated potassium (Kv) channel inhibitor that augments neurite extension via NGF/TrkA signaling pathway. Kv channels may represent molecular targets to modulate SCN development and regeneration and to develop the treatments for spinal cord injury.
Collapse
|
32
|
Zheng H, Lim JY, Seong JY, Hwang SW. The Role of Corticotropin-Releasing Hormone at Peripheral Nociceptors: Implications for Pain Modulation. Biomedicines 2020; 8:biomedicines8120623. [PMID: 33348790 PMCID: PMC7766747 DOI: 10.3390/biomedicines8120623] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/15/2020] [Accepted: 12/15/2020] [Indexed: 12/12/2022] Open
Abstract
Peripheral nociceptors and their synaptic partners utilize neuropeptides for signal transmission. Such communication tunes the excitatory and inhibitory function of nociceptor-based circuits, eventually contributing to pain modulation. Corticotropin-releasing hormone (CRH) is the initiator hormone for the conventional hypothalamic-pituitary-adrenal axis, preparing our body for stress insults. Although knowledge of the expression and functional profiles of CRH and its receptors and the outcomes of their interactions has been actively accumulating for many brain regions, those for nociceptors are still under gradual investigation. Currently, based on the evidence of their expressions in nociceptors and their neighboring components, several hypotheses for possible pain modulations are emerging. Here we overview the historical attention to CRH and its receptors on the peripheral nociception and the recent increases in information regarding their roles in tuning pain signals. We also briefly contemplate the possibility that the stress-response paradigm can be locally intrapolated into intercellular communication that is driven by nociceptor neurons. Such endeavors may contribute to a more precise view of local peptidergic mechanisms of peripheral pain modulation.
Collapse
Affiliation(s)
- Haiyan Zheng
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Korea; (H.Z.); (J.Y.L.); (J.Y.S.)
| | - Ji Yeon Lim
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Korea; (H.Z.); (J.Y.L.); (J.Y.S.)
| | - Jae Young Seong
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Korea; (H.Z.); (J.Y.L.); (J.Y.S.)
| | - Sun Wook Hwang
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Korea; (H.Z.); (J.Y.L.); (J.Y.S.)
- Department of Physiology, College of Medicine, Korea University, Seoul 02841, Korea
- Correspondence: ; Tel.: +82-2-2286-1204; Fax: +82-2-925-5492
| |
Collapse
|
33
|
Sham surgeries for central and peripheral neural injuries persistently enhance pain-avoidance behavior as revealed by an operant conflict test. Pain 2020; 160:2440-2455. [PMID: 31323014 DOI: 10.1097/j.pain.0000000000001642] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Studies using rodent models of neuropathic pain use sham surgery control procedures that cause deep tissue damage. Sham surgeries would thus be expected to induce potentially long-lasting postsurgical pain, but little evidence for such pain has been reported. Operant tests of voluntary behavior can reveal negative motivational and cognitive aspects of pain that may provide sensitive tools for detecting pain-related alterations. In a previously described operant mechanical conflict test involving lengthy familiarization and training, rodents freely choose to either escape from a brightly lit chamber by crossing sharp probes or refuse to cross. Here, we describe a brief (2-day) mechanical conflict protocol that exploits rats' innate exploratory response to a novel environment to detect persistently enhanced pain-avoidance behavior after sham surgeries for 2 neural injury models: thoracic spinal cord injury and chronic constriction injury of the sciatic nerve. Pitting the combined motivations to avoid the bright light and to explore the novel device against pain from crossing noxious probes disclosed a conflicting, hyperalgesia-related reluctance to repeatedly cross the probes after injury. Rats receiving standard sham surgeries demonstrated enhanced pain-like avoidance behavior compared with naive controls, and this behavior was similar to that of corresponding chronic constriction injury or spinal cord injury rats weeks or months after injury. In the case of sham surgery for spinal cord injury, video analysis of voluntary exploratory behavior directed at the probes revealed enhanced forepaw withdrawal responses. These findings have important implications for preclinical investigations into behavioral alterations and physiological mechanisms associated with postsurgical and neuropathic pain.
Collapse
|
34
|
Contribution of T-Type Calcium Channels to Spinal Cord Injury-Induced Hyperexcitability of Nociceptors. J Neurosci 2020; 40:7229-7240. [PMID: 32839232 DOI: 10.1523/jneurosci.0517-20.2020] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 07/22/2020] [Accepted: 07/30/2020] [Indexed: 01/24/2023] Open
Abstract
A hyperexcitable state and spontaneous activity of nociceptors have been suggested to play a critical role in the development of chronic neuropathic pain following spinal cord injury (SCI). In male rats, we employed the action potential-clamp technique to determine the underlying ionic mechanisms responsible for driving SCI-nociceptors to a hyperexcitable state and for triggering their spontaneous activity. We found that the increased activity of low voltage activated T-type calcium channels induced by the injury sustains the bulk (∼60-70%) of the inward current active at subthreshold voltages during the interspike interval in SCI-nociceptors, with a modest contribution (∼10-15%) from tetrodotoxin (TTX)-sensitive and TTX-resistant sodium channels and hyperpolarization-activated cyclic nucleotide-gated (HCN) channels. In current-clamp recordings, inhibition of T-type calcium channels with 1 μm TTA-P2 reduced both the spontaneous and the evoked firing in response to current injections in SCI-nociceptors to a level similar to sham-nociceptors. Electrophysiology in vitro was then combined with the conditioned place preference (CPP) paradigm to determine the relationship between the increased activity of T-type channels in SCI-nociceptors and chronic neuropathic pain following SCI. The size of the interspike T-type calcium current recorded from nociceptors isolated from SCI rats showing TTA-P2-induced CPP (responders) was ∼6 fold greater than the interspike T-type calcium current recorded from nociceptors isolated from SCI rats without TTA-P2-induced CPP (non-responders). Taken together, our data suggest that the increased activity of T-type calcium channels induced by the injury plays a primary role in driving SCI-nociceptors to a hyperexcitable state and contributes to chronic neuropathic pain following SCI.SIGNIFICANCE STATEMENT Chronic neuropathic pain is a major comorbidity of spinal cord injury (SCI), affecting up to 70-80% of patients. Anticonvulsant and tricyclic antidepressant drugs are first line analgesics used to treat SCI-induced neuropathic pain, but their efficacy is very limited. A hyperexcitable state and spontaneous activity of SCI-nociceptors have been proposed as a possible underlying cause for the development of chronic neuropathic pain following SCI. Here, we show that the increased activity of T-type calcium channels induced by the injury plays a major role in driving SCI-nociceptors to a hyperexcitable state and for promoting their spontaneous activity, suggesting that T-type calcium channels may represent a pharmacological target to treat SCI-induced neuropathic pain.
Collapse
|
35
|
Depolarization-Dependent C-Raf Signaling Promotes Hyperexcitability and Reduces Opioid Sensitivity of Isolated Nociceptors after Spinal Cord Injury. J Neurosci 2020; 40:6522-6535. [PMID: 32690613 DOI: 10.1523/jneurosci.0810-20.2020] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 06/16/2020] [Accepted: 07/13/2020] [Indexed: 12/22/2022] Open
Abstract
Chronic pain caused by spinal cord injury (SCI) is notoriously resistant to treatment, particularly by opioids. After SCI, DRG neurons show hyperactivity and chronic depolarization of resting membrane potential (RMP) that is maintained by cAMP signaling through PKA and EPAC. Importantly, SCI also reduces the negative regulation by Gαi of adenylyl cyclase and its production of cAMP, independent of alterations in G protein-coupled receptors and/or G proteins. Opioid reduction of pain depends on coupling of opioid receptors to Gαi/o family members. Combining high-content imaging and cluster analysis, we show that in male rats SCI decreases opioid responsiveness in vitro within a specific subset of small-diameter nociceptors that bind isolectin B4. This SCI effect is mimicked in nociceptors from naive animals by a modest 5 min depolarization of RMP (15 mm K+; -45 mV), reducing inhibition of cAMP signaling by μ-opioid receptor agonists DAMGO and morphine. Disinhibition and activation of C-Raf by depolarization-dependent phosphorylation are central to these effects. Expression of an activated C-Raf reduces sensitivity of adenylyl cyclase to opioids in nonexcitable HEK293 cells, whereas inhibition of C-Raf or treatment with the hyperpolarizing drug retigabine restores opioid responsiveness and blocks spontaneous activity of nociceptors after SCI. Inhibition of ERK downstream of C-Raf also blocks SCI-induced hyperexcitability and depolarization, without direct effects on opioid responsiveness. Thus, depolarization-dependent C-Raf and downstream ERK activity maintain a depolarized RMP and nociceptor hyperactivity after SCI, providing a self-reinforcing mechanism to persistently promote nociceptor hyperexcitability and limit the therapeutic effectiveness of opioids.SIGNIFICANCE STATEMENT Chronic pain induced by spinal cord injury (SCI) is often permanent and debilitating, and usually refractory to treatment with analgesics, including opioids. SCI-induced pain in a rat model has been shown to depend on persistent hyperactivity in primary nociceptors (injury-detecting sensory neurons), associated with a decrease in the sensitivity of adenylyl cyclase production of cAMP to inhibitory Gαi proteins in DRGs. This study shows that SCI and one consequence of SCI (chronic depolarization of resting membrane potential) decrease sensitivity to opioid-mediated inhibition of cAMP and promote hyperactivity of nociceptors by enhancing C-Raf activity. ERK activation downstream of C-Raf is necessary for maintaining ongoing depolarization and hyperactivity, demonstrating an unexpected positive feedback loop to persistently promote pain.
Collapse
|
36
|
Cevikbas F, Lerner EA. Physiology and Pathophysiology of Itch. Physiol Rev 2020; 100:945-982. [PMID: 31869278 PMCID: PMC7474262 DOI: 10.1152/physrev.00017.2019] [Citation(s) in RCA: 146] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 10/31/2019] [Accepted: 12/04/2019] [Indexed: 02/06/2023] Open
Abstract
Itch is a topic to which everyone can relate. The physiological roles of itch are increasingly understood and appreciated. The pathophysiological consequences of itch impact quality of life as much as pain. These dynamics have led to increasingly deep dives into the mechanisms that underlie and contribute to the sensation of itch. When the prior review on the physiology of itching was published in this journal in 1941, itch was a black box of interest to a small number of neuroscientists and dermatologists. Itch is now appreciated as a complex and colorful Rubik's cube. Acute and chronic itch are being carefully scratched apart and reassembled by puzzle solvers across the biomedical spectrum. New mediators are being identified. Mechanisms blur boundaries of the circuitry that blend neuroscience and immunology. Measures involve psychophysics and behavioral psychology. The efforts associated with these approaches are positively impacting the care of itchy patients. There is now the potential to markedly alleviate chronic itch, a condition that does not end life, but often ruins it. We review the itch field and provide a current understanding of the pathophysiology of itch. Itch is a disease, not only a symptom of disease.
Collapse
Affiliation(s)
- Ferda Cevikbas
- Dermira, Inc., Menlo Park, California; and Harvard Medical School and the Cutaneous Biology Research Center at Massachusetts General Hospital, Charlestown, Massachusetts
| | - Ethan A Lerner
- Dermira, Inc., Menlo Park, California; and Harvard Medical School and the Cutaneous Biology Research Center at Massachusetts General Hospital, Charlestown, Massachusetts
| |
Collapse
|
37
|
Paclitaxel Induces Upregulation of Transient Receptor Potential Vanilloid 1 Expression in the Rat Spinal Cord. Int J Mol Sci 2020; 21:ijms21124341. [PMID: 32570786 PMCID: PMC7352737 DOI: 10.3390/ijms21124341] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Accepted: 06/11/2020] [Indexed: 02/07/2023] Open
Abstract
Painful peripheral neuropathy is a common adverse effect of paclitaxel (PTX) treatment. To analyze the contribution of transient receptor potential vanilloid 1 (TRPV1) in the development of PTX-induced mechanical allodynia/hyperalgesia and thermal hyperalgesia, TRPV1 expression in the rat spinal cord was analyzed after intraperitoneal administration of 2 and 4 mg/kg PTX. PTX treatment increased the expression of TRPV1 protein in the spinal cord. Immunohistochemistry showed that PTX (4 mg/kg) treatment increased TRPV1 protein expression in the superficial layers of the spinal dorsal horn 14 days after treatment. Behavioral assessment using the paw withdrawal response showed that PTX-induced mechanical allodynia/hyperalgesia and thermal hyperalgesia after 14 days was significantly inhibited by oral or intrathecal administration of the TRPV1 antagonist AMG9810. We found that intrathecal administration of small interfering RNA (siRNA) to knock down TRPV1 protein expression in the spinal cord significantly decreased PTX-induced mechanical allodynia/hyperalgesia and thermal hyperalgesia. Together, these results demonstrate that TRPV1 receptor expression in spinal cord contributes, at least in part, to the development of PTX-induced painful peripheral neuropathy. TRPV1 receptor antagonists may be useful in the prevention and treatment of PTX-induced peripheral neuropathic pain.
Collapse
|
38
|
Wu Z, Li L, Xie F, Xu G, Dang D, Yang Q. Enhancing KCNQ Channel Activity Improves Neurobehavioral Recovery after Spinal Cord Injury. J Pharmacol Exp Ther 2020; 373:72-80. [PMID: 31969383 PMCID: PMC7076523 DOI: 10.1124/jpet.119.264010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 01/17/2020] [Indexed: 01/08/2023] Open
Abstract
Spinal cord injury (SCI) usually leads to acute neuronal death and delayed secondary degeneration, resulting in sensory dysfunction, paralysis, and chronic pain. Excessive excitation is one of the critical factors leading to secondary neural damage initiated by various insults. KCNQ/Kv7 channels are highly expressed in spinal neurons and axons and play an important role in controlling their excitability. Enhancing KCNQ channel activity by using its specific opener retigabine could thus be a plausible treatment strategy to reduce the pathology after SCI. We produced contusive SCI at T10 in adult male rats, which then received 10 consecutive days' treatment with retigabine or vehicle starting 3 hours or 3 days after contusion. Two different concentrations and two different delivery methods were applied. Delivery of retigabine via Alzet osmotic pumps, but not intraperitoneal injections 3 hours after contusion, promoted recovery of locomotor function. Remarkably, retigabine delivery in both methods significantly attenuated the development of mechanical stimuli-induced hyperreflexia and spontaneous pain; however, no significant difference in the thermal threshold was observed. Although retigabine delivered 3 days after contusion significantly attenuated the development of mechanical hypersensitivity and spontaneous pain, the locomotor function is not improved by the delayed treatments. Finally, we found that early application of retigabine attenuates the inflammatory activity in the spinal cord and increases the survival of white matter after SCI. Our results suggest that decreasing neuronal excitability by targeting KCNQ/Kv7 channels at acute stage aids the recovery of locomotor function and attenuates the development of neuropathic pain after SCI. SIGNIFICANCE STATEMENT: Several pharmacological interventions have been proposed for spinal cord injury (SCI) treatment, but none have been shown to be both effective and safe in clinical trials. Necrotic neuronal death and chronic pain are often the cost of pathological neural excitation after SCI. We show that early, brief application of retigabine could aid locomotor and sensory neurobehavioral recovery after SCI, supporting the use of this drug in the clinic to promote motor and sensory function in patients with SCI.
Collapse
Affiliation(s)
- Zizhen Wu
- The Solomon H Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland (Z.W.); Department of Integrative Biology and Pharmacology, McGovern Medical School at UT Health, Houston, Texas (L.L., F.X.); Department of Critical Medicine, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China (F.X.); and Department of Neuroscience, Cell Biology and Anatomy at University of Texas Medical Branch, Galveston, Texas (G.X., D.D., Q.Y.)
| | - Lin Li
- The Solomon H Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland (Z.W.); Department of Integrative Biology and Pharmacology, McGovern Medical School at UT Health, Houston, Texas (L.L., F.X.); Department of Critical Medicine, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China (F.X.); and Department of Neuroscience, Cell Biology and Anatomy at University of Texas Medical Branch, Galveston, Texas (G.X., D.D., Q.Y.)
| | - Fuhua Xie
- The Solomon H Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland (Z.W.); Department of Integrative Biology and Pharmacology, McGovern Medical School at UT Health, Houston, Texas (L.L., F.X.); Department of Critical Medicine, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China (F.X.); and Department of Neuroscience, Cell Biology and Anatomy at University of Texas Medical Branch, Galveston, Texas (G.X., D.D., Q.Y.)
| | - Guoying Xu
- The Solomon H Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland (Z.W.); Department of Integrative Biology and Pharmacology, McGovern Medical School at UT Health, Houston, Texas (L.L., F.X.); Department of Critical Medicine, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China (F.X.); and Department of Neuroscience, Cell Biology and Anatomy at University of Texas Medical Branch, Galveston, Texas (G.X., D.D., Q.Y.)
| | - Danny Dang
- The Solomon H Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland (Z.W.); Department of Integrative Biology and Pharmacology, McGovern Medical School at UT Health, Houston, Texas (L.L., F.X.); Department of Critical Medicine, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China (F.X.); and Department of Neuroscience, Cell Biology and Anatomy at University of Texas Medical Branch, Galveston, Texas (G.X., D.D., Q.Y.)
| | - Qing Yang
- The Solomon H Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland (Z.W.); Department of Integrative Biology and Pharmacology, McGovern Medical School at UT Health, Houston, Texas (L.L., F.X.); Department of Critical Medicine, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China (F.X.); and Department of Neuroscience, Cell Biology and Anatomy at University of Texas Medical Branch, Galveston, Texas (G.X., D.D., Q.Y.)
| |
Collapse
|
39
|
EPAC1 and EPAC2 promote nociceptor hyperactivity associated with chronic pain after spinal cord injury. NEUROBIOLOGY OF PAIN 2019; 7:100040. [PMID: 31890991 PMCID: PMC6926371 DOI: 10.1016/j.ynpai.2019.100040] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 11/15/2019] [Accepted: 11/20/2019] [Indexed: 12/19/2022]
Abstract
Chronic pain following spinal cord injury (SCI) is associated with electrical hyperactivity (spontaneous and evoked) in primary nociceptors. Cyclic adenosine monophosphate (cAMP) signaling is an important contributor to nociceptor excitability, and knockdown of the cAMP effector, exchange protein activated by cAMP (EPAC), has been shown to relieve pain-like responses in several chronic pain models. To examine potentially distinct roles of each EPAC isoform (EPAC1 and 2) in maintaining chronic pain, we used rat and mouse models of contusive spinal cord injury (SCI). Pharmacological inhibition of EPAC1 or 2 in a rat SCI model was sufficient to reverse SCI-induced nociceptor hyperactivity, indicating that EPAC1 and 2 signaling activity are complementary, with both required to maintain hyperactivity. However, EPAC activation was not sufficient to induce similar hyperactivity in nociceptors from naïve rats, and we observed no change in EPAC protein expression after SCI. In the mouse SCI model, inhibition of both EPAC isoforms through a combination of pharmacological inhibition and genetic deletion was required to reverse SCI-induced nociceptor hyperactivity. This was consistent with our finding that neither EPAC1-/- nor EPAC2-/- mice were protected against SCI-induced chronic pain as assessed with an operant mechanical conflict test. Thus, EPAC1 and 2 activity may play a redundant role in mouse nociceptors, although no corresponding change in EPAC protein expression levels was detected after SCI. Despite some differences between these species, our data demonstrate a fundamental role for both EPAC1 and EPAC2 in mechanisms maintaining nociceptor hyperactivity and chronic pain after SCI.
Collapse
|
40
|
Yasko JR, Moss IL, Mains RE. Transcriptional Profiling of Non-injured Nociceptors After Spinal Cord Injury Reveals Diverse Molecular Changes. Front Mol Neurosci 2019; 12:284. [PMID: 32038157 PMCID: PMC6988781 DOI: 10.3389/fnmol.2019.00284] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 11/08/2019] [Indexed: 01/01/2023] Open
Abstract
Traumatic spinal cord injury (SCI) has devastating implications for patients, including a high predisposition for developing chronic pain distal to the site of injury. Chronic pain develops weeks to months after injury, consequently, patients are treated after irreparable changes have occurred. Nociceptors are central to chronic pain; however, the diversity of this cellular population presents challenges to understanding mechanisms and attributing pain modalities to specific cell types. To begin to address how peripheral sensory neurons below the injury level may contribute to the below-level pain reported by SCI patients, we examined SCI-induced changes in gene expression in lumbar dorsal root ganglia (DRG) below the site of injury. SCI was performed at the T10 vertebral level, with injury produced by a vessel clip with a closing pressure of 15 g for 1 min. Alterations in gene expression produce long-term sensory changes, therefore, we were interested in studying SCI-induced transcripts before the onset of chronic pain, which may trigger changes in downstream signaling pathways and ultimately facilitate the transmission of pain. To examine changes in the nociceptor subpopulation in DRG distal to the site of injury, we retrograde labeled sensory neurons projecting to the hairy hindpaw skin with fluorescent dye and collected the corresponding lumbar (L2–L6) DRG 4 days post-injury. Following dissociation, labeled neurons were purified by fluorescence-activated cell sorting (FACS). RNA was extracted from sorted sensory neurons of naïve, sham, or SCI mice and sequenced. Transcript abundances validated that the desired population of nociceptors were isolated. Cross-comparisons to data sets from similar studies confirmed, we were able to isolate our cells of interest and identify a unique pattern of gene expression within a subpopulation of neurons projecting to the hairy hindpaw skin. Differential gene expression analysis showed high expression levels and significant transcript changes 4 days post-injury in SCI cell populations relevant to the onset of chronic pain. Regulatory interrelationships predicted by pathway analysis implicated changes within the synaptogenesis signaling pathway as well as networks related to inflammatory signaling mechanisms, suggesting a role for synaptic plasticity and a correlation with pro-inflammatory signaling in the transition from acute to chronic pain.
Collapse
Affiliation(s)
- Jessica R Yasko
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT, United States
| | - Isaac L Moss
- Department of Orthopedic Surgery and the Comprehensive Spine Center, University of Connecticut Health Center, Farmington, CT, United States
| | - Richard E Mains
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT, United States
| |
Collapse
|
41
|
Walters ET. Adaptive mechanisms driving maladaptive pain: how chronic ongoing activity in primary nociceptors can enhance evolutionary fitness after severe injury. Philos Trans R Soc Lond B Biol Sci 2019; 374:20190277. [PMID: 31544606 DOI: 10.1098/rstb.2019.0277] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Chronic pain is considered maladaptive by clinicians because it provides no apparent protective or recuperative benefits. Similarly, evolutionary speculations have assumed that chronic pain represents maladaptive or evolutionarily neutral dysregulation of acute pain mechanisms. By contrast, the present hypothesis proposes that chronic pain can be driven by mechanisms that evolved to reduce increased vulnerability to attack from predators and aggressive conspecifics, which often target prey showing physical impairment after severe injury. Ongoing pain and anxiety persisting long after severe injury continue to enhance vigilance and behavioural caution, decreasing the heightened vulnerability to attack that results from motor impairment and disfigurement, thereby increasing survival and reproduction (fitness). This hypothesis is supported by evidence of animals surviving and reproducing after traumatic amputations, and by complex specializations that enable primary nociceptors to detect local and systemic signs of injury and inflammation, and to maintain low-frequency discharge that can promote ongoing pain indefinitely. Ongoing activity in nociceptors involves intricate electrophysiological and anatomical specializations, including inducible alterations in the expression of ion channels and receptors that produce persistent hyperexcitability and hypersensitivity to chemical signals of injury. Clinically maladaptive chronic pain may sometimes result from the recruitment of this powerful evolutionary adaptation to severe bodily injury. This article is part of the Theo Murphy meeting issue 'Evolution of mechanisms and behaviour important for pain'.
Collapse
Affiliation(s)
- Edgar T Walters
- Department of Integrative Biology and Pharmacology, McGovern Medical School at UTHealth, 6431 Fannin Street, Houston, TX 77030, USA
| |
Collapse
|
42
|
MZF1 in the Dorsal Root Ganglia Contributes to the Development and Maintenance of Neuropathic Pain via Regulation of TRPV1. Neural Plast 2019; 2019:2782417. [PMID: 31582966 PMCID: PMC6754943 DOI: 10.1155/2019/2782417] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 04/27/2019] [Accepted: 05/19/2019] [Indexed: 01/09/2023] Open
Abstract
Previous studies have demonstrated that myeloid zinc finger 1 (MZF1) in the dorsal root ganglion (DRG) participates in neuropathic pain induced by chronic-constriction injury (CCI) via regulation of voltage-gated K+ channels (Kv). Emerging evidence indicates that transient receptor potential vanilloid 1 (TRPV1) is involved in the development and maintenance of neuropathic pain. Although it is known that the transcription of TRPV1 is regulated by Kruppel-like zinc-finger transcription factor 7 (Klf7)—and that the structure of TRPV1 is similar to that of Kv—few studies have systematically investigated the relationship between MZF1 and TRPV1 in neuropathic pain. In the present study, we demonstrated that CCI induced an increase in MZF1 and TRPV1 in lumbar-level 4/5 (L4/5) DRGs at 3 days post-CCI and that this increase was persistent until at least 14 days post-CCI. DRG microinjection of rAAV5-MZF1 into the DRGs of naïve rats resulted in a decrease in paw-withdrawal threshold (PWT) and paw-withdrawal latency (PWL) compared with that of the rAAV5-EGFP group, which started at four weeks and lasted until at least eight weeks after microinjection. Additionally, prior microinjection of MZF1 siRNA clearly ameliorated CCI-induced reduction in PWT and PWL at 3 days post-CCI and lasted until at least 7 days post-CCI. Correspondingly, microinjection of MZF1 siRNA subsequent to CCI alleviated the established mechanical allodynia and thermal hyperalgesia induced by CCI, which occurred at 3 days postinjection and lasted until at least 10 days postinjection. Microinjection of rAAV5-MZF1 increased the expression of TRPV1 in DRGs. Microinjection of MZF1 siRNA diminished the CCI-induced increase of TRPV1, but not P2X7R, in DRGs. These findings suggest that MZF1 may contribute to neuropathic pain via regulation of TRPV1 expression in DRGs.
Collapse
|
43
|
Hu Q, Wang Q, Wang C, Tai Y, Liu B, Shao X, Fang J, Liu B. TRPV1 Channel Contributes to the Behavioral Hypersensitivity in a Rat Model of Complex Regional Pain Syndrome Type 1. Front Pharmacol 2019; 10:453. [PMID: 31105572 PMCID: PMC6498414 DOI: 10.3389/fphar.2019.00453] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Accepted: 04/09/2019] [Indexed: 12/31/2022] Open
Abstract
Complex regional pain syndrome type 1 (CRPS-I) is a debilitating pain condition that significantly affects life quality of patients. It remains a clinically challenging condition and the mechanisms of CRPS-I have not been fully elucidated. Here, we investigated the involvement of TRPV1, a non-selective cation channel important for integrating various painful stimuli, in an animal model of CRPS-I. A rat model of chronic post-ischemia pain (CPIP) was established to mimic CRPS-I. TRPV1 expression was significantly increased in hind paw tissue and small to medium-sized dorsal root ganglion (DRG) neurons of CPIP rats. CPIP rats showed increased TRPV1 current density and capsaicin responding rate in small-sized nociceptive DRG neurons. Local pharmacological blockage of TRPV1 with the specific antagonist AMG9810, at a dosage that does not produce hyperthermia or affect thermal perception or locomotor activity, effectively attenuated thermal and mechanical hypersensitivity in bilateral hind paws of CPIP rats and reduced the hyperexcitability of DRG neurons induced by CPIP. CPIP rats showed bilateral spinal astrocyte and microglia activations, which were significantly attenuated by AMG9810 treatment. These findings identified an important role of TRPV1 in mediating thermal and mechanical hypersensitivity in a CRPS-I animal model and further suggest local pharmacological blocking TRPV1 may represent an effective approach to ameliorate CRPS-I.
Collapse
Affiliation(s)
- Qimiao Hu
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou, China
| | - Qiong Wang
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| | - Chuan Wang
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| | - Yan Tai
- Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Boyu Liu
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou, China
| | - Xiaomei Shao
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou, China
| | - Jianqiao Fang
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou, China
| | - Boyi Liu
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou, China
| |
Collapse
|
44
|
Inflammation induces Epac-protein kinase C alpha and epsilon signaling in TRPV1-mediated hyperalgesia. Pain 2019; 159:2383-2393. [PMID: 30015706 DOI: 10.1097/j.pain.0000000000001346] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The exchange proteins activated by cAMP (Epacs) have been shown to play important roles in producing inflammation-induced nociception. Transient receptor potential vanilloid type 1 (TRPV1) is a major receptor processing thermal and chemosensitive nociceptive information. The role of Epacs in modulating the activity of TRPV1 has yet to be determined. Studying the effect of complete Freund adjuvant (CFA)-induced inflammation on capsaicin-activated TRPV1 nociceptive responses in dorsal root ganglia (DRG), we found that CFA produced a large increase in capsaicin-induced responses. The increase was inhibited by Epac1 and Epac2 antagonists. Thus, activation of Epacs is critical in producing enhancement in TRPV1-mediated responses under inflammatory conditions. In addition, the inflammation-induced enhancement of TRPV1 responses was blocked by PKCα and PKCε inhibitors, suggesting the essential roles of these PKCs in enhancing TRPV1 responses. To determine the mechanism underlying the Epac actions on TRPV1, we studied the effects of the Epac activator, 8-(4-chlorophenylthio)-2-O-methyl-cAMP (CPT), on capsaicin-induced nociceptive behavioral responses, capsaicin-activated currents, expression and membrane trafficking of PKC and TRPV1 in DRG. CPT was found to enhance capsaicin-induced nociception and ionic currents. The enhancement was inhibited by PKCα and PKCε inhibitors. In addition, CPT increased the expression of phosphorylated PKCα (pPKCα) and membrane TRPV1 expression in DRG. Studying the colocalization of TRPV1 and pPKCα or pPKCε in DRG slices prepared from CFA-treated rats, we found that pPKCα or pPKCε expressed with TRPV1 in different-sized neurons to exert differential influences on TRPV1 activity. Thus, Epac-PKC signaling is critically important in producing inflammation-induced potentiation of TRPV1 functions.
Collapse
|
45
|
Odem MA, Bavencoffe AG, Cassidy RM, Lopez ER, Tian J, Dessauer CW, Walters ET. Isolated nociceptors reveal multiple specializations for generating irregular ongoing activity associated with ongoing pain. Pain 2018; 159:2347-2362. [PMID: 30015712 PMCID: PMC6193853 DOI: 10.1097/j.pain.0000000000001341] [Citation(s) in RCA: 126] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Ongoing pain has been linked to ongoing activity (OA) in human C-fiber nociceptors, but rodent models of pain-related OA have concentrated on allodynia rather than ongoing pain, and on OA generated in non-nociceptive Aβ fibers rather than C-fiber nociceptors. Little is known about how ongoing pain or nociceptor OA is generated. To define neurophysiological alterations underlying nociceptor OA, we have used isolated dorsal root ganglion neurons that continue to generate OA after removal from animals displaying ongoing pain. We subclassify OA as either spontaneous activity generated solely by alterations intrinsic to the active neuron or as extrinsically driven OA. Both types of OA were implicated previously in nociceptors in vivo and after isolation following spinal cord injury, which produces chronic ongoing pain. Using novel automated algorithms to analyze irregular changes in membrane potential, we have found, in a distinctive, nonaccommodating type of probable nociceptor, induction by spinal cord injury of 3 alterations that promote OA: (1) prolonged depolarization of resting membrane potential, (2) a hyperpolarizing shift in the voltage threshold for action potential generation, and (3) an increase in the incidence of large depolarizing spontaneous fluctuations (DSFs). Can DSFs also be enhanced acutely to promote OA in neurons from uninjured animals? A low dose of serotonin failed to change resting membrane potential but lowered action potential threshold. When combined with artificial depolarization to model inflammation, serotonin also strongly potentiated DSFs and OA. These findings reveal nociceptor specializations for generating OA that may promote ongoing pain in chronic and acute conditions.
Collapse
Affiliation(s)
- Max A. Odem
- Department of Integrative Biology and Pharmacology, McGovern Medical School at UTHealth, Houston, TX, USA
| | - Alexis G. Bavencoffe
- Department of Integrative Biology and Pharmacology, McGovern Medical School at UTHealth, Houston, TX, USA
| | - Ryan M. Cassidy
- Department of Integrative Biology and Pharmacology, McGovern Medical School at UTHealth, Houston, TX, USA
| | - Elia R. Lopez
- Department of Integrative Biology and Pharmacology, McGovern Medical School at UTHealth, Houston, TX, USA
| | - Jinbin Tian
- Department of Integrative Biology and Pharmacology, McGovern Medical School at UTHealth, Houston, TX, USA
| | - Carmen W. Dessauer
- Department of Integrative Biology and Pharmacology, McGovern Medical School at UTHealth, Houston, TX, USA
| | - Edgar T. Walters
- Department of Integrative Biology and Pharmacology, McGovern Medical School at UTHealth, Houston, TX, USA
| |
Collapse
|
46
|
Chhaya SJ, Quiros-Molina D, Tamashiro-Orrego AD, Houlé JD, Detloff MR. Exercise-Induced Changes to the Macrophage Response in the Dorsal Root Ganglia Prevent Neuropathic Pain after Spinal Cord Injury. J Neurotrauma 2018; 36:877-890. [PMID: 30152715 DOI: 10.1089/neu.2018.5819] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Spinal cord injury (SCI) induces neuropathic pain that is refractory to treatment. Central and peripheral immune responses to SCI play critical roles in pain development. Although immune responses in the dorsal horn have been implicated in SCI-pain, immune mechanisms in the periphery, especially in the dorsal root ganglia (DRG), where nociceptor cell bodies reside, have not been well studied. Exercise is an immunomodulator, and we showed previously that early exercise after SCI reduces pain development. However, the mechanisms of exercise-mediated pain reduction are not understood. Therefore, we examined the 1) underlying immune differences in the spinal cord and DRG between rats with and without pain and 2) immunomodulatory effects of exercise in pain reduction. Rats were subjected to a unilateral contusion at C5 and tested for pain development using von Frey and mechanical conflict-avoidance paradigms. A subgroup of rats was exercised on forced running wheels starting at 5 days post-injury for 4 weeks. We observed greater microglial activation in the C7-C8 dorsal horn of rats with SCI-induced pain compared to rats with normal sensation, and early exercise reduced this activation independently of pain behavior. Further, abnormal pain sensation strongly correlated with an increased number of DRG macrophages. Importantly, exercise-treated rats that maintain normal sensation also have a lower number of macrophages in the DRG. Our data suggest that macrophage presence in the DRG may be an important effector of pain development, and early wheel walking exercise may mediate pain prevention by modulating the injury-induced macrophage response in the DRG. Further supportive evidence demonstrated that rats that developed pain despite exercise intervention still displayed a significantly elevated number of macrophages in the DRG. Collectively, these data suggest that macrophage presence in the DRG may be an amenable cellular target for future therapies.
Collapse
Affiliation(s)
- Soha J Chhaya
- Department of Neurobiology and Anatomy, Spinal Cord Research Center, College of Medicine Drexel University Philadelphia, Pennsylvania
| | - Daniel Quiros-Molina
- Department of Neurobiology and Anatomy, Spinal Cord Research Center, College of Medicine Drexel University Philadelphia, Pennsylvania
| | - Alessandra D Tamashiro-Orrego
- Department of Neurobiology and Anatomy, Spinal Cord Research Center, College of Medicine Drexel University Philadelphia, Pennsylvania
| | - John D Houlé
- Department of Neurobiology and Anatomy, Spinal Cord Research Center, College of Medicine Drexel University Philadelphia, Pennsylvania
| | - Megan Ryan Detloff
- Department of Neurobiology and Anatomy, Spinal Cord Research Center, College of Medicine Drexel University Philadelphia, Pennsylvania
| |
Collapse
|
47
|
Wang Y, Ye F, Huang C, Xue F, Li Y, Gao S, Qiu Z, Li S, Chen Q, Zhou H, Song Y, Huang W, Tan W, Wang Z. Bioinformatic Analysis of Potential Biomarkers for Spinal Cord-injured Patients with Intractable Neuropathic Pain. Clin J Pain 2018; 34:825-830. [PMID: 29547407 PMCID: PMC6078488 DOI: 10.1097/ajp.0000000000000608] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Revised: 10/05/2017] [Accepted: 10/22/2017] [Indexed: 12/30/2022]
Abstract
BACKGROUND Neuropathic pain is one of the common complications after spinal cord injury (SCI), affecting individuals' quality of life. The molecular mechanism for neuropathic pain after SCI is still unclear. We aimed to discover potential genes and microRNAs (miRNAs) related to neuropathic pain by the bioinformatics method. METHODS Microarray data of GSE69901 were obtained from Gene Expression Omnibus (GEO) database. Peripheral blood samples from individuals with or without neuropathic pain after SCI were collected. Twelve samples from individuals with neuropathic pain and 13 samples from individuals without pain as controls were included in the downloaded microarray. Differentially expressed genes (DEGs) between the neuropathic pain group and the control group were detected using the GEO2R online tool. Functional enrichment analysis of DEGs was performed using the DAVID database. Protein-protein interaction network was constructed from the STRING database. MiRNAs targeting these DEGs were obtained from the miRNet database. A merged miRNA-DEG network was constructed and analyzed with Cytoscape software. RESULTS In total, 1134 DEGs were identified between individuals with or without neuropathic pain (case and control), and 454 biological processes were enriched. We identified 4 targeted miRNAs, including mir-204-5p, mir-519d-3p, mir-20b-5p, mir-6838-5p, which may be potential biomarkers for SCI patients. CONCLUSION Protein modification and regulation of the biological process of the central nervous system may be a risk factor in SCI. Certain genes and miRNAs may be potential biomarkers for the prediction of and potential targets for the prevention and treatment of neuropathic pain after SCI.
Collapse
Affiliation(s)
- Yimin Wang
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-sen University
- Department of Anesthesiology, Guangdong Second Provincial General Hospital, Guangzhou
| | - Fang Ye
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-sen University
| | - Chanyan Huang
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-sen University
| | - Faling Xue
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-sen University
| | - Yingyuan Li
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-sen University
| | - Shaowei Gao
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-sen University
| | - Zeting Qiu
- Zhongshan School of Medicine, Sun Yat-sen University, Guangdong, China
| | - Si Li
- Zhongshan School of Medicine, Sun Yat-sen University, Guangdong, China
| | - Qinchang Chen
- Zhongshan School of Medicine, Sun Yat-sen University, Guangdong, China
| | - Huaqiang Zhou
- Zhongshan School of Medicine, Sun Yat-sen University, Guangdong, China
| | - Yiyan Song
- Zhongshan School of Medicine, Sun Yat-sen University, Guangdong, China
| | - Wenqi Huang
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-sen University
| | - Wulin Tan
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-sen University
| | - Zhongxing Wang
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-sen University
| |
Collapse
|
48
|
Shiao R, Lee-Kubli CA. Neuropathic Pain After Spinal Cord Injury: Challenges and Research Perspectives. Neurotherapeutics 2018; 15:635-653. [PMID: 29736857 PMCID: PMC6095789 DOI: 10.1007/s13311-018-0633-4] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Neuropathic pain is a debilitating consequence of spinal cord injury (SCI) that remains difficult to treat because underlying mechanisms are not yet fully understood. In part, this is due to limitations of evaluating neuropathic pain in animal models in general, and SCI rodents in particular. Though pain in patients is primarily spontaneous, with relatively few patients experiencing evoked pains, animal models of SCI pain have primarily relied upon evoked withdrawals. Greater use of operant tasks for evaluation of the affective dimension of pain in rodents is needed, but these tests have their own limitations such that additional studies of the relationship between evoked withdrawals and operant outcomes are recommended. In preclinical SCI models, enhanced reflex withdrawal or pain responses can arise from pathological changes that occur at any point along the sensory neuraxis. Use of quantitative sensory testing for identification of optimal treatment approach may yield improved identification of treatment options and clinical trial design. Additionally, a better understanding of the differences between mechanisms contributing to at- versus below-level neuropathic pain and neuropathic pain versus spasticity may shed insights into novel treatment options. Finally, the role of patient characteristics such as age and sex in pathogenesis of neuropathic SCI pain remains to be addressed.
Collapse
Affiliation(s)
- Rani Shiao
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 N. Torrey Pines, La Jolla, California, 92073, USA
| | - Corinne A Lee-Kubli
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 N. Torrey Pines, La Jolla, California, 92073, USA.
| |
Collapse
|
49
|
Sliwinski C, Nees TA, Puttagunta R, Weidner N, Blesch A. Sensorimotor Activity Partially Ameliorates Pain and Reduces Nociceptive Fiber Density in the Chronically Injured Spinal Cord. J Neurotrauma 2018; 35:2222-2238. [PMID: 29706124 DOI: 10.1089/neu.2017.5431] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
A large proportion of patients suffering from spinal cord injury (SCI) develop chronic central neuropathic pain. Previously, we and others have shown that sensorimotor training early after SCI can prevent the development of mechanical allodynia. To determine whether training initiated in the subchronic/chronic phase remains effective, correlates of below-level neuropathic pain were analyzed in the hindpaws 5-10 weeks after a moderate T11 contusion SCI (50 kDyn) in adult female C57BL/6 mice. In a comparison of SCI and sham mice 5 weeks post-injury, about 80% of injured animals developed mechanical hypersensitivity to light mechanical stimuli, whereas testing of noxious stimuli revealed hypo-responsiveness. Thermal sensitivity testing showed a decreased response latency after injury. Without intervention, mechanical and thermal hyper-responsiveness were evident until the end of the experiment (10 weeks). In contrast, treadmill training (2 × 15 min/day; 5 × /week) initiated 6 weeks post-injury resulted in partial amelioration of pain behavior and this effect remained stable. Analysis of calcitonin gene-related peptide (CGRP)-labeled fibers in lamina III-IV of the lumbar dorsal horn revealed an increase in labeling density after SCI. This was not due to changes in the number or size distribution of CGRP-labeled lumbar dorsal root ganglion neurons. Treadmill training reduced the CGRP-labeling density in the spinal cord of injured mice, whereas the density of non-peptidergic isolectin-B4 (IB4)+ fibers showed no changes in lamina IIi and a slight reduction of sparse IB4 labeling in laminae III-IV. Thus, sensorimotor activity initiated in the subchronic/chronic phase of SCI remains effective in ameliorating pain behavior and influencing structural changes of the nociceptive system.
Collapse
Affiliation(s)
| | - Timo A Nees
- 1 Spinal Cord Injury Center, Heidelberg University Hospital , Heidelberg, Germany .,2 Center for Orthopedic and Trauma Surgery, Heidelberg University Hospital , Heidelberg, Germany
| | - Radhika Puttagunta
- 1 Spinal Cord Injury Center, Heidelberg University Hospital , Heidelberg, Germany
| | - Norbert Weidner
- 1 Spinal Cord Injury Center, Heidelberg University Hospital , Heidelberg, Germany
| | - Armin Blesch
- 1 Spinal Cord Injury Center, Heidelberg University Hospital , Heidelberg, Germany .,3 Department of Neurological Surgery and Goodman Campbell Brain and Spine, Stark Neurosciences Research Institute, Indiana University School of Medicine , Indianapolis, Indiana
| |
Collapse
|
50
|
Wang S, Brigoli B, Lim J, Karley A, Chung MK. Roles of TRPV1 and TRPA1 in Spontaneous Pain from Inflamed Masseter Muscle. Neuroscience 2018; 384:290-299. [PMID: 29890293 DOI: 10.1016/j.neuroscience.2018.05.048] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 05/28/2018] [Accepted: 05/31/2018] [Indexed: 01/26/2023]
Abstract
Craniofacial muscle pain, such as spontaneous pain and bite-evoked pain, are major symptoms in patients with temporomandibular disorders and infection. However, the underlying mechanisms of muscle pain, especially mechanisms of highly prevalent spontaneous pain, are poorly understood. Recently, we reported that transient receptor potential vanilloid 1 (TRPV1) contributes to spontaneous pain but only marginally contributes to bite-evoked pain during masseter inflammation. Here, we investigated the role of transient receptor potential ankyrin 1 (TRPA1) in spontaneous and bite-evoked pain during masseter inflammation, and dissected the relative contributions of TRPA1 and TRPV1. Masseter inflammation increased mouse grimace scale (MGS) scores and face wiping behaviors. Pharmacological or genetic inhibition of TRPA1 significantly attenuated MGS but not face wiping behaviors. MGS scores were also attenuated by scavenging putative endogenous ligands for TRPV1 or TRPA1. Simultaneous inhibition of TRPA1 by AP18 and TRPV1 by AMG9810 in masseter muscle resulted in robust inhibition of both MGS and face wiping behaviors. Administration of AP18 or AMG9810 to masseter muscle induced conditioned place preference (CPP). The extent of CPP following simultaneous administration of AP18 and AMG9810 was greater than that induced by the individual antagonists. In contrast, inflammation-induced reduction of bite force was not affected by the inhibition of TRPA1 alone or in combination with TRPV1. These results suggest that simultaneous inhibition of TRPV1 and TRPA1 produces additive relief of spontaneous pain, but does not ameliorate bite-evoked pain during masseter inflammation. Our results provide further evidence that distinct mechanisms underlie spontaneous and bite-evoked pain from inflamed masseter muscle.
Collapse
Affiliation(s)
- Sheng Wang
- Department of Neural and Pain Sciences, School of Dentistry, Program in Neuroscience, Center to Advance Chronic Pain Research, University of Maryland, Baltimore, 650 W. Baltimore Street, Baltimore, MD 21201, USA
| | - Benjamin Brigoli
- Department of Neural and Pain Sciences, School of Dentistry, Program in Neuroscience, Center to Advance Chronic Pain Research, University of Maryland, Baltimore, 650 W. Baltimore Street, Baltimore, MD 21201, USA
| | - Jongseuk Lim
- Department of Neural and Pain Sciences, School of Dentistry, Program in Neuroscience, Center to Advance Chronic Pain Research, University of Maryland, Baltimore, 650 W. Baltimore Street, Baltimore, MD 21201, USA
| | - Alisha Karley
- Department of Neural and Pain Sciences, School of Dentistry, Program in Neuroscience, Center to Advance Chronic Pain Research, University of Maryland, Baltimore, 650 W. Baltimore Street, Baltimore, MD 21201, USA
| | - Man-Kyo Chung
- Department of Neural and Pain Sciences, School of Dentistry, Program in Neuroscience, Center to Advance Chronic Pain Research, University of Maryland, Baltimore, 650 W. Baltimore Street, Baltimore, MD 21201, USA.
| |
Collapse
|